1
|
Wu J, Gu Z, Modica JA, Chen S, Mrksich M, Voth GA. Megamolecule Self-Assembly Networks: A Combined Computational and Experimental Design Strategy. J Am Chem Soc 2024; 146:30553-30564. [PMID: 39451142 DOI: 10.1021/jacs.4c11892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
This work describes the use of computational strategies to design megamolecule building blocks for the self-assembly of lattice networks. The megamolecules are prepared by attaching four Cutinase-SnapTag fusion proteins (CS fusions) to a four-armed linker, followed by functionalizing each fusion with a terpyridine linker. This functionality is designed to participate in a metal-mediated self-assembly process to give networks. This article describes a simulation-guided strategy for the design of megamolecules to optimize the peptide linker in the fusion protein to give conformations that are best suited for self-assembly and therefore streamlines the typically time-consuming and labor-intensive experimental process. We designed 11 candidate megamolecules and identified the most promising linker, (EAAAK)2, along with the optimal experimental conditions through a combination of all-atom molecular dynamics, enhanced sampling, and larger-scale coarse-grained molecular dynamics simulations. Our simulation findings were validated and found to be consistent with the experimental results. Significantly, this study offers valuable insight into the self-assembly of megamolecule networks and provides a novel and general strategy for large biomolecular material designs by using systematic bottom-up coarse-grained simulations.
Collapse
Affiliation(s)
- Jiangbo Wu
- Department of Chemistry, Chicago Center for Theoretical Chemistry, The James Franck Institute, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois 60637, United States
| | - Zhaoyi Gu
- Departments of Chemistry and Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Justin A Modica
- Departments of Chemistry and Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Sijia Chen
- Department of Chemistry, Chicago Center for Theoretical Chemistry, The James Franck Institute, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois 60637, United States
| | - Milan Mrksich
- Departments of Chemistry and Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Gregory A Voth
- Department of Chemistry, Chicago Center for Theoretical Chemistry, The James Franck Institute, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois 60637, United States
| |
Collapse
|
2
|
Abou-Shanab AM, Gaser OA, Soliman MW, Oraby A, Salah RA, Gabr M, Edris AAF, Mohamed I, El-Badri N. Human amniotic membrane scaffold enhances adipose mesenchymal stromal cell mitochondrial bioenergetics promoting their regenerative capacities. Mol Cell Biochem 2024:10.1007/s11010-024-05094-x. [PMID: 39453499 DOI: 10.1007/s11010-024-05094-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 08/12/2024] [Indexed: 10/26/2024]
Abstract
The human amniotic membrane (hAM) has been applied as a scaffold in tissue engineering to sustain stem cells and enhance their regenerative capacities. We investigated the molecular and biochemical regulations of mesenchymal stromal cells (MSCs) cultured on hAM scaffold in a three-dimensional (3D) setting. Culture of adipose-MSCs (AMSCs) on decellularized hAM showed significant improvement in their viability, proliferative capacity, resistance to apoptosis, and enhanced MSC markers expression. These cultured MSCs displayed altered expression of markers associated with pro-angiogenesis and inflammation and demonstrated increased potential for differentiation into adipogenic and osteogenic lineages. The hAM scaffold modulated cellular respiration by upregulating glycolysis in MSCs as evidenced by increased glucose consumption, cellular pyruvate and lactate production, and upregulation of glycolysis markers. These metabolic changes modulated mitochondrial oxidative phosphorylation (OXPHOS) and altered the production of reactive oxygen species (ROS), expression of OXPHOS markers, and total antioxidant capacity. They also significantly boosted the urea cycle and altered the mitochondrial ultrastructure. Similar findings were observed in bone marrow-derived MSCs (BMSCs). Live cell imaging of BMSCs cultured in the same 3D environment revealed dynamic changes in cellular activity and interactions with its niche. These findings provide evidence for the favorable properties of hAM as a biomimetic scaffold for enhancing the in vitro functionality of MSCs and supporting their potential usefulness in clinical applications.
Collapse
Affiliation(s)
- Ahmed M Abou-Shanab
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Ola A Gaser
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Mariam Waleed Soliman
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Alaa Oraby
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Radwa Ayman Salah
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Mahmoud Gabr
- Urology and Nephrology Center, Mansoura University, Mansoura, 35516, Egypt
| | | | - Ihab Mohamed
- Department of Zoology, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt.
| |
Collapse
|
3
|
Yang J, Wang P, Zhang Y, Zhang M, Sun Q, Chen H, Dong L, Chu Z, Xue B, Hoff WD, Zhao C, Wang W, Wei Q, Cao Y. Photo-tunable hydrogels reveal cellular sensing of rapid rigidity changes through the accumulation of mechanical signaling molecules. Cell Stem Cell 2024:S1934-5909(24)00359-X. [PMID: 39437791 DOI: 10.1016/j.stem.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 07/08/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
Cells use traction forces to sense mechanical cues in their environment. While the molecular clutch model effectively explains how cells exert more forces on stiffer substrates, it falls short in addressing their adaptation to dynamic mechanical fluctuations prevalent in tissues and organs. Here, using hydrogel with photo-responsive rigidity, we show that cells' response to rigidity changes is frequency dependent. Strikingly, at certain frequencies, cellular traction forces exceed those on static substrates 4-fold stiffer, challenging the established molecular clutch model. We discover that the discrepancy between the rapid adaptation of traction forces and the slower deactivation of mechanotransduction signaling proteins results in their accumulation, thereby enhancing long-term cellular traction in dynamic settings. Consequently, we propose a new model that melds immediate mechanosensing with extended mechanical signaling. Our study underscores the significance of dynamic rigidity in the development of synthetic biomaterials, emphasizing the importance of considering both immediate and prolonged cellular responses.
Collapse
Affiliation(s)
- Jiapeng Yang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250021, China; College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Peng Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Yu Zhang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250021, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Man Zhang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Qian Sun
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Huiyan Chen
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Liang Dong
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Zhiqin Chu
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong 999077, China; Joint Appointment with School of Biomedical Sciences, The University of Hong Kong, Hong Kong 999077, China
| | - Bin Xue
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Wouter David Hoff
- Department of Physics, Oklahoma State University, Stillwater, OK 74078, USA
| | - Changsheng Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Wei Wang
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Qiang Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China.
| | - Yi Cao
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250021, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Department of Physics, Nanjing University, Nanjing 210093, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China.
| |
Collapse
|
4
|
Liu S, Manshaii F, Chen J, Wang X, Wang S, Yin J, Yang M, Chen X, Yin X, Zhou Y. Unleashing the Potential of Electroactive Hybrid Biomaterials and Self-Powered Systems for Bone Therapeutics. NANO-MICRO LETTERS 2024; 17:44. [PMID: 39417933 PMCID: PMC11486894 DOI: 10.1007/s40820-024-01536-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/08/2024] [Indexed: 10/19/2024]
Abstract
The incidence of large bone defects caused by traumatic injury is increasing worldwide, and the tissue regeneration process requires a long recovery time due to limited self-healing capability. Endogenous bioelectrical phenomena have been well recognized as critical biophysical factors in bone remodeling and regeneration. Inspired by bioelectricity, electrical stimulation has been widely considered an external intervention to induce the osteogenic lineage of cells and enhance the synthesis of the extracellular matrix, thereby accelerating bone regeneration. With ongoing advances in biomaterials and energy-harvesting techniques, electroactive biomaterials and self-powered systems have been considered biomimetic approaches to ensure functional recovery by recapitulating the natural electrophysiological microenvironment of healthy bone tissue. In this review, we first introduce the role of bioelectricity and the endogenous electric field in bone tissue and summarize different techniques to electrically stimulate cells and tissue. Next, we highlight the latest progress in exploring electroactive hybrid biomaterials as well as self-powered systems such as triboelectric and piezoelectric-based nanogenerators and photovoltaic cell-based devices and their implementation in bone tissue engineering. Finally, we emphasize the significance of simulating the target tissue's electrophysiological microenvironment and propose the opportunities and challenges faced by electroactive hybrid biomaterials and self-powered bioelectronics for bone repair strategies.
Collapse
Affiliation(s)
- Shichang Liu
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710018, People's Republic of China
| | - Farid Manshaii
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, 90095, USA
| | - Jinmiao Chen
- Hangzhou Institute of Technology, Xidian University, Hangzhou, 311231, People's Republic of China
| | - Xinfei Wang
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, 90095, USA
| | - Shaolei Wang
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, 90095, USA
| | - Junyi Yin
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, 90095, USA
| | - Ming Yang
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710018, People's Republic of China.
| | - Xuxu Chen
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710018, People's Republic of China.
| | - Xinhua Yin
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710018, People's Republic of China.
| | - Yunlei Zhou
- Hangzhou Institute of Technology, Xidian University, Hangzhou, 311231, People's Republic of China
| |
Collapse
|
5
|
Li S, Li J, Xu J, Shen Y, Shang X, Li H, Wang J, Liu Y, Qiang L, Qiao Z, Wang J, He Y, Hu Y. Removal-Free and Multicellular Suspension Bath-Based 3D Bioprinting. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2406891. [PMID: 39394784 DOI: 10.1002/adma.202406891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/27/2024] [Indexed: 10/14/2024]
Abstract
Suspension bath-based 3D bioprinting (SUB3BP) is effective in creating engineered vascular structures. The transfer of oxygen and nutrients via engineered vascular networks is necessary for tissue or organ survival and integration following transplantation. Existing SUB3BP techniques face challenges in fabricating hierarchical structures with multicellular organization, including issues related to suspension bath removal, restricted material choices, and low accuracy. A next-generation SUB3BP technique that is removal-free and multicellular is presented. A simple, storable, stable, and scalable starch hydrogel design leverages the diverse spectrum of hydrogels available for use in SUB3BP. Starch granules (8.1 µm) create vascular structures with minimal surface roughness (2.5 µm) that simulate more natural vessel walls compared to prior research. The development of cells and organoids, as well as the bioprinting of multicellular skin models with vasculature, demonstrates that starch suspension baths eliminate the removal process and have the potential for fabricating artificial tissue with a hierarchical structure and multicellular distribution.
Collapse
Affiliation(s)
- Shuai Li
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jianping Li
- Zhejiang Key Laboratory of Precision Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jian Xu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yifan Shen
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xiushuai Shang
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Hangyu Li
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jingwen Wang
- Zhejiang Key Laboratory of Precision Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yihao Liu
- Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Lei Qiang
- Key Laboratory of Advanced Technologies of Materials (MOE), School of Materials Science and Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Zhiguang Qiao
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Jinwu Wang
- Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yong He
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yihe Hu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| |
Collapse
|
6
|
Liu Q, Zhang Y, Yu S, Zhao C, Yang Y, Yan J, Wang Y, Liu D, Liu Y, Zhang X. Proanthocyanidins modification of the mineralized collagen scaffold based on synchronous self-assembly/mineralization for bone regeneration. Colloids Surf B Biointerfaces 2024; 245:114290. [PMID: 39383582 DOI: 10.1016/j.colsurfb.2024.114290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/25/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Proteoglycans (PG) is crucial for regulating collagen formation and mineralization during bone tissue development. A wide variety of PG-modified collagen scaffolds have been proposed for bone engineering application to promote biological responses and work as artificial matrices that guide tissue regeneration. However, poor performance of theses biomaterials against infections has led to an unmet need for clinical prevention. Therefore, we utilized proanthocyanidins (PA) to simulate the functions of PG, including mediating the collagen assembly and intrafibrillar mineralization, to optimize scaffolds performance. The excellent antibacterial properties of PA can endow the scaffolds with anti-infection effects in the process of tissue regeneration. When PA was added during fibrillogenesis, the collagen fibrils appeared irregular aggregation and the mineralization degree was reduced. In contrast, the addition of PA after collagen self-assembly improved the latter's ability to act as a deposition template and remarkably promoted mineral ions infiltration, thus enhancing intrafibrillar mineralization. The PA-modified scaffold displayed a highly hydrophilicity behaviour and long-term resistance to degradation. The sustained release of PA effectively inhibited the activity of Staphylococcus aureus. The scaffold also showed excellent biocompatibility and improved bone regeneration in calvarial critical-size defect models. The application of PA enables a dual-function scaffold with favourable intrafibrillar mineralization and anti-bacterial properties for bone regeneration.
Collapse
Affiliation(s)
- Qing Liu
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No. 12 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Ye Zhang
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No. 12 Qixiangtai Road, Heping District, Tianjin 300070, China; Department of Biomaterials, 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
| | - Shuxian Yu
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No. 12 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Chuanze Zhao
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No. 12 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Yuqing Yang
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No. 12 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Jianyu Yan
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No. 12 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Yuge Wang
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No. 12 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Dayong Liu
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No. 12 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Ying Liu
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No. 12 Qixiangtai Road, Heping District, Tianjin 300070, China.
| | - Xu Zhang
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No. 12 Qixiangtai Road, Heping District, Tianjin 300070, China; Tianjin Medical University Institute of Stomatology, No. 12 Qixiangtai Road, Heping District, Tianjin 300070, China.
| |
Collapse
|
7
|
Zhong L, Banigo AT, Zoetebier B, Karperien M. Bioactive Hydrogels Based on Tyramine and Maleimide Functionalized Dextran for Tissue Engineering Applications. Gels 2024; 10:566. [PMID: 39330167 PMCID: PMC11431488 DOI: 10.3390/gels10090566] [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: 07/12/2024] [Revised: 08/20/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024] Open
Abstract
Hydrogels are widely used in tissue engineering due to their ability to form three-dimensional (3D) structures that support cellular functions and mimic the extracellular matrix (ECM). Despite their advantages, dextran-based hydrogels lack intrinsic biological activity, limiting their use in this field. Here, we present a strategy for developing bioactive hydrogels through sequential thiol-maleimide bio-functionalization and enzyme-catalyzed crosslinking. The hydrogel network is formed through the reaction of tyramine moieties in the presence of horseradish peroxidase (HRP) and hydrogen peroxide (H2O2), allowing for tunable gelation time and stiffness by adjusting H2O2 concentrations. Maleimide groups on the hydrogel backbone enable the coupling of thiol-containing bioactive molecules, such as arginylglycylaspartic acid (RGD) peptides, to enhance biological activity. We examined the effects of hydrogel stiffness and RGD concentration on human mesenchymal stem cells (hMSCs) during differentiation and found that hMSCs encapsulated within these hydrogels exhibited over 88% cell viability on day 1 across all conditions, with a slight reduction to 60-81% by day 14. Furthermore, the hydrogels facilitated adipogenic differentiation, as evidenced by positive Oil Red O staining. These findings demonstrate that DexTA-Mal hydrogels create a biocompatible environment that is conducive to cell viability and differentiation, offering a versatile platform for future tissue engineering applications.
Collapse
Affiliation(s)
| | | | | | - Marcel Karperien
- Department of Developmental BioEngineering, Faculty of Science and Technology, TechMed Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands; (L.Z.); (A.T.B.); (B.Z.)
| |
Collapse
|
8
|
Dong L, Li L, Chen H, Cao Y, Lei H. Mechanochemistry: Fundamental Principles and Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2403949. [PMID: 39206931 DOI: 10.1002/advs.202403949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/30/2024] [Indexed: 09/04/2024]
Abstract
Mechanochemistry is an emerging research field at the interface of physics, mechanics, materials science, and chemistry. Complementary to traditional activation methods in chemistry, such as heat, electricity, and light, mechanochemistry focuses on the activation of chemical reactions by directly or indirectly applying mechanical forces. It has evolved as a powerful tool for controlling chemical reactions in solid state systems, sensing and responding to stresses in polymer materials, regulating interfacial adhesions, and stimulating biological processes. By combining theoretical approaches, simulations and experimental techniques, researchers have gained intricate insights into the mechanisms underlying mechanochemistry. In this review, the physical chemistry principles underpinning mechanochemistry are elucidated and a comprehensive overview of recent significant achievements in the discovery of mechanically responsive chemical processes is provided, with a particular emphasis on their applications in materials science. Additionally, The perspectives and insights into potential future directions for this exciting research field are offered.
Collapse
Affiliation(s)
- Liang Dong
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, Jiangsu, 210093, P. R. China
| | - Luofei Li
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, Jiangsu, 210093, P. R. China
| | - Huiyan Chen
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, Jiangsu, 210093, P. R. China
| | - Yi Cao
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, Jiangsu, 210093, P. R. China
| | - Hai Lei
- School of Physics, Zhejiang University, Hangzhou, Zhejiang, 310027, P. R. China
- Institute of Advanced Physics, Zhejiang University, Hangzhou, Zhejiang, 310027, P. R. China
| |
Collapse
|
9
|
Du X, Xing Y, Li Y, Cao M, Wu J, Dong G, Shi Z, Wei X, Qiu M, Gao J, Xu Y, Xu H, Liu D, Dong Y. Gradually Self-Strengthen DNA Supramolecular Hydrogels. Macromol Rapid Commun 2024; 45:e2400177. [PMID: 38636558 DOI: 10.1002/marc.202400177] [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: 03/25/2024] [Indexed: 04/20/2024]
Abstract
The dynamic mechanical strength of the extracellular matrix (ECM) has been demonstrated to play important role in determining the cell behavior. Growing evidences suggest that the gradual stiffening process of the matrix is particularly decisive during tissue development and wound healing. Herein, a novel strategy to prepare hydrogels with gradually enhanced mechanical strength is provided. Such hydrogels could maintain the dynamic properties at their initial states, such as self-healing and shear-thinning properties. With subsequent slow covalent crosslinking, the stability and mechanical properties would be gradually improved. This method is useful for sequence programmability and oxidation strategies, which has provided an alternated tool to study cell behavior during dynamic increase in mechanical strength of ECM.
Collapse
Affiliation(s)
- Xiuji Du
- CAS Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yongzheng Xing
- National Engineering Research Center for Colloidal Materials, School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
| | - Yujie Li
- Key Laboratory of Organic Optoelectronics & Molecular Engineering of the Ministry of Education, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Muqing Cao
- Key Laboratory of Organic Optoelectronics & Molecular Engineering of the Ministry of Education, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Jun Wu
- CAS Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guizhi Dong
- CAS Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ziwei Shi
- CAS Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xunan Wei
- Department of Chemistry, Renmin University of China, Beijing, 100872, China
| | - Miaomiao Qiu
- CAS Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Junjie Gao
- National Engineering Research Center for Colloidal Materials, School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
| | - Yun Xu
- Center for Medical Device Evaluation, China Food and Drug Administration (CFDA), Beijing, 100084, China
| | - Huaping Xu
- Key Laboratory of Organic Optoelectronics & Molecular Engineering of the Ministry of Education, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Dongsheng Liu
- Key Laboratory of Organic Optoelectronics & Molecular Engineering of the Ministry of Education, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Yuanchen Dong
- CAS Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
10
|
Shi J, Liu Y, Ling Y, Tang H. Polysaccharide-protein based scaffolds for cartilage repair and regeneration. Int J Biol Macromol 2024; 274:133495. [PMID: 38944089 DOI: 10.1016/j.ijbiomac.2024.133495] [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: 05/09/2024] [Revised: 06/23/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024]
Abstract
Cartilage repair and regeneration have become a global issue that millions of patients from all over the world need surgical intervention to repair the articular cartilage annually due to the limited self-healing capability of the cartilage tissues. Cartilage tissue engineering has gained significant attention in cartilage repair and regeneration by integration of the chondrocytes (or stem cells) and the artificial scaffolds. Recently, polysaccharide-protein based scaffolds have demonstrated unique and promising mechanical and biological properties as the artificial extracellular matrix of natural cartilage. In this review, we summarize the modification methods for polysaccharides and proteins. The preparation strategies for the polysaccharide-protein based hydrogel scaffolds are presented. We discuss the mechanical, physical and biological properties of the polysaccharide-protein based scaffolds. Potential clinical translation and challenges on the artificial scaffolds are also discussed.
Collapse
Affiliation(s)
- Jin Shi
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou 215123, China
| | - Yu Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou 215123, China
| | - Ying Ling
- Institute of Neuroscience, Soochow University, Suzhou 215123, China.
| | - Haoyu Tang
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou 215123, China.
| |
Collapse
|
11
|
Murali A, Brokesh AM, Cross LM, Kersey AL, Jaiswal MK, Singh I, Gaharwar A. Inorganic Biomaterials Shape the Transcriptome Profile to Induce Endochondral Differentiation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402468. [PMID: 38738803 PMCID: PMC11304299 DOI: 10.1002/advs.202402468] [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: 03/07/2024] [Revised: 03/27/2024] [Indexed: 05/14/2024]
Abstract
Minerals play a vital role, working synergistically with enzymes and other cofactors to regulate physiological functions including tissue healing and regeneration. The bioactive characteristics of mineral-based nanomaterials can be harnessed to facilitate in situ tissue regeneration by attracting endogenous progenitor and stem cells and subsequently directing tissue-specific differentiation. Here, cellular responses of human mesenchymal stem/stromal cells to traditional bioactive mineral-based nanomaterials, such as hydroxyapatite, whitlockite, silicon-dioxide, and the emerging synthetic 2D nanosilicates are investigated. Transcriptome sequencing is utilized to probe the cellular response and determine the significantly affected signaling pathways due to exposure to these inorganic nanomaterials. Transcriptome profiles of stem cells treated with nanosilicates reveals a stabilized skeletal progenitor state suggestive of endochondral differentiation. This observation is bolstered by enhanced deposition of matrix mineralization in nanosilicate treated stem cells compared to control or other treatments. Specifically, use of 2D nanosilicates directs osteogenic differentiation of stem cells via activation of bone morphogenetic proteins and hypoxia-inducible factor 1-alpha signaling pathway. This study provides insight into impact of nanomaterials on cellular gene expression profile and predicts downstream effects of nanomaterial induction of endochondral differentiation.
Collapse
Affiliation(s)
- Aparna Murali
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Anna M. Brokesh
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Lauren M. Cross
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Anna L. Kersey
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Manish K. Jaiswal
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Irtisha Singh
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
- Department of Cell Biology and GeneticsCollege of MedicineTexas A&M UniversityBryanTX77807‐3260USA
- Interdisciplinary Program in Genetics and GenomicsTexas A&M UniversityCollege StationTX77843USA
| | - Akhilesh Gaharwar
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
- Interdisciplinary Program in Genetics and GenomicsTexas A&M UniversityCollege StationTX77843USA
- Department of Material Science and EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| |
Collapse
|
12
|
Gui Y, Deng X, Li N, Zhao L. PRELP reduce cell stiffness and adhesion to promote the growth and metastasis of colorectal cancer cells by binding to integrin α5. Exp Cell Res 2024; 441:114151. [PMID: 38992455 DOI: 10.1016/j.yexcr.2024.114151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 06/19/2024] [Accepted: 07/03/2024] [Indexed: 07/13/2024]
Abstract
PRELP is thought to be an inhibitor of the development and progression of a variety of malignancies. Metastasis is a major cause of death in patients with colorectal cancer, but the mechanism underlying the role of PRELP in colorectal cancer metastasis remains poorly understood. In this study, we found that PRELP was significantly higher in metastatic tissues than in non-metastatic tissues of colorectal cancer and was closely associated with poor prognosis of colorectal cancer patients. PRELP promotes growth and metastasis of colorectal cancer cells. PRELP reduces cell stiffness and adhesion. PRELP promoted EMT in colorectal cancer cells and that PRELP bind to integrin α5 to activate the integrin α5/FAK/AKT signaling pathway. In conclusion, we demonstrate that PRELP is upregulated in metastatic colorectal cancer, providing a potential prognostic marker and therapeutic target for the clinical management of metastatic colorectal cancer from a biomechanical perspective.
Collapse
Affiliation(s)
- Yajun Gui
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 41001l, China; Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China
| | - Xiangying Deng
- Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Namei Li
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 41001l, China; Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China
| | - Lin Zhao
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 41001l, China; Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China.
| |
Collapse
|
13
|
Major GS, Doan VK, Longoni A, Bilek MMM, Wise SG, Rnjak-Kovacina J, Yeo GC, Lim KS. Mapping the microcarrier design pathway to modernise clinical mesenchymal stromal cell expansion. Trends Biotechnol 2024; 42:859-876. [PMID: 38320911 DOI: 10.1016/j.tibtech.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/08/2024] [Accepted: 01/08/2024] [Indexed: 02/08/2024]
Abstract
Microcarrier expansion systems show exciting potential to revolutionise mesenchymal stromal cell (MSC)-based clinical therapies by providing an opportunity for economical large-scale expansion of donor- and patient-derived cells. The poor reproducibility and efficiency of cell expansion on commercial polystyrene microcarriers have driven the development of novel microcarriers with tuneable physical, mechanical, and cell-instructive properties. These new microcarriers show innovation toward improving cell expansion outcomes, although their limited biological characterisation and compatibility with dynamic culture systems suggest the need to realign the microcarrier design pathway. Clear headway has been made toward developing infrastructure necessary for scaling up these technologies; however, key challenges remain in characterising the wholistic effects of microcarrier properties on the biological fate and function of expanded MSCs.
Collapse
Affiliation(s)
- Gretel S Major
- School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Vinh K Doan
- School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Alessia Longoni
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marcela M M Bilek
- School of Biomedical Engineering, University of Sydney, Sydney, Australia; School of Physics, University of Sydney, Sydney, Australia; Charles Perkins Centre, University of Sydney, Sydney, Australia; Sydney Nano Institute, University of Sydney, Sydney, Australia
| | - Steven G Wise
- School of Medical Sciences, University of Sydney, Sydney, Australia; Charles Perkins Centre, University of Sydney, Sydney, Australia
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia; Tyree Institute of Health Engineering, University of New South Wales, Sydney, Australia
| | - Giselle C Yeo
- Charles Perkins Centre, University of Sydney, Sydney, Australia; School of Life and Environmental Sciences, University of Sydney, Sydney, Australia.
| | - Khoon S Lim
- School of Medical Sciences, University of Sydney, Sydney, Australia; Charles Perkins Centre, University of Sydney, Sydney, Australia; Sydney Nano Institute, University of Sydney, Sydney, Australia.
| |
Collapse
|
14
|
Zhu J, Zhu X, Xu Y, Chen X, Ge X, Huang Y, Wang Z. The role of noncoding RNAs in beta cell biology and tissue engineering. Life Sci 2024; 348:122717. [PMID: 38744419 DOI: 10.1016/j.lfs.2024.122717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/29/2024] [Accepted: 05/11/2024] [Indexed: 05/16/2024]
Abstract
The loss or dysfunction of pancreatic β-cells, which are responsible for insulin secretion, constitutes the foundation of all forms of diabetes, a widely prevalent disease worldwide. The replacement of damaged β-cells with regenerated or transplanted cells derived from stem cells is a promising therapeutic strategy. However, inducing the differentiation of stem cells into fully functional glucose-responsive β-cells in vitro has proven to be challenging. Noncoding RNAs (ncRNAs) have emerged as critical regulatory factors governing the differentiation, identity, and function of β-cells. Furthermore, engineered hydrogel systems, biomaterials, and organ-like structures possess engineering characteristics that can provide a three-dimensional (3D) microenvironment that supports stem cell differentiation. This review summarizes the roles and contributions of ncRNAs in maintaining the differentiation, identity, and function of β-cells. And it focuses on regulating the levels of ncRNAs in stem cells to activate β-cell genetic programs for generating alternative β-cells and discusses how to manipulate ncRNA expression by combining hydrogel systems and other tissue engineering materials. Elucidating the patterns of ncRNA-mediated regulation in β-cell biology and utilizing this knowledge to control stem cell differentiation may offer promising therapeutic strategies for generating functional insulin-producing cells in diabetes cell replacement therapy and tissue engineering.
Collapse
Affiliation(s)
- Jiaqi Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Xiaoren Zhu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Yang Xu
- Center of Gallbladder Disease, Shanghai East Hospital, Institute of Gallstone Disease, School of Medicine, Tongji University, Shanghai 200092, China
| | - Xingyou Chen
- Medical School of Nantong University, Nantong 226001, China
| | - Xinqi Ge
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Yan Huang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| | - Zhiwei Wang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| |
Collapse
|
15
|
Gerrits L, Bakker B, Hendriks LD, Engels S, Hammink R, Kouwer PHJ. Tailoring of Physical Properties in Macroporous Poly(isocyanopeptide) Cryogels. Biomacromolecules 2024; 25:3464-3474. [PMID: 38743442 PMCID: PMC11170948 DOI: 10.1021/acs.biomac.4c00086] [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: 01/21/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/16/2024]
Abstract
Over the years, synthetic hydrogels have proven remarkably useful as cell culture matrixes to elucidate the role of the extracellular matrix (ECM) on cell behavior. Yet, their lack of interconnected macropores undermines the widespread use of hydrogels in biomedical applications. To overcome this limitation, cryogels, a class of macroporous hydrogels, are rapidly emerging. Here, we introduce a new, highly elastic, and tunable synthetic cryogel, based on poly(isocyanopeptides) (PIC). Introduction of methacrylate groups on PIC facilitated cryopolymerization through free-radical polymerization and afforded cryogels with an interconnected macroporous structure. We investigated which cryogelation parameters can be used to tune the architectural and mechanical properties of the PIC cryogels by systematically altering cryopolymerization temperature, polymer concentration, and polymer molecular weight. We show that for decreasing cryopolymerization temperatures, there is a correlation between cryogel pore size and stiffness. More importantly, we demonstrate that by simply varying the polymer concentration, we can selectively tune the compressive strength of PIC cryogels without affecting their architecture. This unique feature is highly useful for biomedical applications, as it facilitates decoupling of stiffness from other variables such as pore size. As such, PIC cryogels provide an interesting new biomaterial for scientists to unravel the role of the ECM in cellular functions.
Collapse
Affiliation(s)
- Lotte Gerrits
- Institute
for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen ,Netherlands
| | - Bram Bakker
- Institute
for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen ,Netherlands
| | - Lynn D. Hendriks
- Institute
for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen ,Netherlands
| | - Sjoerd Engels
- Institute
for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen ,Netherlands
| | - Roel Hammink
- Department
of Medical BioSciences,Radboudumc, Geert Grooteplein 26, 6525 GA Nijmegen, The Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen ,Netherlands
| | - Paul H. J. Kouwer
- Institute
for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen ,Netherlands
| |
Collapse
|
16
|
Zhu F, Yan N, Lu X, Xu J, Gu H, Liang J, Cheng K, Wang X, Ma X, Ma N, Zhao X, Chen C, Nie G. Cell-Reprogramming-Inspired Dynamically Responsive Hydrogel Boosts the Induction of Pluripotency via Phase-Separated Biomolecular Condensates. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2211609. [PMID: 36989141 DOI: 10.1002/adma.202211609] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/23/2023] [Indexed: 05/16/2023]
Abstract
Induced pluripotent stem cells (iPSCs) have wide applications in disease modeling, personalized medicine, and tissue engineering. The generation of iPSCs from somatic cells via transcriptional-factor- or chemical molecule-based approaches are time-consuming and inefficient. Here, a cell-reprogramming-inspired dynamically responsive hydrogel is fabricated via a synthetic-biology-based strategy. Human and mouse somatic cells (including senescent cells) are efficiently reprogrammed into iPSCs that exhibit key features of embryonic stem cells. The cell-reprogramming-responsive hydrogel possesses dynamic bioresponsiveness, and it faithfully senses metabolic remodeling and extracellular acidification during cell reprogramming, responding by changing its mechanical properties accordingly. Mechanistic study demonstrates that the autonomous change of the mechanical properties of the cell-reprogramming-responsive hydrogel elicits the formation of Yes-associated protein (YAP) biomolecular condensates with the appropriate timing during cell reprogramming, ensuring a faster and more efficient generation of iPSCs than conventional cell reprogramming approach. Taken together, this study reveals the robust induction of pluripotency by coordination of cell-reprogramming-inspired dynamically responsive hydrogel and phase-separated biomolecular condensates.
Collapse
Affiliation(s)
- Fei Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
| | - Na Yan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xukun Lu
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
- Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Junchao Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haiyan Gu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Keman Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
| | - Xiaona Wang
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiaotu Ma
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
| | - Nana Ma
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- The GBA National Institute for Nanotechnology Innovation, Guangdong, 510700, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- The GBA National Institute for Nanotechnology Innovation, Guangdong, 510700, China
| |
Collapse
|
17
|
Shahrokhtash A, Sutherland DS. Smart Biointerfaces via Click Chemistry-Enabled Nanopatterning of Multiple Bioligands and DNA Force Sensors. ACS APPLIED MATERIALS & INTERFACES 2024; 16:21534-21545. [PMID: 38634566 PMCID: PMC11073048 DOI: 10.1021/acsami.4c00831] [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: 01/15/2024] [Revised: 03/10/2024] [Accepted: 04/08/2024] [Indexed: 04/19/2024]
Abstract
Nanoscale biomolecular placement is crucial for advancing cellular signaling, sensor technology, and molecular interaction studies. Despite this, current methods fall short in enabling large-area nanopatterning of multiple biomolecules while minimizing nonspecific interactions. Using bioorthogonal tags at a submicron scale, we introduce a novel hole-mask colloidal lithography method for arranging up to three distinct proteins, DNA, or peptides on large, fully passivated surfaces. The surfaces are compatible with single-molecule fluorescence microscopy and microplate formats, facilitating versatile applications in cellular and single-molecule assays. We utilize fully passivated and transparent substrates devoid of metals and nanotopographical features to ensure accurate patterning and minimize nonspecific interactions. Surface patterning is achieved using bioorthogonal TCO-tetrazine (inverse electron-demand Diels-Alder, IEDDA) ligation, DBCO-azide (strain-promoted azide-alkyne cycloaddition, SPAAC) click chemistry, and biotin-avidin interactions. These are arranged on surfaces passivated with dense poly(ethylene glycol) PEG brushes crafted through the selective and stepwise removal of sacrificial metallic and polymeric layers, enabling the directed attachment of biospecific tags with nanometric precision. In a proof-of-concept experiment, DNA tension gauge tether (TGT) force sensors, conjugated to cRGD (arginylglycylaspartic acid) in nanoclusters, measured fibroblast integrin tension. This novel application enables the quantification of forces in the piconewton range, which is restricted within the nanopatterned clusters. A second demonstration of the platform to study integrin and epidermal growth factor (EGF) proximal signaling reveals clear mechanotransduction and changes in the cellular morphology. The findings illustrate the platform's potential as a powerful tool for probing complex biochemical pathways involving several molecules arranged with nanometer precision and cellular interactions at the nanoscale.
Collapse
Affiliation(s)
- Ali Shahrokhtash
- Interdisciplinary
Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, 8000Aarhus C, Denmark
- The
Centre for Cellular Signal Patterns (CellPAT), Gustav Wieds Vej 14, 8000 Aarhus C ,Denmark
| | - Duncan S. Sutherland
- Interdisciplinary
Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, 8000Aarhus C, Denmark
- The
Centre for Cellular Signal Patterns (CellPAT), Gustav Wieds Vej 14, 8000 Aarhus C ,Denmark
| |
Collapse
|
18
|
Ryoo H, Kimmel H, Rondo E, Underhill GH. Advances in high throughput cell culture technologies for therapeutic screening and biological discovery applications. Bioeng Transl Med 2024; 9:e10627. [PMID: 38818120 PMCID: PMC11135158 DOI: 10.1002/btm2.10627] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 06/01/2024] Open
Abstract
Cellular phenotypes and functional responses are modulated by the signals present in their microenvironment, including extracellular matrix (ECM) proteins, tissue mechanical properties, soluble signals and nutrients, and cell-cell interactions. To better recapitulate and analyze these complex signals within the framework of more physiologically relevant culture models, high throughput culture platforms can be transformative. High throughput methodologies enable scientists to extract increasingly robust and broad datasets from individual experiments, screen large numbers of conditions for potential hits, better qualify and predict responses for preclinical applications, and reduce reliance on animal studies. High throughput cell culture systems require uniformity, assay miniaturization, specific target identification, and process simplification. In this review, we detail the various techniques that researchers have used to face these challenges and explore cellular responses in a high throughput manner. We highlight several common approaches including two-dimensional multiwell microplates, microarrays, and microfluidic cell culture systems as well as unencapsulated and encapsulated three-dimensional high throughput cell culture systems, featuring multiwell microplates, micromolds, microwells, microarrays, granular hydrogels, and cell-encapsulated microgels. We also discuss current applications of these high throughput technologies, namely stem cell sourcing, drug discovery and predictive toxicology, and personalized medicine, along with emerging opportunities and future impact areas.
Collapse
Affiliation(s)
- Hyeon Ryoo
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Hannah Kimmel
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Evi Rondo
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Gregory H. Underhill
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| |
Collapse
|
19
|
Sun R, Jin X, Bao Y, Cao Z, Gao D, Zhang R, Qiu L, Yuan H, Xing C. Microenvironment with NIR-Controlled ROS and Mechanical Tensions for Manipulating Cell Activities in Wound Healing. NANO LETTERS 2024; 24:3257-3266. [PMID: 38426843 DOI: 10.1021/acs.nanolett.4c00307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The extracellular matrix (ECM) orchestrates cell behavior and tissue regeneration by modulating biochemical and mechanical signals. Manipulating cell-material interactions is crucial for leveraging biomaterials to regulate cell functions. Yet, integrating multiple cues in a single material remains a challenge. Here, near-infrared (NIR)-controlled multifunctional hydrogel platforms, named PIC/CM@NPs, are introduced to dictate fibroblast behavior during wound healing by tuning the matrix oxidative stress and mechanical tensions. PIC/CM@NPs are prepared through cell adhesion-medicated assembly of collagen-like polyisocyanide (PIC) polymers and cell-membrane-coated conjugated polymer nanoparticles (CM@NPs), which closely mimic the fibrous structure and nonlinear mechanics of ECM. Upon NIR stimulation, PIC/CM@NPs composites enhance fibroblast cell proliferation, migration, cytokine production, and myofibroblast activation, crucial for wound closure. Moreover, they exhibit effective and toxin removal antibacterial properties, reducing inflammation. This multifunctional approach accelerates healing by 95%, highlighting the importance of integrating biochemical and biophysical cues in the biomaterial design for advanced tissue regeneration.
Collapse
Affiliation(s)
- Rang Sun
- Key Laboratory of Hebei Province for Molecular Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, P. R. China
| | - Xinyu Jin
- Key Laboratory of Hebei Province for Molecular Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, P. R. China
| | - Yuying Bao
- Key Laboratory of Hebei Province for Molecular Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, P. R. China
| | - Zhanshuo Cao
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin 300130, P. R. China
| | - Dong Gao
- Key Laboratory of Hebei Province for Molecular Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, P. R. China
| | - Ran Zhang
- Key Laboratory of Hebei Province for Molecular Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, P. R. China
| | - Liang Qiu
- Key Laboratory of Hebei Province for Molecular Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, P. R. China
| | - Hongbo Yuan
- Key Laboratory of Hebei Province for Molecular Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, P. R. China
- Molecular Imaging and Photonics, Chemistry Department, KU Leuven, Leuven 3000, Belgium
| | - Chengfen Xing
- Key Laboratory of Hebei Province for Molecular Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, P. R. China
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin 300130, P. R. China
| |
Collapse
|
20
|
Kosmidis Papadimitriou A, Chong SW, Shen Y, Lee OS, Knowles TPJ, Grover LM, Vigolo D. Fabrication of gradient hydrogels using a thermophoretic approach in microfluidics. Biofabrication 2024; 16:025023. [PMID: 38377611 DOI: 10.1088/1758-5090/ad2b05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 02/20/2024] [Indexed: 02/22/2024]
Abstract
The extracellular matrix presents spatially varying physical cues that can influence cell behavior in many processes. Physical gradients within hydrogels that mimic the heterogenous mechanical microenvironment are useful to study the impact of these cues on cellular responses. Therefore, simple and reliable techniques to create such gradient hydrogels are highly desirable. This work demonstrates the fabrication of stiffness gradient Gellan gum (GG) hydrogels by applying a temperature gradient across a microchannel containing hydrogel precursor solution. Thermophoretic migration of components within the precursor solution generates a concentration gradient that mirrors the temperature gradient profile, which translates into mechanical gradients upon crosslinking. Using this technique, GG hydrogels with stiffness gradients ranging from 20 to 90 kPa over 600µm are created, covering the elastic moduli typical of moderately hard to hard tissues. MC3T3 osteoblast cells are then cultured on these gradient substrates, which exhibit preferential migration and enhanced osteogenic potential toward the stiffest region on the gradient. Overall, the thermophoretic approach provides a non-toxic and effective method to create hydrogels with defined mechanical gradients at the micron scale suitable forin vitrobiological studies and potentially tissue engineering applications.
Collapse
Affiliation(s)
| | - Shin Wei Chong
- The University of Sydney, School of Biomedical Engineering, Sydney, NSW 2006, Australia
- The University of Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Yi Shen
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
- The University of Sydney, School of Chemical and Biomolecular Engineering, Sydney, NSW 2006, Australia
- The University of Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Oisin Stefan Lee
- The University of Sydney, School of Biomedical Engineering, Sydney, NSW 2006, Australia
| | - Tuomas P J Knowles
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Liam M Grover
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Daniele Vigolo
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
- The University of Sydney, School of Biomedical Engineering, Sydney, NSW 2006, Australia
- The University of Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
21
|
Mout R, Bretherton RC, Decarreau J, Lee S, Gregorio N, Edman NI, Ahlrichs M, Hsia Y, Sahtoe DD, Ueda G, Sharma A, Schulman R, DeForest CA, Baker D. De novo design of modular protein hydrogels with programmable intra- and extracellular viscoelasticity. Proc Natl Acad Sci U S A 2024; 121:e2309457121. [PMID: 38289949 PMCID: PMC10861882 DOI: 10.1073/pnas.2309457121] [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: 06/06/2023] [Accepted: 12/26/2023] [Indexed: 02/01/2024] Open
Abstract
Relating the macroscopic properties of protein-based materials to their underlying component microstructure is an outstanding challenge. Here, we exploit computational design to specify the size, flexibility, and valency of de novo protein building blocks, as well as the interaction dynamics between them, to investigate how molecular parameters govern the macroscopic viscoelasticity of the resultant protein hydrogels. We construct gel systems from pairs of symmetric protein homo-oligomers, each comprising 2, 5, 24, or 120 individual protein components, that are crosslinked either physically or covalently into idealized step-growth biopolymer networks. Through rheological assessment, we find that the covalent linkage of multifunctional precursors yields hydrogels whose viscoelasticity depends on the crosslink length between the constituent building blocks. In contrast, reversibly crosslinking the homo-oligomeric components with a computationally designed heterodimer results in viscoelastic biomaterials exhibiting fluid-like properties under rest and low shear, but solid-like behavior at higher frequencies. Exploiting the unique genetic encodability of these materials, we demonstrate the assembly of protein networks within living mammalian cells and show via fluorescence recovery after photobleaching (FRAP) that mechanical properties can be tuned intracellularly in a manner similar to formulations formed extracellularly. We anticipate that the ability to modularly construct and systematically program the viscoelastic properties of designer protein-based materials could have broad utility in biomedicine, with applications in tissue engineering, therapeutic delivery, and synthetic biology.
Collapse
Affiliation(s)
- Rubul Mout
- Department of Biochemistry, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
- Stem Cell Program at Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
| | - Ross C. Bretherton
- Department of Bioengineering, University of Washington, Seattle, WA98195
- Department of Chemical Engineering, University of Washington, Seattle, WA98195
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA98195
- Department of Chemistry, University of Washington, Seattle, WA98195
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA98195
| | - Justin Decarreau
- Department of Biochemistry, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Sangmin Lee
- Department of Biochemistry, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Nicole Gregorio
- Department of Bioengineering, University of Washington, Seattle, WA98195
- Department of Chemical Engineering, University of Washington, Seattle, WA98195
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA98195
- Department of Chemistry, University of Washington, Seattle, WA98195
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA98195
| | - Natasha I. Edman
- Department of Biochemistry, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA98195
- Medical Scientist Training Program, University of Washington, Seattle, WA98195
| | - Maggie Ahlrichs
- Department of Biochemistry, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Yang Hsia
- Department of Biochemistry, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Danny D. Sahtoe
- Department of Biochemistry, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
- HHMI, University of Washington, Seattle, WA98195
| | - George Ueda
- Department of Biochemistry, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Alee Sharma
- College of Professional Studies, Northeastern University, Boston, MA02115
| | - Rebecca Schulman
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD21218
- Department of Computer Science, Johns Hopkins University, Baltimore, MD21218
| | - Cole A. DeForest
- Institute for Protein Design, University of Washington, Seattle, WA98195
- Department of Bioengineering, University of Washington, Seattle, WA98195
- Department of Chemical Engineering, University of Washington, Seattle, WA98195
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA98195
- Department of Chemistry, University of Washington, Seattle, WA98195
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA98195
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
- HHMI, University of Washington, Seattle, WA98195
| |
Collapse
|
22
|
Kureel SK, Blair B, Sheetz MP. Recent Advancement in Elimination Strategies and Potential Rejuvenation Targets of Senescence. Adv Biol (Weinh) 2024; 8:e2300461. [PMID: 37857532 DOI: 10.1002/adbi.202300461] [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: 08/29/2023] [Indexed: 10/21/2023]
Abstract
Cellular senescence is a state of exiting the cell cycle, resisting apoptosis, and changing phenotype. Senescent cells (SCs) can be identified by large, distorted morphology and irreversible inability to replicate. In early development, senescence has beneficial roles like tissue patterning and wound healing, where SCs are cleared by the immune system. However, there is a steep rise in SC number as organisms age. The issue with SC accumulation stems from the loss of cellular function, alterations of the microenvironment, and secretions of pro-inflammatory molecules, consisting of cytokines, chemokines, matrix metalloproteinases (MMPs), interleukins, and extracellular matrix (ECM)-associated molecules. This secreted cocktail is referred to as the senescence-associated secretory phenotype (SASP), a hallmark of cellular senescence. The SASP promotes inflammation and displays a bystander effect where paracrine signaling turns proliferating cells into senescent states. To alleviate age-associated diseases, researchers have developed novel methods and techniques to selectively eliminate SCs in aged individuals. Although studies demonstrated that selectively killing SCs improves age-related disorders, there are drawbacks to SC removal. Considering favorable aspects of senescence in the body, this paper reviews recent advancements in elimination strategies and potential rejuvenation targets of senescence to bring researchers in the field up to date.
Collapse
Affiliation(s)
- Sanjay Kumar Kureel
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Brandon Blair
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Michael P Sheetz
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| |
Collapse
|
23
|
Denoeud C, Luo G, Paquet J, Boisselier J, Wosinski P, Moya A, Diallo A, Larochette N, Marinesco S, Meiller A, Becquart P, Moussi H, Vilquin JT, Logeart-Avramoglou D, Gand A, Larreta-Garde V, Pauthe E, Potier E, Petite H. Enzyme-controlled, nutritive hydrogel for mesenchymal stromal cell survival and paracrine functions. Commun Biol 2023; 6:1266. [PMID: 38092861 PMCID: PMC10719273 DOI: 10.1038/s42003-023-05643-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 11/28/2023] [Indexed: 12/17/2023] Open
Abstract
Culture-adapted human mesenchymal stromal cells (hMSCs) are appealing candidates for regenerative medicine applications. However, these cells implanted in lesions as single cells or tissue constructs encounter an ischemic microenvironment responsible for their massive death post-transplantation, a major roadblock to successful clinical therapies. We hereby propose a paradigm shift for enhancing hMSC survival by designing, developing, and testing an enzyme-controlled, nutritive hydrogel with an inbuilt glucose delivery system for the first time. This hydrogel, composed of fibrin, starch (a polymer of glucose), and amyloglucosidase (AMG, an enzyme that hydrolyze glucose from starch), provides physiological glucose levels to fuel hMSCs via glycolysis. hMSCs loaded in these hydrogels and exposed to near anoxia (0.1% pO2) in vitro exhibited improved cell viability and angioinductive functions for up to 14 days. Most importantly, these nutritive hydrogels promoted hMSC viability and paracrine functions when implanted ectopically. Our findings suggest that local glucose delivery via the proposed nutritive hydrogel can be an efficient approach to improve hMSC-based therapeutic efficacy.
Collapse
Affiliation(s)
- Cyprien Denoeud
- University Paris Cité, CNRS, INSERM, ENVA, B3OA, Paris, France
| | - Guotian Luo
- University Paris Cité, CNRS, INSERM, ENVA, B3OA, Paris, France
| | - Joseph Paquet
- University Paris Cité, CNRS, INSERM, ENVA, B3OA, Paris, France
| | - Julie Boisselier
- Biomaterial for Health Group, ERRMECe, University of Cergy-Pontoise, Cergy-Pontoise, France
| | | | - Adrien Moya
- University Paris Cité, CNRS, INSERM, ENVA, B3OA, Paris, France
| | - Ahmad Diallo
- University Paris Cité, CNRS, INSERM, ENVA, B3OA, Paris, France
| | | | | | - Anne Meiller
- Neuroscience Research Center, AniRA-NeuroChem Platform, Lyon, France
| | - Pierre Becquart
- University Paris Cité, CNRS, INSERM, ENVA, B3OA, Paris, France
| | - Hilel Moussi
- University Paris Cité, CNRS, INSERM, ENVA, B3OA, Paris, France
| | - Jean-Thomas Vilquin
- Sorbonne Université, INSERM, AIM, CNRS, Centre de Recherche en Myologie, Hôpital Pitié Salpêtrière, Paris, France
| | | | - Adeline Gand
- Biomaterial for Health Group, ERRMECe, University of Cergy-Pontoise, Cergy-Pontoise, France
| | | | - Emmanuel Pauthe
- Biomaterial for Health Group, ERRMECe, University of Cergy-Pontoise, Cergy-Pontoise, France
| | - Esther Potier
- University Paris Cité, CNRS, INSERM, ENVA, B3OA, Paris, France
| | - Hervé Petite
- University Paris Cité, CNRS, INSERM, ENVA, B3OA, Paris, France.
| |
Collapse
|
24
|
Ashna M, Senthilkumar N, Sanpui P. Human Hair Keratin-Based Hydrogels in Regenerative Medicine: Current Status and Future Directions. ACS Biomater Sci Eng 2023; 9:5527-5547. [PMID: 37734053 DOI: 10.1021/acsbiomaterials.3c00883] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
Regenerative medicine (RM) is a multidisciplinary field that utilizes the inherent regenerative potential of human cells to generate functionally and physiologically acceptable human cells, tissues, and organs in vivo or ex vivo. An appropriate biomaterial scaffold with desired physicochemical properties constitutes an important component of a successful RM approach. Among various forms of biomaterials explored until the present day, hydrogels have emerged as a versatile candidate for tissue engineering and regenerative medicine (TERM) applications such as scaffolds for spatial patterning and delivering therapeutic agents, or substrates to enhance cell growth, differentiation, and migration. Although hydrogels can be prepared from a variety of synthetic polymers as well as biopolymers, the latter are preferred for their inherent biocompatibility. Specifically, keratins are fibrous proteins that have been recently explored for constructing hydrogels useful for RM purposes. The present review discusses the suitability of keratin-based biomaterials in RM, with a particular focus on human hair keratin hydrogels and their use in various RM applications.
Collapse
Affiliation(s)
- Mymuna Ashna
- Department of Biotechnology, BITS Pilani Dubai Campus, Dubai International Academic City, Dubai, United Arab Emirates
| | - Neeharika Senthilkumar
- Department of Biotechnology, BITS Pilani Dubai Campus, Dubai International Academic City, Dubai, United Arab Emirates
| | - Pallab Sanpui
- Department of Biotechnology, BITS Pilani Dubai Campus, Dubai International Academic City, Dubai, United Arab Emirates
| |
Collapse
|
25
|
Ghorbani S, Christine Füchtbauer A, Møllebjerg A, Møller Martensen P, Hvidbjerg Laursen S, Christian Evar Kraft D, Kjems J, Meyer RL, Rahimi K, Foss M, Füchtbauer EM, Sutherland DS. Protein ligand and nanotopography separately drive the phenotype of mouse embryonic stem cells. Biomaterials 2023; 301:122244. [PMID: 37459700 DOI: 10.1016/j.biomaterials.2023.122244] [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: 01/02/2023] [Revised: 07/09/2023] [Accepted: 07/12/2023] [Indexed: 09/06/2023]
Abstract
Biochemical and biomechanical signals regulate stem cell function in the niche environments in vivo. Current in vitro culture of mouse embryonic stem cells (mESC) uses laminin (LN-511) to provide mimetic biochemical signaling (LN-521 for human systems) to maintain stemness. Alternative approaches propose topographical cues to provide biomechanical cues, however combined biochemical and topographic cues may better mimic the in vivo environment, but are largely unexplored for in vitro stem cell expansion. In this study, we directly compare in vitro signals from LN-511 and/or topographic cues to maintain stemness, using systematically-varied submicron pillar patterns or flat surfaces with or without preadsorbed LN-511. The adhesion of cells, colony formation, expression of the pluripotency marker,octamer-binding transcription factor 4 (Oct4), and transcriptome profiling were characterized. We observed that either biochemical or topographic signals could maintain stemness of mESCs in feeder-free conditions, indicated by high-level Oct4 and gene profiling by RNAseq. The combination of LN-511 with nanotopography reduced colony growth, while maintaining stemness markers, shifted the cellular phenotype indicating that the integration of biochemical and topographic signals is antagonistic. Overall, significantly faster (up to 2.5 times) colony growth was observed at nanotopographies without LN-511, suggesting for improved ESC expansion.
Collapse
Affiliation(s)
- Sadegh Ghorbani
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark; The Centre for Cellular Signal Patterns (CELLPAT), Gustav Wieds Vej 14, Aarhus C, 8000, Denmark; Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA.
| | | | - Andreas Møllebjerg
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark
| | | | - Sara Hvidbjerg Laursen
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark
| | - David Christian Evar Kraft
- Department of Dentistry and Oral Health, Faculty of Health, University of Aarhus, Aarhus C, 8000, Denmark
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark; The Centre for Cellular Signal Patterns (CELLPAT), Gustav Wieds Vej 14, Aarhus C, 8000, Denmark; Department of Molecular Biology, University of Aarhus, Aarhus C, 8000, Denmark
| | - Rikke Louise Meyer
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark
| | - Karim Rahimi
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark; Department of Molecular Biology, University of Aarhus, Aarhus C, 8000, Denmark
| | - Morten Foss
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark
| | | | - Duncan S Sutherland
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark; The Centre for Cellular Signal Patterns (CELLPAT), Gustav Wieds Vej 14, Aarhus C, 8000, Denmark.
| |
Collapse
|
26
|
Rubio-Lara JA, Igarashi KJ, Sood S, Johansson A, Sommerkamp P, Yamashita M, Lin DS. Expanding hematopoietic stem cell ex vivo: recent advances and technical considerations. Exp Hematol 2023; 125-126:6-15. [PMID: 37543237 DOI: 10.1016/j.exphem.2023.07.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/07/2023]
Abstract
Hematopoietic stem cells (HSCs) are the most primitive cell type in the hematopoietic hierarchy, which are responsible for sustaining the lifelong production of mature blood and immune cells. Due to their superior long-term regenerative capacity, HSC therapies such as stem cell transplantation have been used in a broad range of hematologic disorders. However, the rarity of this population in vivo considerably limits its clinical applications and large-scale analyses such as screening and safety studies. Therefore, ex vivo culture methods that allow long-term expansion and maintenance of functional HSCs are instrumental in overcoming the difficulties in studying HSC biology and improving HSC therapies. In this perspective, we discuss recent advances and technical considerations for three ex vivo HSC expansion methods including 1) polyvinyl alcohol-based HSC expansion, 2) mesenchymal stromal cell-HSC co-culture, and 3) two-/three-dimensional hydrogel HSC culture. This review summarizes the presentations and discussions from the 2022 International Society for Experimental Hematology (ISEH) Annual Meeting New Investigator Technology Session.
Collapse
Affiliation(s)
| | - Kyomi J Igarashi
- Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Shubhankar Sood
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany; Division of Inflammatory Stress in Stem Cells, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alban Johansson
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Pia Sommerkamp
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Masayuki Yamashita
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Dawn S Lin
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
27
|
Huang F, He Y, Zhang M, Luo K, Li J, Li J, Zhang X, Dong X, Tang J. Progress in Research on Stem Cells in Neonatal Refractory Diseases. J Pers Med 2023; 13:1281. [PMID: 37623531 PMCID: PMC10455340 DOI: 10.3390/jpm13081281] [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: 06/23/2023] [Revised: 08/03/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023] Open
Abstract
With the development and progress of medical technology, the survival rate of premature and low-birth-weight infants has increased, as has the incidence of a variety of neonatal diseases, such as hypoxic-ischemic encephalopathy, intraventricular hemorrhage, bronchopulmonary dysplasia, necrotizing enterocolitis, and retinopathy of prematurity. These diseases cause severe health conditions with poor prognoses, and existing control methods are ineffective for such diseases. Stem cells are a special type of cells with self-renewal and differentiation potential, and their mechanisms mainly include anti-inflammatory and anti-apoptotic properties, reducing oxidative stress, and boosting regeneration. Their paracrine effects can affect the microenvironment in which they survive, thereby affecting the biological characteristics of other cells. Due to their unique abilities, stem cells have been used in treating various diseases. Therefore, stem cell therapy may open up the possibility of treating such neonatal diseases. This review summarizes the research progress on stem cells and exosomes derived from stem cells in neonatal refractory diseases to provide new insights for most researchers and clinicians regarding future treatments. In addition, the current challenges and perspectives in stem cell therapy are discussed.
Collapse
Affiliation(s)
- Fangjun Huang
- Department of Neonatology, West China Second Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Yang He
- Department of Neonatology, West China Second Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Meng Zhang
- Department of Neonatology, West China Second Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Keren Luo
- Department of Neonatology, West China Second Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Jiawen Li
- Department of Neonatology, West China Second Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Jiali Li
- Department of Neonatology, West China Second Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Xinyu Zhang
- Department of Neonatology, West China Second Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Xiaoyan Dong
- Department of Neonatology, West China Second Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Jun Tang
- Department of Neonatology, West China Second Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| |
Collapse
|
28
|
Zhang W, Zha K, Hu W, Xiong Y, Knoedler S, Obed D, Panayi AC, Lin Z, Cao F, Mi B, Liu G. Multifunctional hydrogels: advanced therapeutic tools for osteochondral regeneration. Biomater Res 2023; 27:76. [PMID: 37542353 PMCID: PMC10403923 DOI: 10.1186/s40824-023-00411-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/05/2023] [Indexed: 08/06/2023] Open
Abstract
Various joint pathologies such as osteochondritis dissecans, osteonecrosis, rheumatic disease, and trauma, may result in severe damage of articular cartilage and other joint structures, ranging from focal defects to osteoarthritis (OA). The osteochondral unit is one of the critical actors in this pathophysiological process. New approaches and applications in tissue engineering and regenerative medicine continue to drive the development of OA treatment. Hydrogel scaffolds, a component of tissue engineering, play an indispensable role in osteochondral regeneration. In this review, tissue engineering strategies regarding osteochondral regeneration were highlighted and summarized. The application of hydrogels for osteochondral regeneration within the last five years was evaluated with an emphasis on functionalized physical and chemical properties of hydrogel scaffolds, functionalized delivery hydrogel scaffolds as well as functionalized intelligent response hydrogel scaffolds. Lastly, to serve as guidance for future efforts in the creation of bioinspired hydrogel scaffolds, a succinct summary and new views for specific mechanisms, applications, and existing limitations of the newly designed functionalized hydrogel scaffolds were offered.
Collapse
Affiliation(s)
- Wenqian Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Kangkang Zha
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Weixian Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Yuan Xiong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Samuel Knoedler
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02152, USA
| | - Doha Obed
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02152, USA
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Adriana C Panayi
- Department of Hand, Plastic and Reconstructive Surgery, Microsurgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, 67071, Ludwigshafen/Rhine, Germany
| | - Ze Lin
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Faqi Cao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| | - Bobin Mi
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore.
| | - Guohui Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| |
Collapse
|
29
|
Wu IC, Liou JW, Yang CH, Chen JH, Chen KY, Hung CH. Self-assembly of gelatin and collagen in the polyvinyl alcohol substrate and its influence on cell adhesion, proliferation, shape, spreading and differentiation. Front Bioeng Biotechnol 2023; 11:1193849. [PMID: 37520293 PMCID: PMC10375239 DOI: 10.3389/fbioe.2023.1193849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 07/03/2023] [Indexed: 08/01/2023] Open
Abstract
Culture substrates display profound influence on biological and developmental characteristic of cells cultured in vitro. This study investigates the influence of polyvinyl alcohol (PVA) substrates blended with different concentration of collagen or/and gelatin on the cell adhesion, proliferation, shape, spreading, and differentiation of stem cells. The collagen/gelatin blended PVA substrates were prepared by air drying. During drying, blended collagen or/and gelatin can self-assemble into macro-scale nucleated particles or branched fibrils in the PVA substrates that can be observed under the optical microscope. These collagen/gelatin blended substrates revealed different surface topography, z-average, roughness, surface adhesion and Young's modulus as examined by the atomic force microscope (AFM). The results of Fourier transform infrared spectroscopy (FTIR) analysis indicated that the absorption of amide I (1,600-1,700 cm-1) and amide II (1,500-1,600 cm-1) groups increased with increasing collagen and gelatin concentration blended and the potential of fibril formation. These collagen or/and gelatin blended PVA substrates showed enhanced NIH-3T3 fibroblast adhesion as comparing with the pure PVA, control tissue culture polystyrene, conventional collagen-coated and gelatin-coated wells. These highly adhesive PVA substrates also exhibit inhibited cell spreading and proliferation. It is also found that the shape of NIH-3T3 fibroblasts can be switched between oval, spindle and flattened shapes depending on the concentration of collagen or/and gelatin blended. For inductive differentiation of stem cells, it is found that number and ration of neural differentiation of rat cerebral cortical neural stem cells increase with the decreasing collagen concentration in the collagen-blended PVA substrates. Moreover, the PVA substrates blended with collagen or collagen and gelatin can efficiently support and conduct human pluripotent stem cells to differentiate into Oil-Red-O- and UCP-1-positive brown-adipocyte-like cells via ectodermal lineage without the addition of mitogenic factors. These results provide a useful and alternative platform for controlling cell behavior in vitro and may be helpful for future application in the field of regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- I-Chi Wu
- Institute of Medical Sciences, Tzu Chi University, Hualien City, Taiwan
- Plastic Surgery Division, Surgical Department, Hualien Armed Forces General Hospital, Hualien City, Taiwan
| | - Je-Wen Liou
- Institute of Medical Sciences, Tzu Chi University, Hualien City, Taiwan
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien City, Taiwan
| | - Chin-Hao Yang
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien City, Taiwan
| | - Jia-Hui Chen
- Institute of Medical Sciences, Tzu Chi University, Hualien City, Taiwan
- Department of Surgery, Taipei Tzu Chi Hospital, New Taipei City, Taiwan
| | - Kuan-Yu Chen
- Institute of Medical Sciences, Tzu Chi University, Hualien City, Taiwan
- Department of Surgery, New Taipei City Hospital, New Taipei City, Taiwan
| | - Chih-Huang Hung
- Institute of Medical Sciences, Tzu Chi University, Hualien City, Taiwan
| |
Collapse
|
30
|
Zhang W, Huang X. Stem cell-based drug delivery strategy for skin regeneration and wound healing: potential clinical applications. Inflamm Regen 2023; 43:33. [PMID: 37391780 DOI: 10.1186/s41232-023-00287-1] [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: 03/10/2023] [Accepted: 06/20/2023] [Indexed: 07/02/2023] Open
Abstract
Stem cell-based therapy is widely accepted to be a promising strategy in tissue regenerative medicine. Nevertheless, there are several obstacles to applying stem cells in skin regeneration and wound healing, which includes determining the optimum source, the processing and administration methods of stem cells, and the survival and functions of stem cells in wound sites. Owing to the limitations of applying stem cells directly, this review aims to discuss several stem cell-based drug delivery strategies in skin regeneration and wound healing and their potential clinical applications. We introduced diverse types of stem cells and their roles in wound repair. Moreover, the stem cell-based drug delivery systems including stem cell membrane-coated nanoparticles, stem cell-derived extracellular vesicles, stem cell as drug carriers, scaffold-free stem cell sheets, and stem cell-laden scaffolds were further investigated in the field of skin regeneration and wound healing. More importantly, stem cell membrane-coating nanotechnology confers great advantages compared to other drug delivery systems in a broad field of biomedical contexts. Taken together, the stem cell-based drug delivery strategy holds great promise for treating skin regeneration and wound healing.
Collapse
Affiliation(s)
- Weiyue Zhang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xin Huang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
31
|
Mout R, Bretherton RC, Decarreau J, Lee S, Edman NI, Ahlrichs M, Hsia Y, Sahtoe DD, Ueda G, Gregorio N, Sharma A, Schulman R, DeForest CA, Baker D. De novo design of modular protein hydrogels with programmable intra- and extracellular viscoelasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.02.543449. [PMID: 37398067 PMCID: PMC10312586 DOI: 10.1101/2023.06.02.543449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Relating the macroscopic properties of protein-based materials to their underlying component microstructure is an outstanding challenge. Here, we exploit computational design to specify the size, flexibility, and valency of de novo protein building blocks, as well as the interaction dynamics between them, to investigate how molecular parameters govern the macroscopic viscoelasticity of the resultant protein hydrogels. We construct gel systems from pairs of symmetric protein homo-oligomers, each comprising 2, 5, 24, or 120 individual protein components, that are crosslinked either physically or covalently into idealized step-growth biopolymer networks. Through rheological assessment and molecular dynamics (MD) simulation, we find that the covalent linkage of multifunctional precursors yields hydrogels whose viscoelasticity depends on the crosslink length between the constituent building blocks. In contrast, reversibly crosslinking the homo-oligomeric components with a computationally designed heterodimer results in non-Newtonian biomaterials exhibiting fluid-like properties under rest and low shear, but shear-stiffening solid-like behavior at higher frequencies. Exploiting the unique genetic encodability of these materials, we demonstrate the assembly of protein networks within living mammalian cells and show via fluorescence recovery after photobleaching (FRAP) that mechanical properties can be tuned intracellularly, in correlation with matching formulations formed extracellularly. We anticipate that the ability to modularly construct and systematically program the viscoelastic properties of designer protein-based materials could have broad utility in biomedicine, with applications in tissue engineering, therapeutic delivery, and synthetic biology.
Collapse
Affiliation(s)
- Rubul Mout
- Department of Biochemistry, University of Washington, Seattle, WA 98195
- Institute for Protein Design, University of Washington, Seattle, WA 98195
- Stem Cell Program at Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Ross C. Bretherton
- Department of Bioengineering, University of Washington, Seattle, WA 98195
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195
- Department of Chemistry, University of Washington, Seattle, WA 98195
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA 98195
| | - Justin Decarreau
- Department of Biochemistry, University of Washington, Seattle, WA 98195
- Institute for Protein Design, University of Washington, Seattle, WA 98195
| | - Sangmin Lee
- Department of Biochemistry, University of Washington, Seattle, WA 98195
- Institute for Protein Design, University of Washington, Seattle, WA 98195
| | - Natasha I. Edman
- Department of Biochemistry, University of Washington, Seattle, WA 98195
- Institute for Protein Design, University of Washington, Seattle, WA 98195
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA 98195
- Medical Scientist Training Program, University of Washington, Seattle, WA 98195
| | - Maggie Ahlrichs
- Department of Biochemistry, University of Washington, Seattle, WA 98195
- Institute for Protein Design, University of Washington, Seattle, WA 98195
| | - Yang Hsia
- Department of Biochemistry, University of Washington, Seattle, WA 98195
- Institute for Protein Design, University of Washington, Seattle, WA 98195
| | - Danny D. Sahtoe
- Department of Biochemistry, University of Washington, Seattle, WA 98195
- Institute for Protein Design, University of Washington, Seattle, WA 98195
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195
| | - George Ueda
- Department of Biochemistry, University of Washington, Seattle, WA 98195
- Institute for Protein Design, University of Washington, Seattle, WA 98195
| | - Nicole Gregorio
- Department of Bioengineering, University of Washington, Seattle, WA 98195
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195
- Department of Chemistry, University of Washington, Seattle, WA 98195
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA 98195
| | - Alee Sharma
- Stem Cell Program at Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Rebecca Schulman
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218
- Department of Computer Science, Johns Hopkins University, Baltimore, MD 21218
| | - Cole A. DeForest
- Institute for Protein Design, University of Washington, Seattle, WA 98195
- Department of Bioengineering, University of Washington, Seattle, WA 98195
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195
- Department of Chemistry, University of Washington, Seattle, WA 98195
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA 98195
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA 98195
- Institute for Protein Design, University of Washington, Seattle, WA 98195
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195
| |
Collapse
|
32
|
Yang U, Kang B, Yong MJ, Yang DH, Choi SY, Je JH, Oh SS. Type-Independent 3D Writing and Nano-Patterning of Confined Biopolymers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207403. [PMID: 36825681 PMCID: PMC10161081 DOI: 10.1002/advs.202207403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/07/2023] [Indexed: 05/06/2023]
Abstract
Biopolymers are essential building blocks that constitute cells and tissues with well-defined molecular structures and diverse biological functions. Their three-dimensional (3D) complex architectures are used to analyze, control, and mimic various cells and their ensembles. However, the free-form and high-resolution structuring of various biopolymers remain challenging because their structural and rheological control depend critically on their polymeric types at the submicron scale. Here, direct 3D writing of intact biopolymers is demonstrated using a systemic combination of nanoscale confinement, evaporation, and solidification of a biopolymer-containing solution. A femtoliter solution is confined in an ultra-shallow liquid interface between a fine-tuned nanopipette and a chosen substrate surface to achieve directional growth of biopolymer nanowires via solvent-exclusive evaporation and concurrent solution supply. The evaporation-dependent printing is biopolymer type-independent, therefore, the 3D motor-operated precise nanopipette positioning allows in situ printing of nucleic acids, polysaccharides, and proteins with submicron resolution. By controlling concentrations and molecular weights, several different biopolymers are reproducibly patterned with desired size and geometry, and their 3D architectures are biologically active in various solvents with no structural deformation. Notably, protein-based nanowire patterns exhibit pin-point localization of spatiotemporal biofunctions, including target recognition and catalytic peroxidation, indicating their application potential in organ-on-chips and micro-tissue engineering.
Collapse
Affiliation(s)
- Un Yang
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, South Korea
| | - Byunghwa Kang
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, South Korea
| | - Moon-Jung Yong
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, South Korea
| | - Dong-Hwan Yang
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, South Korea
| | - Si-Young Choi
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, South Korea
| | - Jung Ho Je
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, South Korea
- Nanoblesse, 85-11 (4th fl.) Namwon-Ro, Pohang, Gyeongbuk, 37883, South Korea
| | - Seung Soo Oh
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, South Korea
- Institute for Convergence Research and Education in Advanced Technology (I-CREATE), Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, South Korea
| |
Collapse
|
33
|
Abdal Dayem A, Lee SB, Lim KM, Kim A, Shin HJ, Vellingiri B, Kim YB, Cho SG. Bioactive peptides for boosting stem cell culture platform: Methods and applications. Biomed Pharmacother 2023; 160:114376. [PMID: 36764131 DOI: 10.1016/j.biopha.2023.114376] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Peptides, short protein fragments, can emulate the functions of their full-length native counterparts. Peptides are considered potent recombinant protein alternatives due to their specificity, high stability, low production cost, and ability to be easily tailored and immobilized. Stem cell proliferation and differentiation processes are orchestrated by an intricate interaction between numerous growth factors and proteins and their target receptors and ligands. Various growth factors, functional proteins, and cellular matrix-derived peptides efficiently enhance stem cell adhesion, proliferation, and directed differentiation. For that, peptides can be immobilized on a culture plate or conjugated to scaffolds, such as hydrogels or synthetic matrices. In this review, we assess the applications of a variety of peptides in stem cell adhesion, culture, organoid assembly, proliferation, and differentiation, describing the shortcomings of recombinant proteins and their full-length counterparts. Furthermore, we discuss the challenges of peptide applications in stem cell culture and materials design, as well as provide a brief outlook on future directions to advance peptide applications in boosting stem cell quality and scalability for clinical applications in tissue regeneration.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Soo Bin Lee
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Kyung Min Lim
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Aram Kim
- Department of Urology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Hyun Jin Shin
- Department of Ophthalmology, Research Institute of Medical Science, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda 151401, Punjab, India
| | - Young Bong Kim
- Department of Biomedical Science & Engineering, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
34
|
Mobarakeh ZT, Hasanzadeh E, Farzin A, Goodarzi A, Farahani MS, Shirian S, Mahmoodi N, Zamani N, Karimi A, Ai J. Enhanced sciatic nerve regeneration with fibrin scaffold containing human endometrial stem cells and insulin encapsulated chitosan particles: An in vivo study. Injury 2023:S0020-1383(23)00082-7. [PMID: 36894467 DOI: 10.1016/j.injury.2023.01.041] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 08/05/2022] [Accepted: 01/23/2023] [Indexed: 03/11/2023]
Abstract
BACKGROUND Based on recent advances in tissue engineering and stem cell therapy in nervous system diseases treatments, this study aimed to investigate sciatic nerve regeneration using human endometrial stem cells (hEnSCs) encapsulated fibrin gel containing chitosan nanoparticle loaded by insulin (Ins-CPs). Stem cells and also Insulin (Ins), which is a strong signaling molecule in peripheral nerve regeneration, play an important role in neural tissue engineering. METHODS The fibrin hydrogel scaffold containing insulin loaded chitosan particles was synthesized and characterized. Release profiles of insulin from hydrogel was determined through UV-visible spectroscopy. Also, human endometrial stem cells encapsulated in hydrogel and its cell biocompatibility were assigned. Furthermore, the sciatic nerve crush injury was carried out and prepared fibrin gel was injected at the crush injury site by an 18-gage needle. Eight and twelve weeks later, the recovery of motor and sensory function and histopathological evaluation were assessed. RESULTS The in vitro experiments showed that the insulin can promote hEnSCs proliferation within a certain concentration range. Animals' treatment confirmed that developed fibrin gel containing Ins-CPs and hEnSCs significantly improves motor function and sensory recovery. Hematoxylin and Eosin (H&E) images provided from cross-sectional and, longitudinal-sections of the harvested regenerative nerve showed that regenerative nerve fibers had been formed and accompanied with new blood vessels in the fibrin/insulin/hEnSCs group. CONCLUSION Our results demonstrated that the prepared hydrogel scaffolds containing insulin nanoparticles and hEnSCs could be considered as a potential biomaterial aimed at regeneration of sciatic nerves.
Collapse
Affiliation(s)
- Zahra Taherian Mobarakeh
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Hasanzadeh
- Immunogenetics Research Center, Department of Tissue Engineering & Regenerative Medicine, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ali Farzin
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Goodarzi
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Morteza Sagharjoghi Farahani
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sadegh Shirian
- Department of Pathology, School of Veterinary Medicine, Sharekord University, Shahrekord, Iran
| | - Narges Mahmoodi
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Narges Zamani
- Department of Obstetrics and Gynecology, Emam Complex Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Anita Karimi
- Chronic Respiratory Diseases Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
35
|
Boinapalli Y, Shankar Pandey R, Singh Chauhan A, Sudheesh MS. Physiological relevance of in-vitro cell-nanoparticle interaction studies as a predictive tool in cancer nanomedicine research. Int J Pharm 2023; 632:122579. [PMID: 36603671 DOI: 10.1016/j.ijpharm.2022.122579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/19/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023]
Abstract
Cell uptake study is a routine experiment used as a surrogate to predict in vivo response in cancer nanomedicine research. Cell culture conditions should be designed in such a way that it emulates 'real' physiological conditions and avoid artefacts. It is critical to dissect the steps involved in cellular uptake to understand the physical, chemical, and biological factors responsible for particle internalization. The two-dimensional model (2D) of cell culture is overly simplistic to mimic the complexity of cancer tissues that exist in vivo. It cannot simulate the critical tissue-specific properties like cell-cell interaction and cell-extracellular matrix (ECM) interaction and its influences on the temporal and spatial distribution of nanoparticles (NPs). The three dimensional model organization of heterogenous cancer and normal cells with the ECM acts as a formidable barrier to NP penetration and cellular uptake. The three dimensional cell culture (3D) technology is a breakthrough in this direction that can mimic the barrier properties of the tumor microenvironment (TME). Herein, we discuss the physiological factors that should be considered to bridge the translational gap between in and vitro cell culture studies and in-vivo studies in cancer nanomedicine.
Collapse
Affiliation(s)
- Yamini Boinapalli
- Dept. of Pharmaceutics, Amrita School of Pharmacy, Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi 682041, India
| | - Ravi Shankar Pandey
- SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, C.G. 495009, India
| | - Abhay Singh Chauhan
- Department of Biopharmaceutical Sciences, School of Pharmacy, Medical College of Wisconsin, Milwaukee, WI 53226, United States.
| | - M S Sudheesh
- Dept. of Pharmaceutics, Amrita School of Pharmacy, Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi 682041, India.
| |
Collapse
|
36
|
Álvarez Z, Ortega JA, Sato K, Sasselli IR, Kolberg-Edelbrock AN, Qiu R, Marshall KA, Nguyen TP, Smith CS, Quinlan KA, Papakis V, Syrgiannis Z, Sather NA, Musumeci C, Engel E, Stupp SI, Kiskinis E. Artificial extracellular matrix scaffolds of mobile molecules enhance maturation of human stem cell-derived neurons. Cell Stem Cell 2023; 30:219-238.e14. [PMID: 36638801 PMCID: PMC9898161 DOI: 10.1016/j.stem.2022.12.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/04/2022] [Accepted: 12/13/2022] [Indexed: 01/13/2023]
Abstract
Human induced pluripotent stem cell (hiPSC) technologies offer a unique resource for modeling neurological diseases. However, iPSC models are fraught with technical limitations including abnormal aggregation and inefficient maturation of differentiated neurons. These problems are in part due to the absence of synergistic cues of the native extracellular matrix (ECM). We report on the use of three artificial ECMs based on peptide amphiphile (PA) supramolecular nanofibers. All nanofibers display the laminin-derived IKVAV signal on their surface but differ in the nature of their non-bioactive domains. We find that nanofibers with greater intensity of internal supramolecular motion have enhanced bioactivity toward hiPSC-derived motor and cortical neurons. Proteomic, biochemical, and functional assays reveal that highly mobile PA scaffolds caused enhanced β1-integrin pathway activation, reduced aggregation, increased arborization, and matured electrophysiological activity of neurons. Our work highlights the importance of designing biomimetic ECMs to study the development, function, and dysfunction of human neurons.
Collapse
Affiliation(s)
- Zaida Álvarez
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA; Department of Medicine, Northwestern University, Chicago, IL 60611, USA; Biomaterials for Regenerative Therapies, Institute for Bioengineering of Catalonia (IBEC), Barcelona 08028, Spain
| | - J Alberto Ortega
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona 08907, Spain
| | - Kohei Sato
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA; Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
| | - Ivan R Sasselli
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA; Department of Chemistry, Northwestern University, Evanston, IL 60208, USA; Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián 20014, Spain
| | - Alexandra N Kolberg-Edelbrock
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA; Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Ruomeng Qiu
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA; Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
| | - Kelly A Marshall
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Thao Phuong Nguyen
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Cara S Smith
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA; Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Katharina A Quinlan
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Vasileios Papakis
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Zois Syrgiannis
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA; Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
| | - Nicholas A Sather
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA
| | - Chiara Musumeci
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Elisabeth Engel
- Biomaterials for Regenerative Therapies, Institute for Bioengineering of Catalonia (IBEC), Barcelona 08028, Spain
| | - Samuel I Stupp
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA; Department of Chemistry, Northwestern University, Evanston, IL 60208, USA; Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA; Department of Materials Science and Engineering, Northwestern University, Evanston, IL 60208, USA; Department of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Evangelos Kiskinis
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA; The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
37
|
Lu T, Xia B, Chen G. Advances in polymer-based cell encapsulation and its applications in tissue repair. Biotechnol Prog 2023; 39:e3325. [PMID: 36651921 DOI: 10.1002/btpr.3325] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/06/2023] [Accepted: 01/11/2023] [Indexed: 01/19/2023]
Abstract
Cell microencapsulation is a more widely accepted area of biological encapsulation. In most cases, it involves fixing cells in polymer scaffolds or semi-permeable hydrogel capsules, providing the environment for protecting cells, allowing the exchange of nutrients and oxygen, and protecting cells against the attack of the host immune system by preventing the entry of antibodies and cytotoxic immune cells. Hydrogel encapsulation provides a three-dimensional (3D) environment similar to that experienced in vivo, so it can maintain normal cellular functions to produce tissues similar to those in vivo. Embedded cells can be genetically modified to release specific therapeutic products directly at the target site, thereby eliminating the side effects of systemic treatments. Cellular microcarriers need to meet many extremely high standards regarding their biocompatibility, cytocompatibility, immunoseparation capacity, transport, mechanical, and chemical properties. In this article, we discuss the biopolymer gels used in tissue engineering applications and the brief introduction of cell encapsulation for therapeutic protein production. Also, we review polymer biomaterials and methods for preparing cell microcarriers for biomedical applications. At the same time, in order to improve the application performance of cell microcarriers in vivo, we also summarize the main limitations and improvement strategies of cell encapsulation. Finally, the main applications of polymer cell microcarriers in regenerative medicine are summarized.
Collapse
Affiliation(s)
- Tangfang Lu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, People's Republic of China
| | - Bin Xia
- Engineering Research Center for Waste Oil Recovery Technology and Equipment, Ministry of Education, Chongqing Technology and Business University, Chongqing, People's Republic of China
| | - Guobao Chen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, People's Republic of China
| |
Collapse
|
38
|
Ahn H, Cho Y, Yun GT, Jung KB, Jeong W, Kim Y, Son MY, Lee E, Im SG, Jung HT. Hierarchical Topography with Tunable Micro- and Nanoarchitectonics for Highly Enhanced Cardiomyocyte Maturation via Multi-Scale Mechanotransduction. Adv Healthc Mater 2023; 12:e2202371. [PMID: 36652539 DOI: 10.1002/adhm.202202371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/12/2023] [Indexed: 01/19/2023]
Abstract
Enhancing cardiomyocyte (CM) maturation by topographical cues is a critical issue in cardiac tissue engineering. Thus far, single-scale topographies with a broad range of feature shapes and dimensions have been utilized including grooves, pillars, and fibers. This study reports for the first time a hierarchical structure composed of nano-pillars (nPs) on micro-wrinkles (µWs) for effective maturation of CMs. Through capillary force lithography followed by a wrinkling process, vast size ranges of topographies are fabricated, and the responses of CMs are systematically investigated. Maturation of CMs on the hierarchical structures is highly enhanced compared to a single-scale topography: cardiac differentiation of H9C2s (rat cardiomyocytes) on the hierarchical topography is ≈ 2.8 and ≈ 1.9 times higher than those consisting of single-scale µWs and nPs. Both nPs and µWs have important roles in cardiac maturation, and the aspect ratio (height/diameter) of the nPs and the wavelength of the µWs are important in CM maturation. This enhancement is caused by strong focal adhesion and nucleus mediated mechanotransduction of CMs from the confinement effects of the different wavelengths of µWs and the cellular membrane protrusion on the nPs. This study demonstrates how a large family of hierarchical structures is used for cardiac maturation.
Collapse
Affiliation(s)
- Hyunah Ahn
- National Laboratory for Organic Opto-Electronic Material, Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Korea
| | - Younghak Cho
- Functional Thin Film Laboratory, Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Korea.,KI for NanoCentury, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Korea
| | - Geun-Tae Yun
- National Nanofab Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Korea
| | - Kwang Bo Jung
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
| | - Wonji Jeong
- Functional Thin Film Laboratory, Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Korea.,KI for NanoCentury, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Korea
| | - Yesol Kim
- Functional Thin Film Laboratory, Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Korea.,KI for NanoCentury, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Korea
| | - Mi-Young Son
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
| | - Eunjung Lee
- Functional Thin Film Laboratory, Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Korea.,KI for NanoCentury, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Korea
| | - Sung Gap Im
- Functional Thin Film Laboratory, Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Korea.,KI for NanoCentury, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Korea
| | - Hee-Tae Jung
- National Laboratory for Organic Opto-Electronic Material, Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Korea
| |
Collapse
|
39
|
Xiao W, Yang Y, Chu C, Rung SA, Wang Z, Man Y, Lin J, Qu Y. Macrophage response mediated by extracellular matrix: recent progress. Biomed Mater 2023; 18. [PMID: 36595269 DOI: 10.1088/1748-605x/aca946] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 12/06/2022] [Indexed: 12/12/2022]
Abstract
Biomaterials are one of efficient treatment options for tissue defects in regenerative medicine. Compared to synthetic materials which tend to induce chronic inflammatory response and fibrous capsule, extracellular matrix (ECM) scaffold materials composed of biopolymers are thought to be capable of inducing a pro-regenerative immune microenvironment and facilitate wound healing. Immune cells are the first line of response to implanted biomaterials. In particular, macrophages greatly affect cell behavior and the ultimate treatment outcome based on multiple cell phenotypes with various functions. The macrophage polarization status is considered as a general reflection of the characteristics of the immune microenvironment. Since numerous reports has emphasized the limitation of classical M1/M2 nomenclature, high-resolution techniques such as single-cell sequencing has been applied to recognize distinct macrophage phenotypes involved in host responses to biomaterials. After reviewing latest literatures that explored the immune microenvironment mediated by ECM scaffolds, this paper describe the behaviors of highly heterogeneous and plastic macrophages subpopulations which affect the tissue regeneration. The mechanisms by which ECM scaffolds interact with macrophages are also discussed from the perspectives of the ECM ultrastructure along with the nucleic acid, protein, and proteoglycan compositions, in order to provide targets for potential therapeutic modulation in regenerative medicine.
Collapse
Affiliation(s)
- Wenlan Xiao
- Department of Oral Implantology & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Yang Yang
- Department of Oral Implantology & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Chenyu Chu
- Department of Oral Implantology & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Sheng-An Rung
- Department of Oral Implantology & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Zhanqi Wang
- Department of Oral Implantology & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Yi Man
- Department of Oral Implantology & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Jie Lin
- Department of Oral Implantology & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Yili Qu
- Department of Oral Implantology & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| |
Collapse
|
40
|
Gupta P, Waghmare S, Kar S, Illath K, Rao S, Santra TS. Functionally gradient three-dimensional graphene foam-based polymeric scaffolds for multilayered tissue regeneration. RSC Adv 2023; 13:1245-1255. [PMID: 36686898 PMCID: PMC9812017 DOI: 10.1039/d2ra06018c] [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: 09/23/2022] [Accepted: 12/20/2022] [Indexed: 01/05/2023] Open
Abstract
Physiological bioengineering of multilayered tissues requires an optimized geometric organization with comparable biomechanics. Currently, polymer-reinforced three-dimensional (3D) graphene foams (GFs) are gaining interest in tissue engineering due to their unique morphology, biocompatibility, and similarity to extracellular matrixes. However, the homogeneous reinforcement of single polymers throughout a GF matrix does not provide tissue-level organization. Therefore, a triple-layered structure is developed in a GF matrix to closely mimic native tissue structures of the periodontium of the teeth. The scaffold aims to overcome the issue of layer separation, which generally occurs in multilayered structures due to the poor integration of various layers. The 3D GF matrix was reinforced with a polycaprolactone (PCL), polyvinyl alcohol (PVA), and PCL-hydroxyapatite (HA) mixture, added sequentially, via spin coating, vacuum, and hot air drying. Later, PVA was dissolved to create a middle layer, mimicking the periodontal fibers, while the layers present on either side resembled cementum and alveolar bone, respectively. Scanning electron microscopy and micro-computed tomography revealed the structure of the scaffold with internal differential porosities. The nanoindentation and tensile testing demonstrated the closeness of mechanical properties to that of native tissues. The biocompatibility was assessed by the MTT assay with MG63 cells (human osteosarcoma cells) exhibiting high adhesion and proliferation rate inside the 3D architecture. Summing up, this scaffold has the potential for enhancing the regeneration of various multilayered tissues.
Collapse
Affiliation(s)
- Pallavi Gupta
- Department of Engineering Design, Indian Institute of Technology MadrasChennai 600036India+91-9840041787+91-044-2257-4747
| | - Sonali Waghmare
- Department of Engineering Design, Indian Institute of Technology MadrasChennai 600036India+91-9840041787+91-044-2257-4747
| | - Srabani Kar
- Indian Institute of Science Education and ResearchTirupati 517507India
| | - Kavitha Illath
- Department of Engineering Design, Indian Institute of Technology MadrasChennai 600036India+91-9840041787+91-044-2257-4747
| | - Suresh Rao
- Department of Engineering Design, Indian Institute of Technology MadrasChennai 600036India+91-9840041787+91-044-2257-4747
| | - Tuhin Subhra Santra
- Department of Engineering Design, Indian Institute of Technology MadrasChennai 600036India+91-9840041787+91-044-2257-4747
| |
Collapse
|
41
|
Mongera A, Pochitaloff M, Gustafson HJ, Stooke-Vaughan GA, Rowghanian P, Kim S, Campàs O. Mechanics of the cellular microenvironment as probed by cells in vivo during zebrafish presomitic mesoderm differentiation. NATURE MATERIALS 2023; 22:135-143. [PMID: 36577855 PMCID: PMC9812792 DOI: 10.1038/s41563-022-01433-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 11/03/2022] [Indexed: 05/19/2023]
Abstract
Tissue morphogenesis, homoeostasis and repair require cells to constantly monitor their three-dimensional microenvironment and adapt their behaviours in response to local biochemical and mechanical cues. Yet the mechanical parameters of the cellular microenvironment probed by cells in vivo remain unclear. Here, we report the mechanics of the cellular microenvironment that cells probe in vivo and in situ during zebrafish presomitic mesoderm differentiation. By quantifying both endogenous cell-generated strains and tissue mechanics, we show that individual cells probe the stiffness associated with deformations of the supracellular, foam-like tissue architecture. Stress relaxation leads to a perceived microenvironment stiffness that decreases over time, with cells probing the softest regime. We find that most mechanical parameters, including those probed by cells, vary along the anteroposterior axis as mesodermal progenitors differentiate. These findings expand our understanding of in vivo mechanosensation and might aid the design of advanced scaffolds for tissue engineering applications.
Collapse
Affiliation(s)
- Alessandro Mongera
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
- Department of Pathology, Brigham and Women's Hospital and Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Marie Pochitaloff
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
| | - Hannah J Gustafson
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
- Biomolecular Science and Engineering Program, University of California, Santa Barbara, CA, USA
| | | | - Payam Rowghanian
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
| | - Sangwoo Kim
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
| | - Otger Campàs
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA.
- Center for Systems Biology Dresden, Dresden, Germany.
- Cluster of Excellence Physics of Life, TU Dresden, Dresden, Germany.
| |
Collapse
|
42
|
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: 74] [Impact Index Per Article: 37.0] [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
|
43
|
Cao Y, Tan J, Zhao H, Deng T, Hu Y, Zeng J, Li J, Cheng Y, Tang J, Hu Z, Hu K, Xu B, Wang Z, Wu Y, Lobie PE, Ma S. Bead-jet printing enabled sparse mesenchymal stem cell patterning augments skeletal muscle and hair follicle regeneration. Nat Commun 2022; 13:7463. [PMID: 36460667 PMCID: PMC9718784 DOI: 10.1038/s41467-022-35183-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 11/22/2022] [Indexed: 12/05/2022] Open
Abstract
Transplantation of mesenchymal stem cells (MSCs) holds promise to repair severe traumatic injuries. However, current transplantation practices limit the potential of this technique, either by losing the viable MSCs or reducing the performance of resident MSCs. Herein, we design a "bead-jet" printer, specialized for high-throughput intra-operative formulation and printing of MSCs-laden Matrigel beads. We show that high-density encapsulation of MSCs in Matrigel beads is able to augment MSC function, increasing MSC proliferation, migration, and extracellular vesicle production, compared with low-density bead or high-density bulk encapsulation of the equivalent number of MSCs. We find that the high-density MSCs-laden beads in sparse patterns demonstrate significantly improved therapeutic performance, by regenerating skeletal muscles approaching native-like cell density with reduced fibrosis, and regenerating skin with hair follicle growth and increased dermis thickness. MSC proliferation within 1-week post-transplantation and differentiation at 3 - 4 weeks post-transplantation are suggested to contribute therapy augmentation. We expect this "bead-jet" printing system to strengthen the potential of MSC transplantation.
Collapse
Affiliation(s)
- Yuanxiong Cao
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Jiayi Tan
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Haoran Zhao
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Ting Deng
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Yunxia Hu
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Junhong Zeng
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Jiawei Li
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Yifan Cheng
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Jiyuan Tang
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Zhiwei Hu
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Keer Hu
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Bing Xu
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
- Shenzhen Bay Laboratory, 518055, Shenzhen, China
| | - Zitian Wang
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Yaojiong Wu
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Peter E Lobie
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
- Shenzhen Bay Laboratory, 518055, Shenzhen, China
| | - Shaohua Ma
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China.
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China.
- Shenzhen Bay Laboratory, 518055, Shenzhen, China.
- Institute for Brain and Cognitive Sciences, Tsinghua University, 100084, Beijing, China.
| |
Collapse
|
44
|
Li Z, Yue M, Liu X, Liu Y, Lv L, Zhang P, Zhou Y. The PCK2-glycolysis axis assists three-dimensional-stiffness maintaining stem cell osteogenesis. Bioact Mater 2022; 18:492-506. [PMID: 35415308 PMCID: PMC8971594 DOI: 10.1016/j.bioactmat.2022.03.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 03/15/2022] [Accepted: 03/23/2022] [Indexed: 01/02/2023] Open
Abstract
Understanding mechanisms underlying the heterogeneity of multipotent stem cells offers invaluable insights into biogenesis and tissue development. Extracellular matrix (ECM) stiffness has been acknowledged as a crucial factor regulating stem cell fate. However, how cells sense stiffness cues and adapt their metabolism activity is still unknown. Here we report the novel role of mitochondrial phosphoenolpyruvate carboxykinase (PCK2) in enhancing osteogenesis in 3D ECM via glycolysis. We experimentally mimicked the physical characteristics of 3D trabeculae network of normal and osteoporotic bone with different microstructure and stiffness, observing that PCK2 promotes osteogenesis in 3D ECM with tunable stiffness in vitro and in vivo. Mechanistically, PCK2 enhances the rate-limiting metabolic enzyme pallet isoform phosphofructokinase (PFKP) in 3D ECM, and further activates AKT/extracellular signal-regulated kinase 1/2 (ERK1/2) cascades, which directly regulates osteogenic differentiation of MSCs. Collectively, our findings implicate an intricate crosstalk between cell mechanics and metabolism, and provide new perspectives for strategies of osteoporosis. As the key rate-limiting enzyme of gluconeogenesis, PCK2 manipulates osteogenesis in stiff and soft ECM in vitro and in vivo. PCK2 regulates osteogenic capacity of BMMSCs in 3D ECM with different stiffness, via modulating glycolysis and regulating PFKP-AKT/ERK signaling pathways.
Collapse
Affiliation(s)
- Zheng Li
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Muxin Yue
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Xuenan Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Longwei Lv
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Ping Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
- Corresponding author. Vice Professor of Department of Prosthodontics, School and Hospital of Stomatology of Peking University, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China.
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
- Corresponding author. President of School and Hospital of Stomatology of Peking University, Professor of Department of Prosthodontics, Vice-Director for National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Vice-Director for the National Clinical Research Center of Oral Diseases (PKU), 22 Zhongguancun South Avenue, Haidian District, Beijing, 10081, PR China.
| |
Collapse
|
45
|
Zelinka A, Roelofs AJ, Kandel RA, De Bari C. Cellular therapy and tissue engineering for cartilage repair. Osteoarthritis Cartilage 2022; 30:1547-1560. [PMID: 36150678 DOI: 10.1016/j.joca.2022.07.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 02/02/2023]
Abstract
Articular cartilage (AC) has limited capacity for repair. The first attempt to repair cartilage using tissue engineering was reported in 1977. Since then, cell-based interventions have entered clinical practice in orthopaedics, and several tissue engineering approaches to repair cartilage are in the translational pipeline towards clinical application. Classically, these involve a scaffold, substrate or matrix to provide structure, and cells such as chondrocytes or mesenchymal stromal cells to generate the tissue. We discuss the advantages and drawbacks of the use of various cell types, natural and synthetic scaffolds, multiphasic or gradient-based scaffolds, and self-organizing or self-assembling scaffold-free systems, for the engineering of cartilage constructs. Several challenges persist including achieving zonal tissue organization and integration with the surrounding tissue upon implantation. Approaches to improve cartilage thickness, organization and mechanical properties include mechanical stimulation, culture under hypoxic conditions, and stimulation with growth factors or other macromolecules. In addition, advanced technologies such as bioreactors, biosensors and 3D bioprinting are actively being explored. Understanding the underlying mechanisms of action of cell therapy and tissue engineering approaches will help improve and refine therapy development. Finally, we discuss recent studies of the intrinsic cellular and molecular mechanisms of cartilage repair that have identified novel signals and targets and are inspiring the development of molecular therapies to enhance the recruitment and cartilage reparative activity of joint-resident stem and progenitor cells. A one-fits-all solution is unrealistic, and identifying patients who will respond to a specific targeted treatment will be critical.
Collapse
Affiliation(s)
- A Zelinka
- Lunenfeld Tanenbaum Research Institute, Sinai Health, Dept. Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - A J Roelofs
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | - R A Kandel
- Lunenfeld Tanenbaum Research Institute, Sinai Health, Dept. Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
| | - C De Bari
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK.
| |
Collapse
|
46
|
Reed E, Fellows A, Lu R, Rienks M, Schmidt L, Yin X, Duregotti E, Brandt M, Krasemann S, Hartmann K, Barallobre-Barreiro J, Addison O, Cuello F, Hansen A, Mayr M. Extracellular Matrix Profiling and Disease Modelling in Engineered Vascular Smooth Muscle Cell Tissues. Matrix Biol Plus 2022; 16:100122. [PMID: 36193159 PMCID: PMC9526190 DOI: 10.1016/j.mbplus.2022.100122] [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: 04/13/2022] [Revised: 08/22/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022] Open
Abstract
Aortic smooth muscle cells (SMCs) have an intrinsic role in regulating vessel homeostasis and pathological remodelling. In two-dimensional (2D) cell culture formats, however, SMCs are not embedded in their physiological extracellular matrix (ECM) environment. To overcome the limitations of conventional 2D SMC cultures, we established a 3D in vitro model of engineered vascular smooth muscle cell tissues (EVTs). EVTs were casted from primary murine aortic SMCs by suspending a SMC-fibrin master mix between two flexible silicon-posts at day 0 before prolonged culture up to 14 days. Immunohistochemical analysis of EVT longitudinal sections demonstrated that SMCs were aligned, viable and secretory. Mass spectrometry-based proteomics analysis of murine EVT lysates was performed and identified 135 matrisome proteins. Proteoglycans, including the large aggregating proteoglycan versican, accumulated within EVTs by day 7 of culture. This was followed by the deposition of collagens, elastin-binding proteins and matrix regulators up to day 14 of culture. In contrast to 2D SMC controls, accumulation of versican occurred in parallel to an increase in versikine, a cleavage product mediated by proteases of the A Disintegrin and Metalloproteinase with Thrombospondin motifs (ADAMTS) family. Next, we tested the response of EVTs to stimulation with transforming growth factor beta-1 (TGFβ-1). EVTs contracted in response to TGFβ-1 stimulation with altered ECM composition. In contrast, treatment with the pharmacological activin-like kinase inhibitor (ALKi) SB 431542 suppressed ECM secretion. As a disease stimulus, we performed calcification assays. The ECM acts as a nidus for calcium phosphate deposition in the arterial wall. We compared the onset and extent of calcification in EVTs and 2D SMCs cultured under high calcium and phosphate conditions for 7 days. Calcified EVTs displayed increased tissue stiffness by up to 30 % compared to non-calcified controls. Unlike the rapid calcification of SMCs in 2D cultures, EVTs sustained expression of the calcification inhibitor matrix Gla protein and allowed for better discrimination of the calcification propensity between independent biological replicates. In summary, EVTs are an intuitive and versatile model to investigate ECM synthesis and turnover by SMCs in a 3D environment. Unlike conventional 2D cultures, EVTs provide a more relevant pathophysiological model for retention of the nascent ECM produced by SMCs.
Collapse
Key Words
- 2D, Two-dimensional
- 3D cell culture
- 3D, Three-dimensional
- ADAMTS, A disintegrin and metalloproteinase with thrombospondin motifs
- ALKi, Activin-like kinase inhibitor
- Calcification
- ECM
- ECM, Extracellular matrix
- EHT, Engineered heart tissue
- EVT, Engineered vascular smooth muscle cell tissue
- LC-MS/MS, Liquid chromatography with tandem mass spectrometry
- Proteomics
- SMC, Smooth muscle cell
- Smooth muscle cells
- TCP, Tissue culture polystyrene
- TGFβ-1, Transforming growth factor beta-1
- Tissue engineering
Collapse
Affiliation(s)
- Ella Reed
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| | - Adam Fellows
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Ruifang Lu
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| | - Marieke Rienks
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| | - Lukas Schmidt
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| | - Xiaoke Yin
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| | - Elisa Duregotti
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| | - Mona Brandt
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, University Medical Center Hamburg-Eppendorf, Germany
| | - Susanne Krasemann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Kristin Hartmann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Javier Barallobre-Barreiro
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| | - Owen Addison
- Centre of Oral, Clinical & Translational Sciences, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK
| | - Friederike Cuello
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, University Medical Center Hamburg-Eppendorf, Germany
| | - Arne Hansen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, University Medical Center Hamburg-Eppendorf, Germany
| | - Manuel Mayr
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| |
Collapse
|
47
|
Guan S, Wang Y, Xie F, Wang S, Xu W, Xu J, Sun C. Carboxymethyl Chitosan and Gelatin Hydrogel Scaffolds Incorporated with Conductive PEDOT Nanoparticles for Improved Neural Stem Cell Proliferation and Neuronal Differentiation. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238326. [PMID: 36500418 PMCID: PMC9740948 DOI: 10.3390/molecules27238326] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022]
Abstract
Tissue engineering scaffolds provide biological and physiochemical cures to guide tissue recovery, and electrical signals through the electroactive materials possess tremendous potential to modulate the cell fate. In this study, a novel electroactive hydrogel scaffold was fabricated by assembling poly(3,4-ethylenedioxythiophene) (PEDOT) nanoparticles on a carboxymethyl chitosan/gelatin (CMCS/Gel) composite hydrogel surface via in situ chemical polymerization. The chemical structure, morphology, conductivity, porosity, swelling rate, in vitro biodegradation, and mechanical properties of the prepared hydrogel samples were characterized. The adhesion, proliferation, and differentiation of neural stem cells (NSCs) on conductive hydrogels were investigated. The CMCS/Gel-PEDOT hydrogels exhibited high porosity, excellent water absorption, improved thermal stability, and adequate biodegradability. Importantly, the mechanical properties of the prepared hydrogels were similar to those of brain tissue, with electrical conductivity up to (1.52 ± 0.15) × 10-3 S/cm. Compared to the CMCS/Gel hydrogel, the incorporation of PEDOT nanoparticles significantly improved the adhesion of NSCs, and supported long-term cell growth and proliferation in a three-dimensional (3D) microenvironment. In addition, under the differentiation condition, the conductive hydrogel also significantly enhanced neuronal differentiation with the up-regulation of β-tubulin III expression. These results suggest that CMCS/Gel-PEDOT hydrogels may be an attractive conductive substrate for further studies on neural tissue repair and regeneration.
Collapse
Affiliation(s)
- Shui Guan
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
- Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
- Research & Educational Center for the Control Engineering of Translational Precision Medicine (R-ECCE-TPM), School of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian 116024, China
- Correspondence: (S.G.); (J.X.); (C.S.)
| | - Yangbin Wang
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
- Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Feng Xie
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
- Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Shuping Wang
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
- Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Weiping Xu
- School of Ocean Science and Technology & Panjin Institute of Industrial Technology, Dalian University of Technology, Panjin 124221, China
| | - Jianqiang Xu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
- Correspondence: (S.G.); (J.X.); (C.S.)
| | - Changkai Sun
- Research & Educational Center for the Control Engineering of Translational Precision Medicine (R-ECCE-TPM), School of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian 116024, China
- Correspondence: (S.G.); (J.X.); (C.S.)
| |
Collapse
|
48
|
Zhang R, Hu Z, Wang Y, Qiu R, Wang G, Wang L, Hu B. A biomimetic double network hydrogel ameliorates renal fibrosis and promotes renal regeneration. J Mater Chem B 2022; 10:9424-9437. [PMID: 36378134 DOI: 10.1039/d2tb01939f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are serious global public health issues. Both interconnect closely, and AKI-CKD transition significantly increases the morbidity of CKD and inevitably progresses to end stage renal disease. However, with the current drug delivery system it is hard to achieve precise delivery and apply it to clinical practice due to the local fibrotic milieu of the AKI-CKD transition procedure. Consequently, new treatment options to halt or even reverse AKI-CKD transition are urgently needed. Curcumin and Ac-SDKP were proved to be capable of ameliorating renal injury and restoring renal biological function. However, due to the water-insolubility, poor absorption and ease of degradation features, their utilization based on traditional drug delivery systems was still confined to the laboratory. A new approach for the targeted delivery of curcumin and Ac-SDKP into kidneys is needed. Hydrogels, owing to their capability of targeted-drug delivery and bio-favorable nature, emerge as a promising resolution. Herein, we developed a bioinspired double network hydrogel scaffold loaded with curcumin and N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) to explore the feasibility of drug-loaded hydrogels for treatment of AKI-CKD transition. This double network hydrogel (GCS) was prepared based on gelatin and curcumin-zinc with polydopamine (DOPA) coating and then immobilized with Ac-SDKP on the surface. The prepared hydrogels possessed appropriate porosity, suitable mechanical properties, and excellent biocompatibility. In vitro, the GCS hydrogel was demonstrated to be pro-angiogenic, anti-oxidative and anti-fibrotic. In vivo, after the GCS hydrogel was implanted into partially nephrectomized rat kidneys, local renal fibrosis was observed to be improved significantly, and neo-blood vessels and neonatal renal tubules appeared around the implanted area. We speculated that the GCS hydrogel could ameliorate renal fibrosis and injury significantly and stimulate regeneration in situ. Taken together, this study demonstrated the promising potential of this bioinspired hydrogel scaffold for renal injury repair and renal regeneration.
Collapse
Affiliation(s)
- Rui Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Key Lab for Organ Failure Research, Ministry of Education Guangzhou, Guangdong, 510515, China. .,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Zifan Hu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Yongqin Wang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Key Lab for Organ Failure Research, Ministry of Education Guangzhou, Guangdong, 510515, China.
| | - Renjie Qiu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Guobao Wang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Key Lab for Organ Failure Research, Ministry of Education Guangzhou, Guangdong, 510515, China.
| | - Leyu Wang
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Bianxiang Hu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Key Lab for Organ Failure Research, Ministry of Education Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
49
|
Alfayez E, Veschini L, Dettin M, Zamuner A, Gaetani M, Carreca AP, Najman S, Ghanaati S, Coward T, Di Silvio L. DAR 16-II Primes Endothelial Cells for Angiogenesis Improving Bone Ingrowth in 3D-Printed BCP Scaffolds and Regeneration of Critically Sized Bone Defects. Biomolecules 2022; 12:biom12111619. [PMID: 36358970 PMCID: PMC9687468 DOI: 10.3390/biom12111619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/28/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022] Open
Abstract
Bone is a highly vascularized tissue and relies on the angiogenesis and response of cells in the immediate environmental niche at the defect site for regeneration. Hence, the ability to control angiogenesis and cellular responses during osteogenesis has important implications in tissue-engineered strategies. Self-assembling ionic-complementary peptides have received much interest as they mimic the natural extracellular matrix. Three-dimensional (3D)-printed biphasic calcium phosphate (BCP) scaffolds coated with self-assembling DAR 16-II peptide provide a support template with the ability to recruit and enhance the adhesion of cells. In vitro studies demonstrated prompt the adhesion of both human umbilical vein endothelial cells (HUVEC) and human mesenchymal stem cells (hMSC), favoring endothelial cell activation toward an angiogenic phenotype. The SEM-EDS and protein micro bicinchoninic acid (BCA) assays demonstrated the efficacy of the coating. Whole proteomic analysis of DAR 16-II-treated HUVECs demonstrated the upregulation of proteins involved in cell adhesion (HABP2), migration (AMOTL1), cytoskeletal re-arrangement (SHC1, TMOD2), immuno-modulation (AMBP, MIF), and morphogenesis (COL4A1). In vivo studies using DAR-16-II-coated scaffolds provided an architectural template, promoting cell colonization, osteogenesis, and angiogenesis. In conclusion, DAR 16-II acts as a proactive angiogenic factor when adsorbed onto BCP scaffolds and provides a simple and effective functionalization step to facilitate the translation of tailored 3D-printed BCP scaffolds for clinical applications.
Collapse
Affiliation(s)
- Eman Alfayez
- Faculty of Dentistry, Oral Biology Department, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Faculty of Dentistry, Oral & Craniofacial Sciences King’s College London, London SE1 9RT, UK
| | - Lorenzo Veschini
- Faculty of Dentistry, Oral & Craniofacial Sciences King’s College London, London SE1 9RT, UK
| | - Monica Dettin
- Department of Industrial Engineering, University of Padua, 35131 Padua, Italy
| | - Annj Zamuner
- Department of Civil, Environmental, and Architectural Engineering, University of Padua, 35131 Padua, Italy
| | - Massimiliano Gaetani
- Fondazione Ricerca nel Mediterraneo (Ri.MED) and Department of Laboratory Medicine and Advanced Biotechnologies, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione, 90100 Palermo, Italy
- Chemical Proteomics, Department of Medical Biochemistry and Biophysics, Karolinska Institutet and SciLifeLab (Science for Life Laboratory), SE-17 177 Stockholm, Sweden
| | - Anna P. Carreca
- Fondazione Ricerca nel Mediterraneo (Ri.MED) and Department of Laboratory Medicine and Advanced Biotechnologies, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione, 90100 Palermo, Italy
| | - Stevo Najman
- Faculty of Medicine, University of Niš, 18000 Niš, Serbia
| | - Shahram Ghanaati
- Department for Oral, Cranio-Maxillofacial and Facial Plastic Surgery, Medical Center of the Goethe University, 60323 Frankfurt, Germany
| | - Trevor Coward
- Faculty of Dentistry, Oral & Craniofacial Sciences King’s College London, London SE1 9RT, UK
| | - Lucy Di Silvio
- Faculty of Dentistry, Oral & Craniofacial Sciences King’s College London, London SE1 9RT, UK
- Correspondence: ; Tel.: +44-02-07848-8475
| |
Collapse
|
50
|
Van Tran V, Wi E, Shin SY, Lee D, Kim YA, Ma BC, Chang M. Microgels based on 0D-3D carbon materials: Synthetic techniques, properties, applications, and challenges. CHEMOSPHERE 2022; 307:135981. [PMID: 35964721 DOI: 10.1016/j.chemosphere.2022.135981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 07/22/2022] [Accepted: 08/05/2022] [Indexed: 06/15/2023]
Abstract
Microgels are three-dimensional (3D) colloidal hydrogel particles with outstanding features such as biocompatibility, good mechanical properties, tunable sizes from submicrometer to tens of nanometers, and large surface areas. Because of these unique qualities, microgels have been widely used in various applications. Carbon-based materials (CMs) with various dimensions (0-3D) have recently been investigated as promising candidates for the design and fabrication of microgels because of their large surface area, excellent conductivity, unique chemical stability, and low cost. Here, we provide a critical review of the specific characteristics of CMs that are being incorporated into microgels, as well as the state-of-the art applications of CM-microgels in pollutant adsorption and photodegradation, H2 evoluation, CO2 capture, soil conditioners, water retention, drug delivery, cell encapsulation, and tissue engineering. Advanced preparation techniques for CM-microgel systems are also summarized and discussed. Finally, challenges related to the low colloidal stability of CM-microgels and development strategies are examined. This review shows that CM-microgels have the potential to be widely used in various practical applications.
Collapse
Affiliation(s)
- Vinh Van Tran
- Laser and Thermal Engineering Laboratory, Department of Mechanical Engineering, Gachon University, Seongnam, 13120, South Korea
| | - Eunsol Wi
- Department of Polymer Engineering, Graduate School, Chonnam National University, Gwangju, 61186, South Korea
| | - Seo Young Shin
- Department of Polymer Engineering, Graduate School, Chonnam National University, Gwangju, 61186, South Korea
| | - Daeho Lee
- Laser and Thermal Engineering Laboratory, Department of Mechanical Engineering, Gachon University, Seongnam, 13120, South Korea
| | - Yoong Ahm Kim
- Department of Polymer Engineering, Graduate School, Chonnam National University, Gwangju, 61186, South Korea; School of Polymer Science and Engineering, Chonnam National University, Gwangju, 61186, South Korea; Alan G. MacDiarmid Energy Research Institute, Chonnam National University, Gwangju, 61186, South Korea
| | - Byung Chol Ma
- School of Chemical Engineering, Chonnam National University, Gwangju, 61186, South Korea.
| | - Mincheol Chang
- Department of Polymer Engineering, Graduate School, Chonnam National University, Gwangju, 61186, South Korea; School of Polymer Science and Engineering, Chonnam National University, Gwangju, 61186, South Korea; Alan G. MacDiarmid Energy Research Institute, Chonnam National University, Gwangju, 61186, South Korea.
| |
Collapse
|