1
|
Wang C, Fan M, Heo SC, Adams SM, Li T, Liu Y, Li Q, Loebel C, Burdick JA, Lu XL, Birk DE, Alisafaei F, Mauck RL, Han L. Structure, Mechanics, and Mechanobiology of Fibrocartilage Pericellular Matrix Mediated by Type V Collagen. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e14750. [PMID: 40407177 DOI: 10.1002/advs.202414750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 05/01/2025] [Indexed: 05/28/2025]
Abstract
The pericellular matrix (PCM) is the immediate microniche surrounding cells in various tissues, regulating matrix turnover, cell-matrix interactions, and disease. This study elucidates the structure-mechanical properties and mechanobiology of the PCM in fibrocartilage, using the murine meniscus as the model. The fibrocartilage PCM is comprised of thin, randomly oriented collagen fibrils that entrap proteoglycans, contrasting with the densely packed, highly aligned collagen fibers in the bulk extracellular matrix (ECM). Compared to the ECM, the PCM exhibits lower modulus and greater isotropy, but has similar relative viscoelastic properties. In Col5a1+/- menisci, the reduction of collagen V results in thicker, more heterogeneous collagen fibrils, reduced modulus, loss of isotropy and faster viscoelastic relaxation in the PCM. Such altered PCM leads to impaired matrix-to-cell strain transmission, and in turn, disrupts mechanotransduction of meniscal cells, as illustrated by reduced calcium signaling activities and alters expression of matrix genes. In vitro, Col5a1+/- cells produce a weakened PCM with inferior properties and reduced protection of cells against tensile stretch. These findings highlight the PCM as a distinctive microstructure in fibrocartilage mechanobiology, underscoring a pivotal role of collagen V in PCM function. Targeting the PCM or its constituents offers potential for improving meniscus regeneration, osteoarthritis intervention and broader fibrocartilage-related therapies.
Collapse
Affiliation(s)
- Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Mingyue Fan
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Su Chin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sheila M Adams
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Thomas Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Yuchen Liu
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Qing Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Claudia Loebel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jason A Burdick
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, 80309, USA
| | - X Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, DE, 19716, USA
| | - David E Birk
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Farid Alisafaei
- Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Administration Medical Center, Philadelphia, PA, 19104, USA
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| |
Collapse
|
2
|
Garcia‐Aponte OF, Kahlenberg S, Kouroupis D, Egger D, Kasper C. Effects of Hydrogels on Mesenchymal Stem/Stromal Cells Paracrine Activity and Extracellular Vesicles Production. J Extracell Vesicles 2025; 14:e70057. [PMID: 40091440 PMCID: PMC11911545 DOI: 10.1002/jev2.70057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/10/2024] [Accepted: 02/11/2025] [Indexed: 03/19/2025] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are a valuable source of paracrine factors, as they have a remarkable secretory capacity, and there is a sizeable knowledge base to develop industrial and clinical production protocols. Promising cell-free approaches for tissue regeneration and immunomodulation are driving research towards secretome applications, among which extracellular vesicles (EVs) are steadily gaining attention. However, the manufacturing and application of EVs is limited by insufficient yields, knowledge gaps, and low standardization. Facing these limitations, hydrogels represent a versatile three-dimensional (3D) culture platform that can incorporate extracellular matrix (ECM) components to mimic the natural stem cell environment in vitro; via these niche-mimicking properties, hydrogels can regulate MSCs' morphology, adhesion, proliferation, differentiation and secretion capacities. However, the impact of the hydrogel's architectural, biochemical and biomechanical properties on the production of EVs remains poorly understood, as the field is still in its infancy and the interdependency of culture parameters compromises the comparability of the studies. Therefore, this review summarizes and discusses the reported effects of hydrogel encapsulation and culture on the secretion of MSC-EVs. Considering the effects of cell-material interactions on the overall paracrine activity of MSCs, we identify persistent challenges from low standardization and process control, and outline future paths of research, such as the synergic use of hydrogels and bioreactors to enhance MSC-EV generation.
Collapse
Affiliation(s)
- Oscar Fabian Garcia‐Aponte
- Department of Biotechnology and Food Science, Institute of Cell and Tissue Culture TechnologiesUniversity of Natural Resources and Life SciencesViennaAustria
| | - Simon Kahlenberg
- Department of Biotechnology and Food Science, Institute of Cell and Tissue Culture TechnologiesUniversity of Natural Resources and Life SciencesViennaAustria
| | - Dimitrios Kouroupis
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of MedicineUniversity of MiamiMiamiFloridaUSA
- Diabetes Research Institute & Cell Transplant Center, Miller School of MedicineUniversity of MiamiMiamiFloridaUSA
| | - Dominik Egger
- Institute of Cell Biology and BiophysicsLeibniz University HannoverHannoverGermany
| | - Cornelia Kasper
- Department of Biotechnology and Food Science, Institute of Cell and Tissue Culture TechnologiesUniversity of Natural Resources and Life SciencesViennaAustria
| |
Collapse
|
3
|
Ma X, Zhu X, Lv S, Yang C, Wang Z, Liao M, Zhou B, Zhang Y, Sun S, Chen P, Liu Z, Chen H. 3D bioprinting of prefabricated artificial skin with multicomponent hydrogel for skin and hair follicle regeneration. Theranostics 2025; 15:2933-2950. [PMID: 40083946 PMCID: PMC11898285 DOI: 10.7150/thno.104854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/27/2025] [Indexed: 03/16/2025] Open
Abstract
Background: The timely management of large-scale wounds and the regeneration of skin appendages constitute major clinical issues. The production of high-precision and customizable artificial skin via 3D bioprinting offers a feasible means to surmount the predicament, within which the selection of bioactive materials and seed cells is critical. This study is aimed at employing skin stem cells and multicomponent hydrogels to prefabricate artificial skin through 3D bioprinting, which enables the regeneration of skin and its appendages. Methods and Results: We employed gelatin methacrylate (GelMA) and hyaluronic acid methacrylate (HAMA) as bioactive materials, in conjunction with epidermal stem cells (Epi-SCs) and skin-derived precursors (SKPs), to fabricate artificial skin utilizing 3D bioprinting. The photosensitive multicomponent hydrogel, comprising 5% GelMA and 0.5% HAMA, demonstrated excellent printability, suitable solubility and swelling rates, as well as stable mechanical properties. Moreover, this hydrogel exhibited exceptional biocompatibility, effectively facilitating the proliferation of SKPs while maintaining the cellular characteristics of both SKPs and Epi-SCs. The transplantation of this artificial skin into cutaneous wounds in nude mice led to complete wound healing and functional tissue regeneration. The regenerated tissue comprised epidermis, dermis, hair follicles, blood vessels, and sebaceous glands, closely resembling native skin. Remarkably, the artificial skin demonstrated sustained tissue regeneration capacity even after 12 h of in vitro culture, facilitating comprehensive functional skin regeneration. Conclusions: Our research presented a skin repair strategy for prefabricated cell-loaded artificial skin, thereby successfully facilitating the regeneration of the epidermis, dermis, hair follicles, blood vessels, and sebaceous glands within the wound.
Collapse
Affiliation(s)
- Xiaoxiao Ma
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, People's Republic of China
- East China Institute of Digital Medical Engineering, Shangrao, 334000, People's Republic of China
| | - Xiaohui Zhu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, People's Republic of China
- Peptide and Small Molecule Drug RD Platform, Furong laboratory, Hunan Normal University, Changsha, 410081, Hunan, People's Republic of China
| | - Sheng Lv
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, People's Republic of China
| | - Chunyan Yang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, People's Republic of China
| | - Zihao Wang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, People's Republic of China
| | - Meilan Liao
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, People's Republic of China
| | - Bohao Zhou
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, People's Republic of China
| | - Yiming Zhang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, People's Republic of China
| | - Shiyu Sun
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, People's Republic of China
| | - Ping Chen
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, People's Republic of China
| | - Zhonghua Liu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, People's Republic of China
| | - Haiyan Chen
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, People's Republic of China
- East China Institute of Digital Medical Engineering, Shangrao, 334000, People's Republic of China
- Peptide and Small Molecule Drug RD Platform, Furong laboratory, Hunan Normal University, Changsha, 410081, Hunan, People's Republic of China
| |
Collapse
|
4
|
Zhou Y, Guo Y, Zhang M, Quan S, Li J. The role of RAP2 in regulation of cell volume on bone marrow mesenchymal stem cell fate determination. J Mol Histol 2025; 56:79. [PMID: 39903386 DOI: 10.1007/s10735-025-10362-1] [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: 07/24/2024] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
The extracellular matrix guides cell behavior through mechanical properties, which plays a role in determining cell function and can even influence stem cell fate. Compared with adherent culture, the three-dimensional culture environment is closer to the growth conditions in vivo, but is limited by standardization of material properties and observation and measurement methods. Therefore, it is necessary to study the relationship among the three-dimensional morphological characteristics of cells, cytoskeleton, and stem cell differentiation under adherent culture conditions. Here, we control the cell volume by adjusting the cell density, microfilament cytoskeleton tension, and osmotic pressure of the culture environment, and analyze the cell morphological features and differentiation to the osteoblastic and adipogenic lineages. Based on the in vitro and in vivo results, we identify cell volume as the true reflection of the cytoskeleton tension under stress stimuli compared with cell spreading area. By adjusting cell volume, cytoskeletal tension and cell differentiation can be regulated without affecting cell spreading area. Further study shows that the Ras-related small GTPase RAP2 inhibits the activity of mechanical transducers Lamin A/C and YAP1, playing an important role in cell volume regulation of cell differentiation. In summary, our results support the close relationship between cell volume and cytoskeleton tension. The regulatory role of cell volume on cell differentiation is modulated, at least in part, by RAP2-related mechanosensitive pathways. Our insights into how cell volume regulates cell differentiation may build a bridge between two-dimensional and three-dimensional mechanical studies in cell biology.
Collapse
Affiliation(s)
- Yimei Zhou
- State Key Laboratory of Oral Diseases, National Center of Stomatology, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, 14#, 3rd Section, Renmin South Road, Chengdu, 610041, China
| | - Yutong Guo
- Department of Orthodontics, National Center for 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, Peking University School and Hospital of Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, Beijing, 100081, PR China
| | - Mei Zhang
- State Key Laboratory of Oral Diseases, National Center of Stomatology, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, 14#, 3rd Section, Renmin South Road, Chengdu, 610041, China
| | - Shuqi Quan
- State Key Laboratory of Oral Diseases, National Center of Stomatology, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, 14#, 3rd Section, Renmin South Road, Chengdu, 610041, China
| | - Juan Li
- State Key Laboratory of Oral Diseases, National Center of Stomatology, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, 14#, 3rd Section, Renmin South Road, Chengdu, 610041, China.
| |
Collapse
|
5
|
Meteling M, Johnbosco C, Wolfel A, Conceição F, Govindaraj K, Moreira Teixeira L, Leijten J. High-Throughput Single-Cell Analysis of Local Nascent Protein Deposition in 3D Microenvironments via Extracellular Protein Identification Cytometry (EPIC). ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2415981. [PMID: 39629556 PMCID: PMC11817916 DOI: 10.1002/adma.202415981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/17/2024] [Indexed: 02/13/2025]
Abstract
Extracellular matrix (ECM) guides cell behavior and tissue fate. Cell populations are notoriously heterogeneous leading to large variations in cell behavior at the single-cell level. Although insights into population heterogeneity are valuable for fundamental biology, regenerative medicine, and drug testing, current ECM analysis techniques only provide either averaged population-level data or single-cell data from a limited number of cells. Here, extracellular protein identification cytometry (EPIC) is presented as a novel platform technology that enables high-throughput measurements of local nascent protein deposition at single-cell level. Specifically, human primary chondrocytes are microfluidically encapsulated in enzymatically crosslinked microgels of 16 picoliter at kHz rates, forming large libraries of discrete 3D single-cell microniches in which ECM can be deposited. ECM proteins are labeled using fluorescence immunostaining to allow for nondestructive analysis via flow cytometry. This approach reveals population heterogeneity in matrix deposition at unprecedented throughput, allowing for the identification and fluorescent activated cell sorting-mediated isolation of cellular subpopulations. Additionally, it is demonstrated that inclusion of a second cell into microgels allows for studying the effect of cell-cell contact on matrix deposition. In summary, EPIC enables high-throughput single-cell analysis of nascent proteins in 3D microenvironments, which is anticipated to advance fundamental knowledge and tissue engineering applications.
Collapse
Affiliation(s)
- Marieke Meteling
- Leijten LaboratoryDepartment of Developmental BioengineeringFaculty of Science and Technology, Technical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Castro Johnbosco
- Leijten LaboratoryDepartment of Developmental BioengineeringFaculty of Science and Technology, Technical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Alexis Wolfel
- Leijten LaboratoryDepartment of Developmental BioengineeringFaculty of Science and Technology, Technical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Francisco Conceição
- Department of Advanced Organ bioengineering and TherapeuticsFaculty of Science and Technology, Technical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Kannan Govindaraj
- Leijten LaboratoryDepartment of Developmental BioengineeringFaculty of Science and Technology, Technical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Liliana Moreira Teixeira
- Department of Advanced Organ bioengineering and TherapeuticsFaculty of Science and Technology, Technical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Jeroen Leijten
- Leijten LaboratoryDepartment of Developmental BioengineeringFaculty of Science and Technology, Technical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| |
Collapse
|
6
|
Diez-Guardia V, Tian Y, Guo Y, Li J, Cui S, Dreiss CA, Gentleman E, Wang X. Controlled Release of Human Dental Pulp Stem Cell-Derived Exosomes from Hydrogels Attenuates Temporomandibular Joint Osteoarthritis. Adv Healthc Mater 2024:e2402923. [PMID: 39713912 DOI: 10.1002/adhm.202402923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/27/2024] [Indexed: 12/24/2024]
Abstract
Temporomandibular joint osteoarthritis (TMJOA) is a painful inflammatory condition that limits mouth opening. Cell-derived exosomes, which have anti-inflammatory effects, are emerging as a treatment for TMJOA. Injection of dental pulp stem cells (DPSCs), which secrete exosomes, can moderate tissue damage in a rat model of TMJOA. However, injected exosomes are quickly cleared, necessitating repeated injections for therapeutic efficacy. Here, vinyl sulfone-modified hyaluronic acid (HA-VS) hydrogels, suitable for encapsulating exosomes are formulated. HA-VS hydrogels degrade in the presence of hyaluronidase and allow for the release of beads of similar size to exosomes over 3 to 6 days. In a rat model of TMJOA, injection of exosomes or exosomes within HA-VS hydrogels significantly attenuated damage-mediated subchondral bone loss as determined by micro-computed tomography, and reduced inflammatory and tissue damage scores as assessed by histology. Overall, DPSCs-derived exosomes attenuated joint damage, but treatment with exosomes within HA-VS hydrogels shows additional protective effects on subchondral bone maintenance and integrity. These findings confirm the protective effects of DPSCs-derived exosomes in moderating tissue damage in TMJOA and suggest that combining exosomes with HA hydrogels can further promote their therapeutic effects.
Collapse
Affiliation(s)
- Victor Diez-Guardia
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
| | - Yajing Tian
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, 100081, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, 100081, China
- Center of Stomatology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yunzhe Guo
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
| | - Jiaying Li
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
| | - Shengjie Cui
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, 100081, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, 100081, China
| | - Cécile A Dreiss
- Institute of Pharmaceutical Science, King's College London, London, SE1 9NH, UK
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
- Department of Biomedical Sciences, University of Lausanne, Lausanne, 1005, Switzerland
| | - Xuedong Wang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, 100081, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, 100081, China
| |
Collapse
|
7
|
Wang Y, Sun C, Liu Z, Zhang S, Gao K, Yi F, Zhou W, Liu H. Nanoengineered Endocytic Biomaterials for Stem Cell Therapy. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202410714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Indexed: 01/05/2025]
Abstract
AbstractStem cells, ideal for the tissue repair and regeneration, possess extraordinary capabilities of multidirectional differentiation and self‐renewal. However, the limited spontaneous differentiation potential makes it challenging to harness them for tissue repair without external intervention. Although conventional approaches using biomolecules, small organic molecules, and ions have shown specific and effective functions, they face challenges such as in vivo diffusion and degradation, poor internalization, and side effects on adjacent cells. Nanoengineered biomaterials offer a solution by solidifying and nanosizing these soluble regulating molecules and ions, facilitating their uptake by stem cells. Once inside lysosomes, these nanoparticles release their contents in a controlled “molecule or ion storm,” efficiently altering the intracellular biological and chemical microenvironment to tune the differentiation of stem cells. This newly emerged approach for regulating stem cell fate has attracted much attention in recent years. This method has shown promising results and is poised to enhance clinical stem cell therapy. This review provides an overview of the design principles for nanoengineered biomaterials, discusses the categories and characteristics of nanoparticles, summarizes the application of nanoparticles in tissue repair and regeneration, and discusses the direction of nanoparticle‐enhanced stem cell therapy and prospects for its clinical application in regenerative medicine.
Collapse
Affiliation(s)
- Yingxue Wang
- Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Chunhui Sun
- Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Zhaoying Liu
- Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Shengmin Zhang
- Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Ke Gao
- Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Fan Yi
- School of Basic Medical Sciences Shandong University Jinan 250012 P. R. China
| | - Wenjuan Zhou
- School of Basic Medical Sciences Shandong University Jinan 250012 P. R. China
| | - Hong Liu
- Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
- State Key Laboratory of Crystal Materials Shandong University Jinan 250100 P. R. China
| |
Collapse
|
8
|
Li M. Atomic force microscopy as a nanomechanical tool for cancer liquid biopsy. Biochem Biophys Res Commun 2024; 734:150637. [PMID: 39226737 DOI: 10.1016/j.bbrc.2024.150637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/24/2024] [Accepted: 08/30/2024] [Indexed: 09/05/2024]
Abstract
Liquid biopsies have been receiving tremendous attention for their potential to reshape cancer management. Though current studies of cancer liquid biopsy primarily focus on applying biochemical assays to characterize the genetic/molecular profiles of circulating tumor cells (CTCs) and their secondary products shed from tumor sites in bodily fluids, delineating the nanomechanical properties of tumor-associated materials in liquid biopsy specimens yields complementary insights into the biology of tumor dissemination and evolution. Particularly, atomic force microscopy (AFM) has become a standard and versatile toolbox for characterizing the mechanical properties of living biological systems at the micro/nanoscale, and AFM has been increasingly utilized to probe the nanomechanical properties of various tumor-derived analytes in liquid biopsies, including CTCs, tumor-associated cells, circulating tumor DNA (ctDNA) molecules, and extracellular vesicles (EVs), offering additional possibilities for understanding cancer pathogenesis from the perspective of mechanobiology. Herein, the applications of AFM in cancer liquid biopsy are summarized, and the challenges and future directions of AFM as a nanomechanical analysis tool in cancer liquid biopsy towards clinical utility are discussed and envisioned.
Collapse
Affiliation(s)
- Mi Li
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China.
| |
Collapse
|
9
|
Hu X, Bao M. Advances in micropatterning technology for mechanotransduction research. MECHANOBIOLOGY IN MEDICINE 2024; 2:100066. [PMID: 40395493 PMCID: PMC12082312 DOI: 10.1016/j.mbm.2024.100066] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/19/2024] [Accepted: 03/24/2024] [Indexed: 05/22/2025]
Abstract
Micropatterning is a sophisticated technique that precisely manipulates the spatial distribution of cell adhesion proteins on various substrates across multiple scales. This precise control over adhesive regions facilitates the manipulation of architectures and physical constraints for single or multiple cells. Furthermore, it allows for an in-depth analysis of how chemical and physical properties influence cellular functionality. In this comprehensive review, we explore the current understanding of the impact of geometrical confinement on cellular functions across various dimensions, emphasizing the benefits of micropatterning in addressing fundamental biological queries. We advocate that utilizing directed self-organization via physical confinement and morphogen gradients on micropatterned surfaces represents an innovative approach to generating functional tissue and controlling morphogenesis in vitro. Integrating this technique with cutting-edge technologies, micropatterning presents a significant potential to bridge a crucial knowledge gap in understanding core biological processes.
Collapse
Affiliation(s)
- Xinyu Hu
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen, the Netherlands
| | - Min Bao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325001, Zhejiang, China
| |
Collapse
|
10
|
Rozans S, Moghaddam AS, Wu Y, Atanasoff K, Nino L, Dunne K, Pashuck ET. Quantifying and Controlling the Proteolytic Degradation of Cell Adhesion Peptides. ACS Biomater Sci Eng 2024; 10:4916-4926. [PMID: 38968389 PMCID: PMC11322908 DOI: 10.1021/acsbiomaterials.4c00736] [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: 04/19/2024] [Revised: 06/10/2024] [Accepted: 06/18/2024] [Indexed: 07/07/2024]
Abstract
Peptides are widely used within biomaterials to improve cell adhesion, incorporate bioactive ligands, and enable cell-mediated degradation of the matrix. While many of the peptides incorporated into biomaterials are intended to be present throughout the life of the material, their stability is not typically quantified during culture. In this work, we designed a series of peptide libraries containing four different N-terminal peptide functionalizations and three C-terminal functionalizations to better understand how simple modifications can be used to reduce the nonspecific degradation of peptides. We tested these libraries with three cell types commonly used in biomaterials research, including mesenchymal stem/stromal cells (hMSCs), endothelial cells, and macrophages, and quantified how these cell types nonspecifically degraded peptides as a function of terminal amino acid and chemistry. We found that peptides in solution which contained N-terminal amines were almost entirely degraded by 48 h, irrespective of the terminal amino acid, and that degradation occurred even at high peptide concentrations. Peptides with C-terminal carboxylic acids also had significant degradation when cultured with the cells. We found that simple modifications to the termini could significantly reduce or completely abolish nonspecific degradation when soluble peptides were added to cells cultured on tissue culture plastic or within hydrogel matrices, and that functionalizations which mimicked peptide conjugations to hydrogel matrices significantly slowed nonspecific degradation. We also found that there were minimal differences in peptide degradation across cell donors and that sequences mimicking different peptides commonly used to functionalize biomaterials all had significant nonspecific degradation. Finally, we saw that there was a positive trend between RGD stability and hMSC spreading within hydrogels, indicating that improving the stability of peptides within biomaterial matrices may improve the performance of engineered matrices.
Collapse
Affiliation(s)
- Samuel
J. Rozans
- Department of Bioengineering, Lehigh University, 7
Asa Drive, Suite 205, Bethlehem, PA 18015, United States
| | - Abolfazl Salehi Moghaddam
- Department of Bioengineering, Lehigh University, 7
Asa Drive, Suite 205, Bethlehem, PA 18015, United States
| | - Yingjie Wu
- Department of Bioengineering, Lehigh University, 7
Asa Drive, Suite 205, Bethlehem, PA 18015, United States
| | - Kayleigh Atanasoff
- Department of Bioengineering, Lehigh University, 7
Asa Drive, Suite 205, Bethlehem, PA 18015, United States
| | - Liliana Nino
- Department of Bioengineering, Lehigh University, 7
Asa Drive, Suite 205, Bethlehem, PA 18015, United States
| | - Katelyn Dunne
- Department of Bioengineering, Lehigh University, 7
Asa Drive, Suite 205, Bethlehem, PA 18015, United States
| | - E. Thomas Pashuck
- Department of Bioengineering, Lehigh University, 7
Asa Drive, Suite 205, Bethlehem, PA 18015, United States
| |
Collapse
|
11
|
Lei H, Sun J, Dai Z, Wo K, Zhang J, Wang Y, Zhao B, Fan W, Wang J, Shi Y, Yang C, Su B, Luo Z, Wu J, Chen L, Chu Y. Remote coupling of electrical and mechanical cues by diurnal photothermal irradiation synergistically promotes bone regeneration. J Nanobiotechnology 2024; 22:410. [PMID: 38992774 PMCID: PMC11238389 DOI: 10.1186/s12951-024-02671-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 06/25/2024] [Indexed: 07/13/2024] Open
Abstract
Recapitulating the natural extracellular physical microenvironment has emerged as a promising method for tissue regeneration, as multiple physical interventions, including ultrasound, thermal and electrical therapy, have shown great potential. However, simultaneous coupling of multiple physical cues to highly bio-mimick natural characteristics for improved tissue regeneration still remains formidable. Coupling of intrinsic electrical and mechanical cues has been regarded as an effective way to modulate tissue repair. Nevertheless, precise and convenient manipulation on coupling of mechano-electrical signals within extracellular environment to facilitate tissue regeneration remains challengeable. Herein, a photothermal-sensitive piezoelectric membrane was designed for simultaneous integration of electrical and mechanical signals in response to NIR irradiation. The high-performance mechano-electrical coupling under NIR exposure synergistically triggered the promotion of osteogenic differentiation of stem cells and enhances bone defect regeneration by increasing cellular mechanical sensing, attachment, spreading and cytoskeleton remodeling. This study highlights the coupling of mechanical signals and electrical cues for modulation of osteogenesis, and sheds light on alternative bone tissue engineering therapies with multiple integrated physical cues for tissue repair.
Collapse
Affiliation(s)
- Haoqi Lei
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Jiwei Sun
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Zhiyin Dai
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Keqi Wo
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Junyuan Zhang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Yifan Wang
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Baoying Zhao
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Wenjie Fan
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Jinyu Wang
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Yunsong Shi
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Cheng Yang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Bin Su
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zhiqiang Luo
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Junjie Wu
- Department of Orthodontics, School of Stomatology, Air Force Medical University, Xi'An, 710032, China.
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China.
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| | - Yingying Chu
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China.
| |
Collapse
|
12
|
Rajeev A, Kansara K, Bhatia D. Navigating the challenges and exploring the perspectives associated with emerging novel biomaterials. Biomater Sci 2024; 12:3565-3581. [PMID: 38832912 DOI: 10.1039/d4bm00376d] [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: 06/06/2024]
Abstract
The field of biomaterials is a continuously evolving interdisciplinary field encompassing biological sciences, materials sciences, chemical sciences, and physical sciences with a multitude of applications realized every year. However, different biomaterials developed for different applications have unique challenges in the form of biological barriers, and addressing these challenges simultaneously is also a challenge. Nevertheless, immense progress has been made through the development of novel materials with minimal adverse effects such as DNA nanostructures, specific synthesis strategies based on supramolecular chemistry, and modulating the shortcomings of existing biomaterials through effective functionalization techniques. This review discusses all these aspects of biomaterials, including the challenges at each level of their development and application, proposed countermeasures for these challenges, and some future directions that may have potential benefits.
Collapse
Affiliation(s)
- Ashwin Rajeev
- Department of Biosciences and Bioengineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat - 382355, India.
| | - Krupa Kansara
- Department of Biosciences and Bioengineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat - 382355, India.
| | - Dhiraj Bhatia
- Department of Biosciences and Bioengineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat - 382355, India.
| |
Collapse
|
13
|
Wang C, Fan M, Heo SJ, Adams SM, Li T, Liu Y, Li Q, Loebel C, Alisafaei F, Burdick JA, Lu XL, Birk DE, Mauck RL, Han L. Structure-Mechanics Principles and Mechanobiology of Fibrocartilage Pericellular Matrix: A Pivotal Role of Type V Collagen. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600498. [PMID: 38979323 PMCID: PMC11230444 DOI: 10.1101/2024.06.26.600498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The pericellular matrix (PCM) is the immediate microniche surrounding resident cells in various tissue types, regulating matrix turnover, cell-matrix cross-talk and disease initiation. This study elucidated the structure-mechanical properties and mechanobiological functions of the PCM in fibrocartilage, a family of connective tissues that sustain complex tensile and compressive loads in vivo. Studying the murine meniscus as the model tissue, we showed that fibrocartilage PCM contains thinner, random collagen fibrillar networks that entrap proteoglycans, a structure distinct from the densely packed, highly aligned collagen fibers in the bulk extracellular matrix (ECM). In comparison to the ECM, the PCM has a lower modulus and greater isotropy, but similar relative viscoelastic properties. In Col5a1 +/- menisci, the reduction of collagen V, a minor collagen localized in the PCM, resulted in aberrant fibril thickening with increased heterogeneity. Consequently, the PCM exhibited a reduced modulus, loss of isotropy and faster viscoelastic relaxation. This disrupted PCM contributes to perturbed mechanotransduction of resident meniscal cells, as illustrated by reduced intracellular calcium signaling, as well as upregulated biosynthesis of lysyl oxidase and tenascin C. When cultured in vitro, Col5a1 +/- meniscal cells synthesized a weakened nascent PCM, which had inferior properties towards protecting resident cells against applied tensile stretch. These findings underscore the PCM as a distinctive microstructure that governs fibrocartilage mechanobiology, and highlight the pivotal role of collagen V in PCM function. Targeting the PCM or its molecular constituents holds promise for enhancing not only meniscus regeneration and osteoarthritis intervention, but also addressing diseases across various fibrocartilaginous tissues.
Collapse
Affiliation(s)
- Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Mingyue Fan
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Su-Jin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Sheila M. Adams
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Thomas Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Yuchen Liu
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Qing Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Claudia Loebel
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Farid Alisafaei
- Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ 07102, United States
| | - Jason A. Burdick
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, United States
| | - X. Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - David E. Birk
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Robert L. Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Administration Medical Center, Philadelphia, PA 19104, United States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| |
Collapse
|
14
|
Duran P, Yang BA, Plaster E, Eiken M, Loebel C, Aguilar CA. Tracking of Nascent Matrix Deposition during Muscle Stem Cell Activation across Lifespan Using Engineered Hydrogels. Adv Biol (Weinh) 2024; 8:e2400091. [PMID: 38616175 DOI: 10.1002/adbi.202400091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/22/2024] [Indexed: 04/16/2024]
Abstract
Adult stem cells occupy a niche that contributes to their function, but how stem cells rebuild their microenvironment after injury remains an open-ended question. Herein, biomaterial-based systems and metabolic labeling are utilized to evaluate how skeletal muscle stem cells deposit extracellular matrix. Muscle stem cells and committed myoblasts are observed to generate less nascent matrix than muscle resident fibro-adipogenic progenitors. When cultured on substrates that matched the stiffness of physiological uninjured and injured muscles, muscle stem cells increased nascent matrix deposition with activation kinetics. Reducing the ability to deposit nascent matrix by an inhibitor of vesicle trafficking (Exo-1) attenuated muscle stem cell function and mimicked impairments observed from muscle stem cells isolated from old muscles. Old muscle stem cells are observed to deposit less nascent matrix than young muscle stem cells, which is rescued with therapeutic supplementation of insulin-like growth factors. These results highlight the role of nascent matrix production with muscle stem cell activation.
Collapse
Affiliation(s)
- Pamela Duran
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Benjamin A Yang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Eleanor Plaster
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Madeline Eiken
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Claudia Loebel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Materials Science & Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Carlos A Aguilar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
15
|
Hartmann B, Fleischhauer L, Nicolau M, Jensen THL, Taran FA, Clausen-Schaumann H, Reuten R. Profiling native pulmonary basement membrane stiffness using atomic force microscopy. Nat Protoc 2024; 19:1498-1528. [PMID: 38429517 DOI: 10.1038/s41596-024-00955-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 11/27/2023] [Indexed: 03/03/2024]
Abstract
Mammalian cells sense and react to the mechanics of their immediate microenvironment. Therefore, the characterization of the biomechanical properties of tissues with high spatial resolution provides valuable insights into a broad variety of developmental, homeostatic and pathological processes within living organisms. The biomechanical properties of the basement membrane (BM), an extracellular matrix (ECM) substructure measuring only ∼100-400 nm across, are, among other things, pivotal to tumor progression and metastasis formation. Although the precise assignment of the Young's modulus E of such a thin ECM substructure especially in between two cell layers is still challenging, biomechanical data of the BM can provide information of eminent diagnostic potential. Here we present a detailed protocol to quantify the elastic modulus of the BM in murine and human lung tissue, which is one of the major organs prone to metastasis. This protocol describes a streamlined workflow to determine the Young's modulus E of the BM between the endothelial and epithelial cell layers shaping the alveolar wall in lung tissues using atomic force microscopy (AFM). Our step-by-step protocol provides instructions for murine and human lung tissue extraction, inflation of these tissues with cryogenic cutting medium, freezing and cryosectioning of the tissue samples, and AFM force-map recording. In addition, it guides the reader through a semi-automatic data analysis procedure to identify the pulmonary BM and extract its Young's modulus E using an in-house tailored user-friendly AFM data analysis software, the Center for Applied Tissue Engineering and Regenerative Medicine processing toolbox, which enables automatic loading of the recorded force maps, conversion of the force versus piezo-extension curves to force versus indentation curves, calculation of Young's moduli and generation of Young's modulus maps, where the pulmonary BM can be identified using a semi-automatic spatial filtering tool. The entire protocol takes 1-2 d.
Collapse
Affiliation(s)
- Bastian Hartmann
- Munich University of Applied Sciences, Center for Applied Tissue Engineering and Regenerative Medicine - CANTER, Munich, Germany
- Center for Nanoscience, Munich, Germany
| | - Lutz Fleischhauer
- Munich University of Applied Sciences, Center for Applied Tissue Engineering and Regenerative Medicine - CANTER, Munich, Germany
- Center for Nanoscience, Munich, Germany
| | - Monica Nicolau
- Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Department of Obstetrics and Gynecology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Thomas Hartvig Lindkær Jensen
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Pathology, Rigshospitalet, Copenhagen, Denmark
| | - Florin-Andrei Taran
- Department of Obstetrics and Gynecology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Hauke Clausen-Schaumann
- Munich University of Applied Sciences, Center for Applied Tissue Engineering and Regenerative Medicine - CANTER, Munich, Germany.
- Center for Nanoscience, Munich, Germany.
| | - Raphael Reuten
- Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg, Freiburg, Germany.
- Department of Obstetrics and Gynecology, Medical Center, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
16
|
Yan Z, Kavanagh T, Harrabi RDS, Lust ST, Tang C, Beavil R, Müller MM, Beavil A, Ameer-Beg S, da Silva RM, Gentleman E. FRET Sensor-Modified Synthetic Hydrogels for Real-Time Monitoring of Cell-Derived Matrix Metalloproteinase Activity using Fluorescence Lifetime Imaging. ADVANCED FUNCTIONAL MATERIALS 2024; 34:adfm.202309711. [PMID: 38779415 PMCID: PMC7615971 DOI: 10.1002/adfm.202309711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Indexed: 05/25/2024]
Abstract
Matrix remodeling plays central roles in a range of physiological and pathological processes and is driven predominantly by the activity of matrix metalloproteinases (MMPs), which degrade extracellular matrix (ECM) proteins. Our understanding of how MMPs regulate cell and tissue dynamics is often incomplete as in vivo approaches are lacking and many in vitro strategies cannot provide high-resolution, quantitative measures of enzyme activity in situ within tissue-like 3D microenvironments. Here, we incorporate a Förster resonance energy transfer (FRET) sensor of MMP activity into fully synthetic hydrogels that mimic many properties of the native ECM. We then use fluorescence lifetime imaging to provide a real-time, fluorophore concentration-independent quantification of MMP activity, establishing a highly accurate, readily adaptable platform for studying MMP dynamics in situ. MCF7 human breast cancer cells encapsulated within hydrogels highlight the detection of MMP activity both locally, at the sub-micron level, and within the bulk hydrogel. Our versatile platform may find use in a range of biological studies to explore questions in the dynamics of cancer metastasis, development, and tissue repair by providing high-resolution, quantitative and in situ readouts of local MMP activity within native tissue-like environments.
Collapse
Affiliation(s)
- Ziqian Yan
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, UK
| | - Thomas Kavanagh
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer and Pharmaceutical Sciences, King’s College London, London, UK
| | | | - Suzette T. Lust
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, UK
| | - Chunling Tang
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, UK
| | - Rebecca Beavil
- Randall Centre of Cell and Molecular Biophysics, King’s College London, London, UK
| | | | - Andrew Beavil
- Randall Centre of Cell and Molecular Biophysics, King’s College London, London, UK
| | - Simon Ameer-Beg
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer and Pharmaceutical Sciences, King’s College London, London, UK
| | | | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, UK
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
17
|
Rozans SJ, Moghaddam AS, Wu Y, Atanasoff K, Nino L, Dunne K, Pashuck ET. Quantifying and controlling the proteolytic degradation of cell adhesion peptides. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.19.590329. [PMID: 38712239 PMCID: PMC11071418 DOI: 10.1101/2024.04.19.590329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Peptides are widely used within biomaterials to improve cell adhesion, incorporate bioactive ligands, and enable cell-mediated degradation of the matrix. While many of the peptides incorporated into biomaterials are intended to be present throughout the life of the material, their stability is not typically quantified during culture. In this work we designed a series of peptide libraries containing four different N-terminal peptide functionalizations and three C-terminal functionalization to better understand how simple modifications can be used to reduce non-specific degradation of peptides. We tested these libraries with three cell types commonly used in biomaterials research, including mesenchymal stem/stromal cells (hMSCs), endothelial cells, and macrophages, and quantified how these cell types non-specifically degraded peptide as a function of terminal amino acid and chemistry. We found that peptides in solution which contained N-terminal amines were almost entirely degraded by 48 hours, irrespective of the terminal amino acid, and that degradation occurred even at high peptide concentrations. Peptides with C-terminal carboxylic acids also had significant degradation when cultured with cells. We found that simple modifications to the termini could significantly reduce or completely abolish non-specific degradation when soluble peptides were added to cells cultured on tissue culture plastic or within hydrogel matrices, and that functionalizations which mimicked peptide conjugations to hydrogel matrices significantly slowed non-specific degradation. We also found that there were minimal differences across cell donors, and that sequences mimicking different peptides commonly-used to functionalized biomaterials all had significant non-specific degradation. Finally, we saw that there was a positive trend between RGD stability and hMSC spreading within hydrogels, indicating that improving the stability of peptides within biomaterial matrices may improve the performance of engineered matrices.
Collapse
|
18
|
Eiken MK, Childs CJ, Brastrom LK, Frum T, Plaster EM, Shachaf O, Pfeiffer S, Levine JE, Alysandratos KD, Kotton DN, Spence JR, Loebel C. Nascent matrix deposition supports alveolar organoid formation from aggregates in synthetic hydrogels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.19.585720. [PMID: 38562781 PMCID: PMC10983987 DOI: 10.1101/2024.03.19.585720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Human induced pluripotent stem cell (iPSC) derived alveolar organoids have emerged as a system to model the alveolar epithelium in homeostasis and disease. However, alveolar organoids are typically grown in Matrigel, a mouse-sarcoma derived basement membrane matrix that offers poor control over matrix properties, prompting the development of synthetic hydrogels as a Matrigel alternative. Here, we develop a two-step culture method that involves pre-aggregation of organoids in hydrogel-based microwells followed by embedding in a synthetic hydrogel that supports alveolar organoid growth, while also offering considerable control over organoid and hydrogel properties. We find that the aggregated organoids secrete their own nascent extracellular matrix (ECM) both in the microwells and upon embedding in the synthetic hydrogels. Thus, the synthetic gels described here allow us to de-couple exogenous and nascent ECM in order to interrogate the role of ECM in organoid formation.
Collapse
Affiliation(s)
- Madeline K. Eiken
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, USA
| | - Charlie J. Childs
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lindy K. Brastrom
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Tristan Frum
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Eleanor M. Plaster
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, USA
| | - Orren Shachaf
- Department of Biomedical Engineering, University of Texas, Austin, TX, USA
| | - Suzanne Pfeiffer
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, USA
| | - Justin E. Levine
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, USA
| | - Konstantinos-Dionysios Alysandratos
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA
- The Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Darrell N. Kotton
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA
- The Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Jason R. Spence
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Program in Cell and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Claudia Loebel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, USA
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
19
|
Ferreira SA, Tallia F, Heyraud A, Walker SA, Salzlechner C, Jones JR, Rankin SM. 3D printed hybrid scaffolds do not induce adverse inflammation in mice and direct human BM-MSC chondrogenesis in vitro. BIOMATERIALS AND BIOSYSTEMS 2024; 13:100087. [PMID: 38312434 PMCID: PMC10835132 DOI: 10.1016/j.bbiosy.2024.100087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/26/2023] [Accepted: 01/08/2024] [Indexed: 02/06/2024] Open
Abstract
Biomaterials that can improve the healing of articular cartilage lesions are needed. To address this unmet need, we developed novel 3D printed silica/poly(tetrahydrofuran)/poly(ε-caprolactone) (SiO2/PTHF/PCL-diCOOH) hybrid scaffolds. Our aim was to carry out essential studies to advance this medical device towards functional validation in pre-clinical trials. First, we show that the chemical composition, microarchitecture and mechanical properties of these scaffolds were not affected by sterilisation with gamma irradiation. To evaluate the systemic and local immunogenic reactivity of the sterilised 3D printed hybrid scaffolds, they were implanted subcutaneously into Balb/c mice. The scaffolds did not trigger a systemic inflammatory response over one week of implantation. The interaction between the host immune system and the implanted scaffold elicited a local physiological reaction with infiltration of mononuclear cells without any signs of a chronic inflammatory response. Then, we investigated how these 3D printed hybrid scaffolds direct chondrogenesis in vitro. Human bone marrow-derived mesenchymal stem/stromal cells (hBM-MSCs) seeded within the 3D printed hybrid scaffolds were cultured under normoxic or hypoxic conditions, with or without chondrogenic supplements. Chondrogenic differentiation assessed by both gene expression and protein production analyses showed that 3D printed hybrid scaffolds support hBM-MSC chondrogenesis. Articular cartilage-specific extracellular matrix deposition within these scaffolds was enhanced under hypoxic conditions (1.7 or 3.7 fold increase in the median of aggrecan production in basal or chondrogenic differentiation media). Our findings show that 3D printed SiO2/PTHF/PCL-diCOOH hybrid scaffolds have the potential to support the regeneration of cartilage tissue.
Collapse
Affiliation(s)
| | | | - Agathe Heyraud
- Department of Materials, Imperial College London, London, UK
| | - Simone A. Walker
- National Heart & Lung Institute, Imperial College London, London, UK
| | | | - Julian R. Jones
- Department of Materials, Imperial College London, London, UK
| | - Sara M. Rankin
- National Heart & Lung Institute, Imperial College London, London, UK
| |
Collapse
|
20
|
Krattiger LA, Moser LO, Odabasi R, Odriozola A, Simona BR, Djonov V, Tibbitt MW, Ehrbar M. Recovery of Therapeutically Ablated Engineered Blood-Vessel Networks on a Plug-and-Play Platform. Adv Healthc Mater 2024; 13:e2301142. [PMID: 37946678 DOI: 10.1002/adhm.202301142] [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: 06/20/2023] [Revised: 10/13/2023] [Indexed: 11/12/2023]
Abstract
Limiting the availability of key angiogenesis-promoting factors is a successful strategy to ablate tumor-supplying blood vessels or to reduce excessive vasculature in diabetic retinopathy. However, the efficacy of such anti-angiogenic therapies (AATs) varies with tumor type, and regrowth of vessels is observed upon termination of treatment. The ability to understand and develop AATs remains limited by a lack of robust in vitro systems for modeling the recovery of vascular networks. Here, complex 3D micro-capillary networks are engineered by sequentially seeding human bone marrow-derived mesenchymal stromal cells and human umbilical vein endothelial cells (ECs) on a previously established, synthetic plug-and-play hydrogel platform. In the tightly interconnected vascular networks that form this way, the two cell types share a basement membrane-like layer and can be maintained for several days of co-culture. Pre-formed networks degrade in the presence of bevacizumab. Upon treatment termination, vessel structures grow back to their original positions after replenishment with new ECs, which also integrate into unperturbed established networks. The data suggest that this plug-and-play platform enables the screening of drugs with blood-vessel inhibiting functions. It is believed that this platform could be of particular interest in studying resistance or recovery mechanisms to AAT treatment.
Collapse
Affiliation(s)
- Lisa A Krattiger
- Department of Obstetrics, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, Zurich, 8091, Switzerland
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, Zürich, 8092, Switzerland
| | - Lukas O Moser
- Department of Obstetrics, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, Zurich, 8091, Switzerland
| | - Rodi Odabasi
- Department of Obstetrics, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, Zurich, 8091, Switzerland
| | - Adolfo Odriozola
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, Bern, 3012, Switzerland
| | - Benjamin R Simona
- Ectica Technologies AG, Raeffelstrasse 24, Zurich, 8045, Switzerland
| | - Valentin Djonov
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, Bern, 3012, Switzerland
| | - Mark W Tibbitt
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, Zürich, 8092, Switzerland
| | - Martin Ehrbar
- Department of Obstetrics, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, Zurich, 8091, Switzerland
| |
Collapse
|
21
|
Cha J, Ding EA, Carvalho EM, Fowler A, Aghi MK, Kumar S. Glioma Cells Secrete Collagen VI to Facilitate Invasion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.12.571198. [PMID: 38168332 PMCID: PMC10760023 DOI: 10.1101/2023.12.12.571198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
While glioblastoma (GBM) progression is associated with extensive extracellular matrix (ECM) secretion, the causal contributions of ECM secretion to invasion remain unclear. Here we investigate these contributions by combining engineered materials, proteomics, analysis of patient data, and a model of bevacizumab-resistant GBM. We find that GBM cells cultured in engineered 3D hyaluronic acid hydrogels secrete ECM prior to invasion, particularly in the absence of exogenous ECM ligands. Proteomic measurements reveal extensive secretion of collagen VI, and collagen VI-associated transcripts are correspondingly enriched in microvascular proliferation regions of human GBMs. We further show that bevacizumab-resistant GBM cells deposit more collagen VI than their responsive counterparts, which is associated with marked cell-ECM stiffening. COL6A3 deletion in GBM cells reduces invasion, β-catenin signaling, and expression of mesenchymal markers, and these effects are amplified in hypoxia. Our studies strongly implicate GBM cell-derived collagen VI in microenvironmental remodeling to facilitate invasion.
Collapse
Affiliation(s)
- Junghwa Cha
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA
| | - Erika A Ding
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
| | - Emily M Carvalho
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
| | - Annabelle Fowler
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA
| | - Manish K Aghi
- Department of Neurosurgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
- Department of Bioengineering and Therapeutic Sciences University of California San Francisco, CA 94158, USA
| |
Collapse
|
22
|
Xu J, Zhang Z, Ren X, Zhang Y, Zhou Y, Lan X, Guo L. In situ photo-crosslinked hydrogel promotes oral mucosal wound healing through sustained delivery of ginsenoside Rg1. Front Bioeng Biotechnol 2023; 11:1252574. [PMID: 37840668 PMCID: PMC10569426 DOI: 10.3389/fbioe.2023.1252574] [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: 07/04/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
Oral mucosal wounds exhibit an increased susceptibility to inflammation as a consequence of their direct exposure to a diverse range of microorganisms. This causes pain, slow healing, and other complications that interfere with patients' daily activities like eating and speaking. Consequently, patients experience a significant decline in their overall quality of life. Therefore, the pursuit of novel treatment approaches is of great importance. In this study, ginsenoside Rg1, a natural active substance extracted from ginseng root, was chosen as a therapeutic agent. It was encapsulated in a screened photo-crosslinked hydrogel scaffold for the treatment of mucosal defects in the rat palate. The results demonstrated that Rg1-hydrogel possessed excellent physical and chemical properties, and that oral mucosa wounds treated with Rg1-hydrogel exhibited the greatest healing performance, as evidenced by more pronounced wound re-epithelialization, increased collagen deposition, and decreased inflammatory infiltration. Subsequent investigations in molecular biology confirmed that Rg1-hydrogel stimulated the secretion of repair-related factors and inhibited the secretion of inflammatory factors. This study demonstrated that the hydrogel containing ginsenoside Rg1 significantly promotes oral mucosal tissue healing in vivo. Based on the findings, it can be inferred that the Rg1-hydrogel has promising prospects for the therapeutic management of oral mucosal wounds.
Collapse
Affiliation(s)
- Jie Xu
- Department of Oral Prosthodontics, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
- Institute of Stomatology, Southwest Medical University, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
- Oral and Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, Luzhou, China
| | - Zhenghao Zhang
- Department of Oral Prosthodontics, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
- Institute of Stomatology, Southwest Medical University, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
- Oral and Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, Luzhou, China
| | - Xiaofeng Ren
- Department of Oral Prosthodontics, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
- Institute of Stomatology, Southwest Medical University, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
- Oral and Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, Luzhou, China
| | - Yunan Zhang
- Department of Oral Prosthodontics, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
- Oral and Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, Luzhou, China
| | - Yang Zhou
- Department of Oral Prosthodontics, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
- Oral and Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, Luzhou, China
| | - Xiaorong Lan
- Institute of Stomatology, Southwest Medical University, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
- Oral and Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, Luzhou, China
| | - Ling Guo
- Department of Oral Prosthodontics, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
- Institute of Stomatology, Southwest Medical University, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
- Oral and Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, Luzhou, China
| |
Collapse
|
23
|
Zhu Y, Zhang M, Sun Q, Wang X, Li X, Li Q. Advanced Mechanical Testing Technologies at the Cellular Level: The Mechanisms and Application in Tissue Engineering. Polymers (Basel) 2023; 15:3255. [PMID: 37571149 PMCID: PMC10422338 DOI: 10.3390/polym15153255] [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: 07/11/2023] [Revised: 07/24/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Mechanics, as a key physical factor which affects cell function and tissue regeneration, is attracting the attention of researchers in the fields of biomaterials, biomechanics, and tissue engineering. The macroscopic mechanical properties of tissue engineering scaffolds have been studied and optimized based on different applications. However, the mechanical properties of the overall scaffold materials are not enough to reveal the mechanical mechanism of the cell-matrix interaction. Hence, the mechanical detection of cell mechanics and cellular-scale microenvironments has become crucial for unraveling the mechanisms which underly cell activities and which are affected by physical factors. This review mainly focuses on the advanced technologies and applications of cell-scale mechanical detection. It summarizes the techniques used in micromechanical performance analysis, including atomic force microscope (AFM), optical tweezer (OT), magnetic tweezer (MT), and traction force microscope (TFM), and analyzes their testing mechanisms. In addition, the application of mechanical testing techniques to cell mechanics and tissue engineering scaffolds, such as hydrogels and porous scaffolds, is summarized and discussed. Finally, it highlights the challenges and prospects of this field. This review is believed to provide valuable insights into micromechanics in tissue engineering.
Collapse
Affiliation(s)
- Yingxuan Zhu
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Mengqi Zhang
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Qingqing Sun
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaofeng Wang
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaomeng Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Qian Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
24
|
Saraswathibhatla A, Indana D, Chaudhuri O. Cell-extracellular matrix mechanotransduction in 3D. Nat Rev Mol Cell Biol 2023; 24:495-516. [PMID: 36849594 PMCID: PMC10656994 DOI: 10.1038/s41580-023-00583-1] [Citation(s) in RCA: 265] [Impact Index Per Article: 132.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2023] [Indexed: 03/01/2023]
Abstract
Mechanical properties of extracellular matrices (ECMs) regulate essential cell behaviours, including differentiation, migration and proliferation, through mechanotransduction. Studies of cell-ECM mechanotransduction have largely focused on cells cultured in 2D, on top of elastic substrates with a range of stiffnesses. However, cells often interact with ECMs in vivo in a 3D context, and cell-ECM interactions and mechanisms of mechanotransduction in 3D can differ from those in 2D. The ECM exhibits various structural features as well as complex mechanical properties. In 3D, mechanical confinement by the surrounding ECM restricts changes in cell volume and cell shape but allows cells to generate force on the matrix by extending protrusions and regulating cell volume as well as through actomyosin-based contractility. Furthermore, cell-matrix interactions are dynamic owing to matrix remodelling. Accordingly, ECM stiffness, viscoelasticity and degradability often play a critical role in regulating cell behaviours in 3D. Mechanisms of 3D mechanotransduction include traditional integrin-mediated pathways that sense mechanical properties and more recently described mechanosensitive ion channel-mediated pathways that sense 3D confinement, with both converging on the nucleus for downstream control of transcription and phenotype. Mechanotransduction is involved in tissues from development to cancer and is being increasingly harnessed towards mechanotherapy. Here we discuss recent progress in our understanding of cell-ECM mechanotransduction in 3D.
Collapse
Affiliation(s)
| | - Dhiraj Indana
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA.
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA.
| |
Collapse
|
25
|
Günay KA, Chang TL, Skillin NP, Rao VV, Macdougall LJ, Cutler AA, Silver JS, Brown TE, Zhang C, Yu CCJ, Olwin BB, Boyden ES, Anseth KS. Photo-expansion microscopy enables super-resolution imaging of cells embedded in 3D hydrogels. NATURE MATERIALS 2023; 22:777-785. [PMID: 37217701 PMCID: PMC10590656 DOI: 10.1038/s41563-023-01558-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 04/17/2023] [Indexed: 05/24/2023]
Abstract
Hydrogels are extensively used as tunable, biomimetic three-dimensional cell culture matrices, but optically deep, high-resolution images are often difficult to obtain, limiting nanoscale quantification of cell-matrix interactions and outside-in signalling. Here we present photopolymerized hydrogels for expansion microscopy that enable optical clearance and tunable ×4.6-6.7 homogeneous expansion of not only monolayer cell cultures and tissue sections, but cells embedded within hydrogels. The photopolymerized hydrogels for expansion microscopy formulation relies on a rapid photoinitiated thiol/acrylate mixed-mode polymerization that is not inhibited by oxygen and decouples monomer diffusion from polymerization, which is particularly beneficial when expanding cells embedded within hydrogels. Using this technology, we visualize human mesenchymal stem cells and their interactions with nascently deposited proteins at <120 nm resolution when cultured in proteolytically degradable synthetic polyethylene glycol hydrogels. Results support the notion that focal adhesion maturation requires cellular fibronectin deposition; nuclear deformation precedes cellular spreading; and human mesenchymal stem cells display cell-surface metalloproteinases for matrix remodelling.
Collapse
Affiliation(s)
- Kemal Arda Günay
- Department of Chemical and Biological Engineering and the BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Tze-Ling Chang
- Department of Chemical and Biological Engineering and the BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Nathaniel P Skillin
- Department of Chemical and Biological Engineering and the BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Varsha V Rao
- Department of Chemical and Biological Engineering and the BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Laura J Macdougall
- Department of Chemical and Biological Engineering and the BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Alicia A Cutler
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA
| | - Jason S Silver
- Department of Chemical and Biological Engineering and the BioFrontiers Institute, University of Colorado, Boulder, CO, USA
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA
| | - Tobin E Brown
- Department of Chemical and Biological Engineering and the BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Chi Zhang
- McGovern Institute, MIT, Cambridge, MA, USA
- HHMI, Cambridge, MA, USA
- Departments of Brain and Cognitive Sciences, Media Arts and Sciences, and Biological Engineering, MIT, Cambridge, MA, USA
| | - Chih-Chieh Jay Yu
- McGovern Institute, MIT, Cambridge, MA, USA
- HHMI, Cambridge, MA, USA
- Departments of Brain and Cognitive Sciences, Media Arts and Sciences, and Biological Engineering, MIT, Cambridge, MA, USA
| | - Bradley B Olwin
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA
| | - Edward S Boyden
- McGovern Institute, MIT, Cambridge, MA, USA
- HHMI, Cambridge, MA, USA
- Departments of Brain and Cognitive Sciences, Media Arts and Sciences, and Biological Engineering, MIT, Cambridge, MA, USA
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering and the BioFrontiers Institute, University of Colorado, Boulder, CO, USA.
| |
Collapse
|
26
|
Peng L, Matellan C, Bosch‐Fortea M, Gonzalez‐Molina J, Frigerio M, Salentinig S, del Rio Hernandez A, Gautrot JE. Mesenchymal Stem Cells Sense the Toughness of Nanomaterials and Interfaces. Adv Healthc Mater 2023; 12:e2203297. [PMID: 36717365 PMCID: PMC11468436 DOI: 10.1002/adhm.202203297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/20/2023] [Indexed: 02/01/2023]
Abstract
Stem cells are known to sense and respond to the mechanical properties of biomaterials. In turn, cells exert forces on their environment that can lead to striking changes in shape, size and contraction of associated tissues, and may result in mechanical disruption and functional failure. However, no study has so far correlated stem cell phenotype and biomaterials toughness. Indeed, disentangling toughness-mediated cell response from other mechanosensing processes has remained elusive as it is particularly challenging to uncouple Youngs' or shear moduli from toughness, within a range relevant to cell-generated forces. In this report, it is shown how the design of the macromolecular architecture of polymer nanosheets regulates interfacial toughness, independently of interfacial shear storage modulus, and how this controls the expansion of mesenchymal stem cells at liquid interfaces. The viscoelasticity and toughness of poly(l-lysine) nanosheets assembled at liquid-liquid interfaces is characterised via interfacial shear rheology. The local (microscale) mechanics of nanosheets are characterised via magnetic tweezer-assisted interfacial microrheology and the thickness of these assemblies is determined from in situ ellipsometry. Finally, the response of mesenchymal stem cells to adhesion and culture at corresponding interfaces is investigated via immunostaining and confocal microscopy.
Collapse
Affiliation(s)
- Lihui Peng
- Institute of BioengineeringQueen Mary University of LondonMile End RoadE1 4NSLondonUK
- Cellular and Molecular Biomechanical LaboratoryDepartment of BioengineeringImperial College LondonLondonSW7 2AZUK
| | - Carlos Matellan
- School of Engineering and Materials ScienceQueen MaryUniversity of LondonMile End RoadLondonE1 4NSUK
| | - Minerva Bosch‐Fortea
- Institute of BioengineeringQueen Mary University of LondonMile End RoadE1 4NSLondonUK
- Cellular and Molecular Biomechanical LaboratoryDepartment of BioengineeringImperial College LondonLondonSW7 2AZUK
| | - Jordi Gonzalez‐Molina
- Institute of BioengineeringQueen Mary University of LondonMile End RoadE1 4NSLondonUK
- Cellular and Molecular Biomechanical LaboratoryDepartment of BioengineeringImperial College LondonLondonSW7 2AZUK
| | - Matteo Frigerio
- Department of ChemistryUniversity of FribourgChemin du Musée 9Fribourg1700Switzerland
| | - Stefan Salentinig
- Department of ChemistryUniversity of FribourgChemin du Musée 9Fribourg1700Switzerland
| | - Armando del Rio Hernandez
- School of Engineering and Materials ScienceQueen MaryUniversity of LondonMile End RoadLondonE1 4NSUK
| | - Julien E. Gautrot
- Institute of BioengineeringQueen Mary University of LondonMile End RoadE1 4NSLondonUK
- Cellular and Molecular Biomechanical LaboratoryDepartment of BioengineeringImperial College LondonLondonSW7 2AZUK
| |
Collapse
|
27
|
Matthiesen I, Jury M, Rasti Boroojeni F, Ludwig SL, Holzreuter M, Buchmann S, Åman Träger A, Selegård R, Winkler TE, Aili D, Herland A. Astrocyte 3D culture and bioprinting using peptide functionalized hyaluronan hydrogels. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2023; 24:2165871. [PMID: 36733710 PMCID: PMC9888471 DOI: 10.1080/14686996.2023.2165871] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/20/2022] [Accepted: 01/04/2023] [Indexed: 06/18/2023]
Abstract
Astrocytes play an important role in the central nervous system, contributing to the development of and maintenance of synapses, recycling of neurotransmitters, and the integrity and function of the blood-brain barrier. Astrocytes are also linked to the pathophysiology of various neurodegenerative diseases. Astrocyte function and organization are tightly regulated by interactions mediated by the extracellular matrix (ECM). Engineered hydrogels can mimic key aspects of the ECM and can allow for systematic studies of ECM-related factors that govern astrocyte behaviour. In this study, we explore the interactions between neuroblastoma (SH-SY5Y) and glioblastoma (U87) cell lines and human fetal primary astrocytes (FPA) with a modular hyaluronan-based hydrogel system. Morphological analysis reveals that FPA have a higher degree of interactions with the hyaluronan-based gels compared to the cell lines. This interaction is enhanced by conjugation of cell-adhesion peptides (cRGD and IKVAV) to the hyaluronan backbone. These effects are retained and pronounced in 3D bioprinted structures. Bioprinted FPA using cRGD functionalized hyaluronan show extensive and defined protrusions and multiple connections between neighboring cells. Possibilities to tailor and optimize astrocyte-compatible ECM-mimicking hydrogels that can be processed by means of additive biofabrication can facilitate the development of advanced tissue and disease models of the central nervous system.
Collapse
Affiliation(s)
- Isabelle Matthiesen
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
- CVRM Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Michael Jury
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Fatemeh Rasti Boroojeni
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Saskia L. Ludwig
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Muriel Holzreuter
- AIMES, Center for Integrated Medical and Engineering Science, Department of Neuroscience, Karolinska Institute, Solna, Sweden
| | - Sebastian Buchmann
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
- AIMES, Center for Integrated Medical and Engineering Science, Department of Neuroscience, Karolinska Institute, Solna, Sweden
- Division of Nanobiotechnology, Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden
| | - Andrea Åman Träger
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Robert Selegård
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Thomas E. Winkler
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
- Institute of Microtechnology & Center of Pharmaceutical Engineering, Technische Universität Braunschweig, Braunschweig, Germany
| | - Daniel Aili
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Anna Herland
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
- AIMES, Center for Integrated Medical and Engineering Science, Department of Neuroscience, Karolinska Institute, Solna, Sweden
- Division of Nanobiotechnology, Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden
| |
Collapse
|
28
|
Chen X, Zhu M, Tang Y, Xie H, Fan X. Methine initiated polypropylene-based disposable face masks aging validated by micromechanical properties loss of atomic force microscopy. JOURNAL OF HAZARDOUS MATERIALS 2023; 441:129831. [PMID: 36084457 PMCID: PMC9398948 DOI: 10.1016/j.jhazmat.2022.129831] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/06/2022] [Accepted: 08/21/2022] [Indexed: 06/15/2023]
Abstract
The contagious coronavirus disease-2019 pandemic has led to an increasing number of disposable face masks (DFMs) abandoned in the environment, when they are exposed to the air condition, the broken of chemical bond induced aging is inevitably occurred which meantime would cause a drastic decrease of the mechanical flexibility. However, the understanding of between chemical bond change related to aging and its micromechanical loss is limited due to the lack of refined techniques. Herein, the atomic force microscopy (AFM) technique was firstly used to observe the aging process induced by methine of the polypropylene-based DFMs. By comparing the micromechanical properties loss, the influences of humidity and light density on the DFM aging were systematically studied in the early 72 h, and it revealed that the increasing scissions number of the easiest attacked methine (Ct-H) can gradually decrease the micromechanical properties of the polypropylene (PP)-based DFM. Furthermore, the results are also validated by the in- situ FTIR and XPS analysis. This work discloses that an aging process can be initially estimated with the micromechanical changes observed by AFM, which offers fundamental data to manage this important emerging plastic pollution during COVID-19 pandemic.
Collapse
Affiliation(s)
- Xueqin Chen
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 510632, China
| | - Mude Zhu
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 510632, China
| | - Yi Tang
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 510632, China
| | - Huiyuan Xie
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 510632, China
| | - Xiaoyun Fan
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
29
|
Schot M, Araújo-Gomes N, van Loo B, Kamperman T, Leijten J. Scalable fabrication, compartmentalization and applications of living microtissues. Bioact Mater 2023; 19:392-405. [PMID: 35574053 PMCID: PMC9062422 DOI: 10.1016/j.bioactmat.2022.04.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/18/2022] [Accepted: 04/06/2022] [Indexed: 10/27/2022] Open
Abstract
Living microtissues are used in a multitude of applications as they more closely resemble native tissue physiology, as compared to 2D cultures. Microtissues are typically composed of a combination of cells and materials in varying combinations, which are dictated by the applications' design requirements. Their applications range wide, from fundamental biological research such as differentiation studies to industrial applications such as cruelty-free meat production. However, their translation to industrial and clinical settings has been hindered due to the lack of scalability of microtissue production techniques. Continuous microfluidic processes provide an opportunity to overcome this limitation as they offer higher throughput production rates as compared to traditional batch techniques, while maintaining reproducible control over microtissue composition and size. In this review, we provide a comprehensive overview of the current approaches to engineer microtissues with a focus on the advantages of, and need for, the use of continuous processes to produce microtissues in large quantities. Finally, an outlook is provided that outlines the required developments to enable large-scale microtissue fabrication using continuous processes.
Collapse
Affiliation(s)
- Maik Schot
- Department of Developmental Bioengineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522NB, Enschede, the Netherlands
| | - Nuno Araújo-Gomes
- Department of Developmental Bioengineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522NB, Enschede, the Netherlands
| | - Bas van Loo
- Department of Developmental Bioengineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522NB, Enschede, the Netherlands
| | - Tom Kamperman
- Department of Developmental Bioengineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522NB, Enschede, the Netherlands
| | - Jeroen Leijten
- Department of Developmental Bioengineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522NB, Enschede, the Netherlands
| |
Collapse
|
30
|
Blache U, Ford EM, Ha B, Rijns L, Chaudhuri O, Dankers PY, Kloxin AM, Snedeker JG, Gentleman E. Engineered hydrogels for mechanobiology. NATURE REVIEWS. METHODS PRIMERS 2022; 2:98. [PMID: 37461429 PMCID: PMC7614763 DOI: 10.1038/s43586-022-00179-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/17/2022] [Indexed: 07/20/2023]
Abstract
Cells' local mechanical environment can be as important in guiding cellular responses as many well-characterized biochemical cues. Hydrogels that mimic the native extracellular matrix can provide these mechanical cues to encapsulated cells, allowing for the study of their impact on cellular behaviours. Moreover, by harnessing cellular responses to mechanical cues, hydrogels can be used to create tissues in vitro for regenerative medicine applications and for disease modelling. This Primer outlines the importance and challenges of creating hydrogels that mimic the mechanical and biological properties of the native extracellular matrix. The design of hydrogels for mechanobiology studies is discussed, including appropriate choice of cross-linking chemistry and strategies to tailor hydrogel mechanical cues. Techniques for characterizing hydrogels are explained, highlighting methods used to analyze cell behaviour. Example applications for studying fundamental mechanobiological processes and regenerative therapies are provided, along with a discussion of the limitations of hydrogels as mimetics of the native extracellular matrix. The article ends with an outlook for the field, focusing on emerging technologies that will enable new insights into mechanobiology and its role in tissue homeostasis and disease.
Collapse
Affiliation(s)
- Ulrich Blache
- Fraunhofer Institute for Cell Therapy and Immunology and Fraunhofer Cluster of Excellence for Immune-Mediated Disease, Leipzig, Germany
| | - Eden M. Ford
- Department of Chemical and Biomolecular Engineering, University of Delaware, DE, USA
| | - Byunghang Ha
- Department of Mechanical Engineering, Stanford University, CA, USA
| | - Laura Rijns
- Institute for Complex Molecular Systems, Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, CA, USA
| | - Patricia Y.W. Dankers
- Institute for Complex Molecular Systems, Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | - April M. Kloxin
- Department of Chemical and Biomolecular Engineering, University of Delaware, DE, USA
- Department of Material Science and Engineering, University of Delaware, DE, USA
| | - Jess G. Snedeker
- University Hospital Balgrist and ETH Zurich, Zurich, Switzerland
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King’s College London, London SE1 9RT, UK
| |
Collapse
|
31
|
Kahle ER, Patel N, Sreenivasappa HB, Marcolongo MS, Han L. Targeting cell-matrix interface mechanobiology by integrating AFM with fluorescence microscopy. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 176:67-81. [PMID: 36055517 PMCID: PMC9691605 DOI: 10.1016/j.pbiomolbio.2022.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/14/2022] [Accepted: 08/18/2022] [Indexed: 06/15/2023]
Abstract
Mechanosensing at the interface of a cell and its surrounding microenvironment is an essential driving force of physiological processes. Understanding molecular activities at the cell-matrix interface has the potential to provide novel targets for improving tissue regeneration and early disease intervention. In the past few decades, the advancement of atomic force microscopy (AFM) has offered a unique platform for probing mechanobiology at this crucial microdomain. In this review, we describe key advances under this topic through the use of an integrated system of AFM (as a biomechanical testing tool) with complementary immunofluorescence (IF) imaging (as an in situ navigation system). We first describe the body of work investigating the micromechanics of the pericellular matrix (PCM), the immediate cell micro-niche, in healthy, diseased, and genetically modified tissues, with a focus on articular cartilage. We then summarize the key findings in understanding cellular biomechanics and mechanotransduction, in which, molecular mechanisms governing transmembrane ion channel-mediated mechanosensing, cytoskeleton remodeling, and nucleus remodeling have been studied in various cell and tissue types. Lastly, we provide an overview of major technical advances that have enabled more in-depth studies of mechanobiology, including the integration of AFM with a side-view microscope, multiple optomicroscopy, a fluorescence recovery after photobleaching (FRAP) module, and a tensile stretching device. The innovations described here have contributed greatly to advancing the fundamental knowledge of extracellular matrix biomechanics and cell mechanobiology for improved understanding, detection, and intervention of various diseases.
Collapse
Affiliation(s)
- Elizabeth R Kahle
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Neil Patel
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Harini B Sreenivasappa
- Cell Imaging Center, Office of Research and Innovation, Drexel University, PA 19104, United States
| | - Michele S Marcolongo
- Department of Mechanical Engineering, Villanova University, Villanova, PA 19085, United States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States.
| |
Collapse
|
32
|
Fischer NG, Aparicio C. Junctional epithelium and hemidesmosomes: Tape and rivets for solving the "percutaneous device dilemma" in dental and other permanent implants. Bioact Mater 2022; 18:178-198. [PMID: 35387164 PMCID: PMC8961425 DOI: 10.1016/j.bioactmat.2022.03.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/14/2022] [Accepted: 03/12/2022] [Indexed: 02/06/2023] Open
Abstract
The percutaneous device dilemma describes etiological factors, centered around the disrupted epithelial tissue surrounding non-remodelable devices, that contribute to rampant percutaneous device infection. Natural percutaneous organs, in particular their extracellular matrix mediating the "device"/epithelium interface, serve as exquisite examples to inspire longer lasting long-term percutaneous device design. For example, the tooth's imperviousness to infection is mediated by the epithelium directly surrounding it, the junctional epithelium (JE). The hallmark feature of JE is formation of hemidesmosomes, cell/matrix adhesive structures that attach surrounding oral gingiva to the tooth's enamel through a basement membrane. Here, the authors survey the multifaceted functions of the JE, emphasizing the role of the matrix, with a particular focus on hemidesmosomes and their five main components. The authors highlight the known (and unknown) effects dental implant - as a model percutaneous device - placement has on JE regeneration and synthesize this information for application to other percutaneous devices. The authors conclude with a summary of bioengineering strategies aimed at solving the percutaneous device dilemma and invigorating greater collaboration between clinicians, bioengineers, and matrix biologists.
Collapse
Affiliation(s)
- Nicholas G. Fischer
- MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 16-212 Moos Tower, 515 Delaware St. SE, Minneapolis, MN, 55455, USA
| | - Conrado Aparicio
- MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 16-212 Moos Tower, 515 Delaware St. SE, Minneapolis, MN, 55455, USA
- Division of Basic Research, Faculty of Odontology, UIC Barcelona – Universitat Internacional de Catalunya, C/. Josep Trueta s/n, 08195, Sant Cugat del Valles, Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), C/. Baldiri Reixac 10-12, 08028, Barcelona, Spain
| |
Collapse
|
33
|
Ke W, Ma L, Wang B, Song Y, Luo R, Li G, Liao Z, Shi Y, Wang K, Feng X, Li S, Hua W, Yang C. N-cadherin mimetic hydrogel enhances MSC chondrogenesis through cell metabolism. Acta Biomater 2022; 150:83-95. [DOI: 10.1016/j.actbio.2022.07.050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 07/17/2022] [Accepted: 07/26/2022] [Indexed: 02/07/2023]
|
34
|
Barnett H, Shevchuk M, Peppas NA, Caldorera-Moore M. Influence of extracellular cues of hydrogel biomaterials on stem cell fate. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2022; 33:1324-1347. [PMID: 35297325 DOI: 10.1080/09205063.2022.2054398] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 03/10/2022] [Accepted: 03/14/2022] [Indexed: 06/14/2023]
Abstract
Tissue engineering is a multidisciplinary field that focuses on creating functional tissue through the combination of biomimetic scaffolds, a cell source, and biochemical/physiochemical cues. Stem cells are often used as the cell source due to their multipotent properties and autologous sourcing; however, the combination of physical and chemical cues that regulate their behavior creates challenges in reproducibly directing them to a specific fate. Hydrogel biomaterials are widely explored as tissue scaffolds due to their innate biomimetic properties and tailorability. For these constructs to be successful, properties such as surface chemistry and spatial configuration, stiffness, and degradability of the biomaterial used for the scaffold framework should be analogous to the natural environment of the tissue they are repairing/replacing. This is imperative, as cues from the surrounding extracellular matrix (ECM) influence stem cell behavior and direct cell differentiation to a specific lineage. Hydrogels offer great promise as tools to control stem cell fate, as researchers can modulate the degradation rates, mechanical properties, swelling behavior, and chemical properties of the biomaterial scaffold to mimic the instructive cues of the native ECM. Discussion of the advantages and challenges of utilizing hydrogel biomaterials as the basis of tissue scaffolds is reviewed herein, as well as specific examples of hydrogels in tissue engineering and advances in hydrogel research to achieve desired cell phenotypes.
Collapse
Affiliation(s)
- Haley Barnett
- School of Sciences, University of Louisiana Monroe, Monroe, LA, USA
| | - Mariya Shevchuk
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute of Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Nicholas A Peppas
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute of Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Department of Pediatrics, and Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| | - Mary Caldorera-Moore
- Department of Biomedical Engineering, Louisiana Tech University, Ruston, LA, USA
| |
Collapse
|
35
|
Baek J, Lopez PA, Lee S, Kim TS, Kumar S, Schaffer DV. Egr1 is a 3D matrix-specific mediator of mechanosensitive stem cell lineage commitment. SCIENCE ADVANCES 2022; 8:eabm4646. [PMID: 35427160 PMCID: PMC9012469 DOI: 10.1126/sciadv.abm4646] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 02/28/2022] [Indexed: 05/31/2023]
Abstract
While extracellular matrix (ECM) mechanics strongly regulate stem cell commitment, the field's mechanistic understanding of this phenomenon largely derives from simplified two-dimensional (2D) culture substrates. Here, we found a 3D matrix-specific mechanoresponsive mechanism for neural stem cell (NSC) differentiation. NSC lineage commitment in 3D is maximally stiffness sensitive in the range of 0.1 to 1.2 kPa, a narrower and more brain-mimetic range than we had previously identified in 2D (0.75 to 75 kPa). Transcriptomics revealed stiffness-dependent up-regulation of early growth response 1 (Egr1) in 3D but not in 2D. Egr1 knockdown enhanced neurogenesis in stiff ECMs by driving β-catenin nuclear localization and activity in 3D, but not in 2D. Mechanical modeling and experimental studies under osmotic pressure indicate that stiff 3D ECMs are likely to stimulate Egr1 via increases in confining stress during cell volumetric growth. To our knowledge, Egr1 represents the first 3D-specific stem cell mechanoregulatory factor.
Collapse
Affiliation(s)
- Jieung Baek
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Paola A. Lopez
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- UC Berkeley–UC San Francisco Graduate Program in Bioengineering, Berkeley, CA 94720, USA
| | - Sangmin Lee
- Department of Mechanical Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Taek-Soo Kim
- Department of Mechanical Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
- UC Berkeley–UC San Francisco Graduate Program in Bioengineering, Berkeley, CA 94720, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - David V. Schaffer
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
- UC Berkeley–UC San Francisco Graduate Program in Bioengineering, Berkeley, CA 94720, USA
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Helen Wills Neuroscience Institute, Berkeley, CA 94720, USA
| |
Collapse
|
36
|
Liu AP, Appel EA, Ashby PD, Baker BM, Franco E, Gu L, Haynes K, Joshi NS, Kloxin AM, Kouwer PHJ, Mittal J, Morsut L, Noireaux V, Parekh S, Schulman R, Tang SKY, Valentine MT, Vega SL, Weber W, Stephanopoulos N, Chaudhuri O. The living interface between synthetic biology and biomaterial design. NATURE MATERIALS 2022; 21:390-397. [PMID: 35361951 PMCID: PMC10265650 DOI: 10.1038/s41563-022-01231-3] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 03/07/2022] [Indexed: 06/14/2023]
Abstract
Recent far-reaching advances in synthetic biology have yielded exciting tools for the creation of new materials. Conversely, advances in the fundamental understanding of soft-condensed matter, polymers and biomaterials offer new avenues to extend the reach of synthetic biology. The broad and exciting range of possible applications have substantial implications to address grand challenges in health, biotechnology and sustainability. Despite the potentially transformative impact that lies at the interface of synthetic biology and biomaterials, the two fields have, so far, progressed mostly separately. This Perspective provides a review of recent key advances in these two fields, and a roadmap for collaboration at the interface between the two communities. We highlight the near-term applications of this interface to the development of hierarchically structured biomaterials, from bioinspired building blocks to 'living' materials that sense and respond based on the reciprocal interactions between materials and embedded cells.
Collapse
Affiliation(s)
- Allen P Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA.
| | - Eric A Appel
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, USA
| | - Paul D Ashby
- Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Elisa Franco
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Luo Gu
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Karmella Haynes
- Wallace H. Coulter Department of Biomedical Engineering, Emory University, Atlanta, GA, USA
| | - Neel S Joshi
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, USA
| | - April M Kloxin
- Department of Chemical and Biomolecular Engineering and Materials Science and Engineering, University of Delaware, Newark, DE, USA
| | - Paul H J Kouwer
- Institute for Molecules and Materials, Radboud University, Nijmegen, the Netherlands
| | - Jeetain Mittal
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Leonardo Morsut
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Vincent Noireaux
- School of Physics and Astronomy, University of Minnesota, Minneapolis, MN, USA
| | - Sapun Parekh
- Department of Biomedical Engineering, University of Texas, Austin, Austin, TX, USA
| | - Rebecca Schulman
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Sindy K Y Tang
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Megan T Valentine
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Sebastián L Vega
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ, USA
| | - Wilfried Weber
- Faculty of Biology and Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | | | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
37
|
Narkar AR, Tong Z, Soman P, Henderson JH. Smart biomaterial platforms: Controlling and being controlled by cells. Biomaterials 2022; 283:121450. [PMID: 35247636 PMCID: PMC8977253 DOI: 10.1016/j.biomaterials.2022.121450] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/23/2022] [Accepted: 02/26/2022] [Indexed: 02/07/2023]
Abstract
Across diverse research and application areas, dynamic functionality-such as programmable changes in biochemical property, in mechanical property, or in microscopic or macroscopic architecture-is an increasingly common biomaterials design criterion, joining long-studied criteria such as cytocompatibility and biocompatibility, drug release kinetics, and controlled degradability or long-term stability in vivo. Despite tremendous effort, achieving dynamic functionality while simultaneously maintaining other desired design criteria remains a significant challenge. Reversible dynamic functionality, rather than one-time or one-way dynamic functionality, is of particular interest but has proven especially challenging. Such reversible functionality could enable studies that address the current gap between the dynamic nature of in vivo biological and biomechanical processes, such as cell traction, cell-extracellular matrix (ECM) interactions, and cell-mediated ECM remodeling, and the static nature of the substrates and ECM constructs used to study the processes. This review assesses dynamic materials that have traditionally been used to control cell activity and static biomaterial constructs, experimental and computational techniques, with features that may inform continued advances in reversible dynamic materials. Taken together, this review presents a perspective on combining the reversibility of smart materials and the in-depth dynamic cell behavior probed by static polymers to design smart bi-directional ECM platforms that can reversibly and repeatedly communicate with cells.
Collapse
Affiliation(s)
- Ameya R Narkar
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, NY, 13244, United States; Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, 13244, United States.
| | - Zhuoqi Tong
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, NY, 13244, United States; Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, 13244, United States.
| | - Pranav Soman
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, NY, 13244, United States; Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, 13244, United States.
| | - James H Henderson
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, NY, 13244, United States; Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, 13244, United States.
| |
Collapse
|
38
|
Bernal PN, Bouwmeester M, Madrid-Wolff J, Falandt M, Florczak S, Rodriguez NG, Li Y, Größbacher G, Samsom RA, van Wolferen M, van der Laan LJW, Delrot P, Loterie D, Malda J, Moser C, Spee B, Levato R. Volumetric Bioprinting of Organoids and Optically Tuned Hydrogels to Build Liver-Like Metabolic Biofactories. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2110054. [PMID: 35166410 DOI: 10.1002/adma.202110054] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/28/2022] [Indexed: 06/14/2023]
Abstract
Organ- and tissue-level biological functions are intimately linked to microscale cell-cell interactions and to the overarching tissue architecture. Together, biofabrication and organoid technologies offer the unique potential to engineer multi-scale living constructs, with cellular microenvironments formed by stem cell self-assembled structures embedded in customizable bioprinted geometries. This study introduces the volumetric bioprinting of complex organoid-laden constructs, which capture key functions of the human liver. Volumetric bioprinting via optical tomography shapes organoid-laden gelatin hydrogels into complex centimeter-scale 3D structures in under 20 s. Optically tuned bioresins enable refractive index matching of specific intracellular structures, countering the disruptive impact of cell-mediated light scattering on printing resolution. This layerless, nozzle-free technique poses no harmful mechanical stresses on organoids, resulting in superior viability and morphology preservation post-printing. Bioprinted organoids undergo hepatocytic differentiation showing albumin synthesis, liver-specific enzyme activity, and remarkably acquired native-like polarization. Organoids embedded within low stiffness gelatins (<2 kPa) are bioprinted into mathematically defined lattices with varying degrees of pore network tortuosity, and cultured under perfusion. These structures act as metabolic biofactories in which liver-specific ammonia detoxification can be enhanced by the architectural profile of the constructs. This technology opens up new possibilities for regenerative medicine and personalized drug testing.
Collapse
Affiliation(s)
- Paulina Nuñez Bernal
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
| | - Manon Bouwmeester
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CT, The Netherlands
| | - Jorge Madrid-Wolff
- Laboratory of Applied Photonics Devices, École Polytechnique Fédéral Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Marc Falandt
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CT, The Netherlands
| | - Sammy Florczak
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
| | - Nuria Ginés Rodriguez
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
| | - Yang Li
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
| | - Gabriel Größbacher
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
| | - Roos-Anne Samsom
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CT, The Netherlands
| | - Monique van Wolferen
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CT, The Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, 3015GD, The Netherlands
| | - Paul Delrot
- Readily3D SA, EPFL Innovation Park, Building A, Lausanne, CH-1015, Switzerland
| | - Damien Loterie
- Readily3D SA, EPFL Innovation Park, Building A, Lausanne, CH-1015, Switzerland
| | - Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CT, The Netherlands
| | - Christophe Moser
- Laboratory of Applied Photonics Devices, École Polytechnique Fédéral Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Bart Spee
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CT, The Netherlands
| | - Riccardo Levato
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CT, The Netherlands
| |
Collapse
|
39
|
Fischer NG, Kobe AC, Dai J, He J, Wang H, Pizarek JA, De Jong DA, Ye Z, Huang S, Aparicio C. Tapping basement membrane motifs: Oral junctional epithelium for surface-mediated soft tissue attachment to prevent failure of percutaneous devices. Acta Biomater 2022; 141:70-88. [PMID: 34971784 PMCID: PMC8898307 DOI: 10.1016/j.actbio.2021.12.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 01/08/2023]
Abstract
Teeth, long-lasting percutaneous organs, feature soft tissue attachment through adhesive structures, hemidesmosomes, in the junctional epithelium basement membrane adjacent to teeth. This soft tissue attachment prevents bacterial infection of the tooth despite the rich - and harsh - microbial composition of the oral cavity. Conversely, millions of percutaneous devices (catheters, dental, and orthopedic implants) fail from infection yearly. Standard of care antibiotic usage fuels antimicrobial resistance and is frequently ineffective. Infection prevention strategies, like for dental implants, have failed in generating durable soft tissue adhesion - like that seen with the tooth - to prevent bacterial colonization at the tissue-device interface. Here, inspired by the impervious natural attachment of the junctional epithelium to teeth, we synthesized four cell adhesion peptide (CAPs) nanocoatings, derived from basement membranes, to promote percutaneous device soft tissue attachment. The two leading nanocoatings upregulated integrin-mediated hemidesmosomes, selectively increased keratinocyte proliferation compared to fibroblasts, which cannot form hemidesmosomes, and expression of junctional epithelium adhesive markers. CAP nanocoatings displayed marked durability under simulated clinical conditions and the top performer CAP nanocoating was validated in a percutaneous implant murine model. Basement membrane CAP nanocoatings, inspired by the tooth and junctional epithelium, may provide an alternative anti-infective strategy for percutaneous devices to mitigate the worldwide threat of antimicrobial resistance. STATEMENT OF SIGNIFICANCE: Prevention and management of medical device infection is a significant healthcare challenge. Overzealous antibiotic use has motivated alternative material innovations to prevent infection. Here, we report implant cell adhesion peptide nanocoatings that mimic a long-lasting, natural "medical device," the tooth, through formation of cell adhesive structures called hemidesmosomes. Such nanocoatings sidestep the use of antimicrobial or antibiotic elements to form a soft-tissue seal around implants. The top performing nanocoatings prompted expression of hemidesmosomes and defensive factors to mimic the tooth and was validated in an animal model. Application of cell adhesion peptide nanocoatings may provide an alternative to preventing, rather that necessarily treating, medical device infection across a range of device indications, like dental implants.
Collapse
Affiliation(s)
- Nicholas G Fischer
- MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 16-212 Moos Tower, 515 Delaware St. SE, Minneapolis, Minnesota 55455, United States
| | - Alexandra C Kobe
- MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 16-212 Moos Tower, 515 Delaware St. SE, Minneapolis, Minnesota 55455, United States
| | - Jinhong Dai
- Institute of Stomatology, School and Hospital of Stomatology, Department of Prosthodontics, Wenzhou Medical University, 373 Xueyuan Xi Road, Wenzhou, Zhejiang 325027, China
| | - Jiahe He
- MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 16-212 Moos Tower, 515 Delaware St. SE, Minneapolis, Minnesota 55455, United States
| | - Hongning Wang
- Institute of Stomatology, School and Hospital of Stomatology, Department of Prosthodontics, Wenzhou Medical University, 373 Xueyuan Xi Road, Wenzhou, Zhejiang 325027, China
| | - John A Pizarek
- MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 16-212 Moos Tower, 515 Delaware St. SE, Minneapolis, Minnesota 55455, United States; United States Navy Dental Corps, Naval Medical Leader and Professional Development Command, 8955 Wood Road Bethesda, MD 20889, United States
| | - David A De Jong
- MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 16-212 Moos Tower, 515 Delaware St. SE, Minneapolis, Minnesota 55455, United States
| | - Zhou Ye
- MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 16-212 Moos Tower, 515 Delaware St. SE, Minneapolis, Minnesota 55455, United States
| | - Shengbin Huang
- Institute of Stomatology, School and Hospital of Stomatology, Department of Prosthodontics, Wenzhou Medical University, 373 Xueyuan Xi Road, Wenzhou, Zhejiang 325027, China
| | - Conrado Aparicio
- MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 16-212 Moos Tower, 515 Delaware St. SE, Minneapolis, Minnesota 55455, United States.
| |
Collapse
|
40
|
Aprile P, Whelan IT, Sathy BN, Carroll SF, Kelly DJ. Soft Hydrogel Environments that Facilitate Cell Spreading and Aggregation Preferentially Support Chondrogenesis of Adult Stem Cells. Macromol Biosci 2022; 22:e2100365. [PMID: 35171524 DOI: 10.1002/mabi.202100365] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/14/2022] [Indexed: 11/10/2022]
Abstract
Mesenchymal stem/stromal cells (MSCs) represent a promising cell type for treating damaged and diseased synovial joints. The therapeutic potential of MSCs will be facilitated by the engineering of biomaterial environments capable of directing their fate. Here we explored the interplay between matrix elasticity and cell morphology in regulating the chondrogenic differentiation of MSCs when seeded onto or encapsulated within hydrogels made of interpenetrating networks (IPN) of alginate and collagen type I. This IPN system enabled the independent control of substrate stiffness (in 2D and in 3D) and cell morphology (3D only). In a 2D culture environment, the expression of chondrogenic markers SOX9, ACAN and COL2 increased on a soft substrate, which correlated with increased SMAD2/3 nuclear localization, enhanced MSCs condensation and the formation of larger cellular aggregates. The encapsulation of spread MSCs within a soft IPN dramatically increased the expression of cartilage-specific genes, which was linked to higher levels of cellular condensation and nuclear SMAD2/3 localization. Surprisingly, cells forced to adopt a more rounded morphology within the same soft IPNs expressed higher levels of the osteogenic markers RUNX2 and COL1. The insight provided by this study suggests that a mechanobiology informed approach to biomaterial development will be integral to the development of successful cartilage tissue engineering strategies. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Paola Aprile
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Ian T Whelan
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.,CÚRAM Center for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - Binulal N Sathy
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.,Centre for Nanoscience and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Simon F Carroll
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Daniel J Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.,CÚRAM Center for Research in Medical Devices, National University of Ireland, Galway, Ireland.,The Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, Trinity College Dublin, Ireland
| |
Collapse
|
41
|
Kahle ER, Han B, Chandrasekaran P, Phillips ER, Mulcahey MK, Lu XL, Marcolongo MS, Han L. Molecular Engineering of Pericellular Microniche via Biomimetic Proteoglycans Modulates Cell Mechanobiology. ACS NANO 2022; 16:1220-1230. [PMID: 35015500 PMCID: PMC9271520 DOI: 10.1021/acsnano.1c09015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Molecular engineering of biological tissues using synthetic mimics of native matrix molecules can modulate the mechanical properties of the cellular microenvironment through physical interactions with existing matrix molecules, and in turn, mediate the corresponding cell mechanobiology. In articular cartilage, the pericellular matrix (PCM) is the immediate microniche that regulates cell fate, signaling, and metabolism. The negatively charged osmo-environment, as endowed by PCM proteoglycans, is a key biophysical cue for cell mechanosensing. This study demonstrated that biomimetic proteoglycans (BPGs), which mimic the ultrastructure and polyanionic nature of native proteoglycans, can be used to molecularly engineer PCM micromechanics and cell mechanotransduction in cartilage. Upon infiltration into bovine cartilage explant, we showed that localization of BPGs in the PCM leads to increased PCM micromodulus and enhanced chondrocyte intracellular calcium signaling. Applying molecular force spectroscopy, we revealed that BPGs integrate with native PCM through augmenting the molecular adhesion of aggrecan, the major PCM proteoglycan, at the nanoscale. These interactions are enabled by the biomimetic "bottle-brush" ultrastructure of BPGs and facilitate the integration of BPGs within the PCM. Thus, this class of biomimetic molecules can be used for modulating molecular interactions of pericellular proteoglycans and harnessing cell mechanosensing. Because the PCM is a prevalent feature of various cell types, BPGs hold promising potential for improving regeneration and disease modification for not only cartilage-related healthcare but many other tissues and diseases.
Collapse
Affiliation(s)
- Elizabeth R. Kahle
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Biao Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Prashant Chandrasekaran
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Evan R. Phillips
- Department of Materials Science and Engineering, Drexel University, Philadelphia, PA 19104, United States
| | - Mary K. Mulcahey
- Department of Orthopaedic Surgery, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - X. Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - Michele S. Marcolongo
- Department of Materials Science and Engineering, Drexel University, Philadelphia, PA 19104, United States
- Department of Mechanical Engineering, Villanova University, Villanova, PA 19085, United States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| |
Collapse
|
42
|
Liu Z, Cai M, Zhang X, Yu X, Wang S, Wan X, Wang ZL, Li L. Cell-Traction-Triggered On-Demand Electrical Stimulation for Neuron-Like Differentiation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2106317. [PMID: 34655105 DOI: 10.1002/adma.202106317] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/15/2021] [Indexed: 06/13/2023]
Abstract
Electromechanical interaction of cells and extracellular matrix are ubiquitous in biological systems. Understanding the fundamentals of this interaction and feedback is critical to design next-generation electroactive tissue engineering scaffold. Herein, based on elaborately modulating the dynamic mechanical forces in cell microenvironment, the design of a smart piezoelectric scaffold with suitable stiffness analogous to that of collagen for on-demand electrical stimulation is reported. Specifically, it generated a piezoelectric potential, namely a piezopotential, to stimulate stem cell differentiation with cell traction as a loop feedback signal, thereby avoiding the unfavorable effect of early electrical stimulation on cell spreading and adhesion. This is the first time to adapt to the dynamic microenvironment of cells and meet the electrical stimulation of cells in different states by a constant scaffold, diminishing the cumbersomeness of inducing material transformation or trigging by an external stimulus. This in situ on-demand electrical stimulation based on cell-traction-mediated piezopotential paves the way for smart scaffolds design and future bioelectronic therapies.
Collapse
Affiliation(s)
- Zhirong Liu
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Mingjun Cai
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
| | - Xiaodi Zhang
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, P. R. China
| | - Xin Yu
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, P. R. China
| | - Shu Wang
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, P. R. China
| | - Xingyi Wan
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zhong Lin Wang
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0245, USA
| | - Linlin Li
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
43
|
Kamperman T, Henke S, Crispim JF, Willemen NGA, Dijkstra PJ, Lee W, Offerhaus HL, Neubauer M, Smink AM, de Vos P, de Haan BJ, Karperien M, Shin SR, Leijten J. Tethering Cells via Enzymatic Oxidative Crosslinking Enables Mechanotransduction in Non-Cell-Adhesive Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2102660. [PMID: 34476848 PMCID: PMC8530967 DOI: 10.1002/adma.202102660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/10/2021] [Indexed: 05/14/2023]
Abstract
Cell-matrix interactions govern cell behavior and tissue function by facilitating transduction of biomechanical cues. Engineered tissues often incorporate these interactions by employing cell-adhesive materials. However, using constitutively active cell-adhesive materials impedes control over cell fate and elicits inflammatory responses upon implantation. Here, an alternative cell-material interaction strategy that provides mechanotransducive properties via discrete inducible on-cell crosslinking (DOCKING) of materials, including those that are inherently non-cell-adhesive, is introduced. Specifically, tyramine-functionalized materials are tethered to tyrosines that are naturally present in extracellular protein domains via enzyme-mediated oxidative crosslinking. Temporal control over the stiffness of on-cell tethered 3D microniches reveals that DOCKING uniquely enables lineage programming of stem cells by targeting adhesome-related mechanotransduction pathways acting independently of cell volume changes and spreading. In short, DOCKING represents a bioinspired and cytocompatible cell-tethering strategy that offers new routes to study and engineer cell-material interactions, thereby advancing applications ranging from drug delivery, to cell-based therapy, and cultured meat.
Collapse
Affiliation(s)
- Tom Kamperman
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
- Division of Engineering in MedicineBrigham and Women's HospitalHarvard Medical School65 Landsdowne StreetCambridgeMA02139USA
| | - Sieger Henke
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - João F. Crispim
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Niels G. A. Willemen
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Pieter J. Dijkstra
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Wooje Lee
- Optical SciencesMESA+ Institute for NanotechnologyUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Herman L. Offerhaus
- Optical SciencesMESA+ Institute for NanotechnologyUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Martin Neubauer
- Physical Chemistry IIUniversity of BayreuthUniversitätsstrasse 30D‐95447BayreuthGermany
| | - Alexandra M. Smink
- Department of Pathology and Medical BiologySection of ImmunoendocrinologyUniversity of GroningenUniversity Medical Center GroningenHanzeplein 1 (EA11)Groningen9713 GZThe Netherlands
| | - Paul de Vos
- Department of Pathology and Medical BiologySection of ImmunoendocrinologyUniversity of GroningenUniversity Medical Center GroningenHanzeplein 1 (EA11)Groningen9713 GZThe Netherlands
| | - Bart J. de Haan
- Department of Pathology and Medical BiologySection of ImmunoendocrinologyUniversity of GroningenUniversity Medical Center GroningenHanzeplein 1 (EA11)Groningen9713 GZThe Netherlands
| | - Marcel Karperien
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Su Ryon Shin
- Division of Engineering in MedicineBrigham and Women's HospitalHarvard Medical School65 Landsdowne StreetCambridgeMA02139USA
| | - Jeroen Leijten
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| |
Collapse
|
44
|
Vernerey FJ, Lalitha Sridhar S, Muralidharan A, Bryant SJ. Mechanics of 3D Cell-Hydrogel Interactions: Experiments, Models, and Mechanisms. Chem Rev 2021; 121:11085-11148. [PMID: 34473466 DOI: 10.1021/acs.chemrev.1c00046] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hydrogels are highly water-swollen molecular networks that are ideal platforms to create tissue mimetics owing to their vast and tunable properties. As such, hydrogels are promising cell-delivery vehicles for applications in tissue engineering and have also emerged as an important base for ex vivo models to study healthy and pathophysiological events in a carefully controlled three-dimensional environment. Cells are readily encapsulated in hydrogels resulting in a plethora of biochemical and mechanical communication mechanisms, which recapitulates the natural cell and extracellular matrix interaction in tissues. These interactions are complex, with multiple events that are invariably coupled and spanning multiple length and time scales. To study and identify the underlying mechanisms involved, an integrated experimental and computational approach is ideally needed. This review discusses the state of our knowledge on cell-hydrogel interactions, with a focus on mechanics and transport, and in this context, highlights recent advancements in experiments, mathematical and computational modeling. The review begins with a background on the thermodynamics and physics fundamentals that govern hydrogel mechanics and transport. The review focuses on two main classes of hydrogels, described as semiflexible polymer networks that represent physically cross-linked fibrous hydrogels and flexible polymer networks representing the chemically cross-linked synthetic and natural hydrogels. In this review, we highlight five main cell-hydrogel interactions that involve key cellular functions related to communication, mechanosensing, migration, growth, and tissue deposition and elaboration. For each of these cellular functions, recent experiments and the most up to date modeling strategies are discussed and then followed by a summary of how to tune hydrogel properties to achieve a desired functional cellular outcome. We conclude with a summary linking these advancements and make the case for the need to integrate experiments and modeling to advance our fundamental understanding of cell-matrix interactions that will ultimately help identify new therapeutic approaches and enable successful tissue engineering.
Collapse
Affiliation(s)
- Franck J Vernerey
- Department of Mechanical Engineering, University of Colorado at Boulder, 1111 Engineering Drive, Boulder, Colorado 80309-0428, United States.,Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States
| | - Shankar Lalitha Sridhar
- Department of Mechanical Engineering, University of Colorado at Boulder, 1111 Engineering Drive, Boulder, Colorado 80309-0428, United States
| | - Archish Muralidharan
- Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States
| | - Stephanie J Bryant
- Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States.,Department of Chemical and Biological Engineering, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309-0596, United States.,BioFrontiers Institute, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309-0596, United States
| |
Collapse
|
45
|
Niu Y, Stadler FJ, Yang X, Deng F, Liu G, Xia H. HA-coated collagen nanofibers for urethral regeneration via in situ polarization of M2 macrophages. J Nanobiotechnology 2021; 19:283. [PMID: 34551762 PMCID: PMC8456673 DOI: 10.1186/s12951-021-01000-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/15/2021] [Indexed: 12/16/2022] Open
Abstract
In situ tissue engineering utilizes the regenerative potential of the human body to control cell function for tissue regeneration and has shown considerable prospect in urology. However, many problems are still to be understood, especially the interactions between scaffolds and host macrophages at the wound site and how these interactions direct tissue integration and regeneration. This study was designed to evaluate the efficacy of hyaluronic acid (HA) functionalized collagen nanofibers in modulating the pro-healing phenotype expression of macrophages for urethral regeneration. Tubular HA-collagen nanofibers with HA-coating were prepared by coaxial electrospinning. The formation of a thin HA-coating atop each collagen nanofiber endowed its nanofibrous mats with higher anisotropic wettability and mechanical softness. The macrophages growing on the surface of HA-collagen nanofibers showed an elongated shape, while collagen nanofibers' surface exhibited a pancake shape. Immunofluorescence and ELISA analysis showed that elongation could promote the expression of M2 phenotype marker and reduce the secretion of inflammatory cytokines. In vivo experiments showed that tubular HA-collagen nanofibers significantly facilitate male puppy urethral regeneration after injury. In the regenerated urethra bridged by tubular HA-collagen nanofibers, anti-inflammatory M2 macrophages are recruited to the surface of the scaffold, which can promote angiogenesis and endogenous urothelial progenitor cell proliferation.
Collapse
Affiliation(s)
- Yuqing Niu
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Florian J Stadler
- Nanshan District Key Lab for Biopolymers and Safety Evaluation, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518055, Guangdong, China
| | - Xu Yang
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Fuming Deng
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Guochang Liu
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Huimin Xia
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China.
| |
Collapse
|
46
|
Integrin and syndecan binding peptide-conjugated alginate hydrogel for modulation of nucleus pulposus cell phenotype. Biomaterials 2021; 277:121113. [PMID: 34492582 DOI: 10.1016/j.biomaterials.2021.121113] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/17/2021] [Accepted: 08/27/2021] [Indexed: 01/05/2023]
Abstract
Biomaterial based strategies have been widely explored to preserve and restore the juvenile phenotype of cells of the nucleus pulposus (NP) in degenerated intervertebral discs (IVD). With aging and maturation, NP cells lose their ability to produce necessary extracellular matrix and proteoglycans, accelerating disc degeneration. Previous studies have shown that integrin or syndecan binding peptide motifs from laminin can induce NP cells from degenerative human discs to re-express juvenile NP-specific cell phenotype and biosynthetic activity. Here, we engineered alginate hydrogels to present integrin- and syndecan-binding peptides alone or in combination (cyclic RGD and AG73, respectively) to introduce bioactive features into the alginate gels. We demonstrated human NP cells cultured upon and within alginate hydrogels presented with cRGD and AG73 peptides exhibited higher cell viability, biosynthetic activity, and NP-specific protein expression over alginate alone. Moreover, the combination of the two peptide motifs elicited markers of the NP-specific cell phenotype, including N-Cadherin, despite differences in cell morphology and multicellular cluster formation between 2D and 3D cultures. These results represent a promising step toward understanding how distinct adhesive peptides can be combined to guide NP cell fate. In the future, these insights may be useful to rationally design hydrogels for NP cell-transplantation based therapies for IVD degeneration.
Collapse
|
47
|
Alaohali A, Salzlechner C, Zaugg LK, Suzano F, Martinez A, Gentleman E, Sharpe PT. GSK3 Inhibitor-Induced Dentinogenesis Using a Hydrogel. J Dent Res 2021; 101:46-53. [PMID: 34152872 PMCID: PMC8721547 DOI: 10.1177/00220345211020652] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Small-molecule drugs targeting glycogen synthase kinase 3 (GSK3) as inhibitors of the protein kinase activity are able to stimulate reparative dentine formation. To develop this approach into a viable clinical treatment for exposed pulp lesions, we synthesized a novel, small-molecule noncompetitive adenosine triphosphate (ATP) drug that can be incorporated into a biodegradable hydrogel for placement by syringe into the tooth. This new drug, named NP928, belongs to the thiadiazolidinone (TDZD) family and has equivalent activity to similar drugs of this family such as tideglusib. However, NP928 is more water soluble than other TDZD drugs, making it more suitable for direct delivery into pulp lesions. We have previously reported that biodegradable marine collagen sponges can successfully deliver TDZD drugs to pulp lesions, but this involves in-theater preparation of the material, which is not ideal in a clinical context. To improve surgical handling and delivery, here we incorporated NP928 into a specifically tailored hydrogel that can be placed by syringe into a damaged tooth. This hydrogel is based on biodegradable hyaluronic acid and can be gelled in situ upon dental blue light exposure, similarly to other common dental materials. NP928 released from hyaluronic acid-based hydrogels upregulated Wnt/β-catenin activity in pulp stem cells and fostered reparative dentine formation compared to marine collagen sponges delivering equivalent concentrations of NP928. This drug-hydrogel combination has the potential to be rapidly developed into a therapeutic procedure that is amenable to general dental practice.
Collapse
Affiliation(s)
- A Alaohali
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK.,Department of Dental and Oral Health, Prince Sultan Military College of Health Sciences, Dammam, Saudi Arabia
| | - C Salzlechner
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
| | - L K Zaugg
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK.,Department of Reconstructive Dentistry, University Center for Dental Medicine, University of Basel, Basel, Switzerland
| | - F Suzano
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
| | - A Martinez
- Centro de Investigaciones Biologicas-CSIC, Madrid, Spain
| | - E Gentleman
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
| | - P T Sharpe
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
| |
Collapse
|
48
|
Lust ST, Hoogland D, Norman MDA, Kerins C, Omar J, Jowett GM, Yu TTL, Yan Z, Xu JZ, Marciano D, da Silva RMP, Dreiss CA, Lamata P, Shipley RJ, Gentleman E. Selectively Cross-Linked Tetra-PEG Hydrogels Provide Control over Mechanical Strength with Minimal Impact on Diffusivity. ACS Biomater Sci Eng 2021; 7:4293-4304. [PMID: 34151570 PMCID: PMC7611660 DOI: 10.1021/acsbiomaterials.0c01723] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
![]()
Synthetic hydrogels
formed from poly(ethylene glycol) (PEG) are
widely used to study how cells interact with their extracellular matrix.
These in vivo-like 3D environments provide a basis
for tissue engineering and cell therapies but also for research into
fundamental biological questions and disease modeling. The physical
properties of PEG hydrogels can be modulated to provide mechanical
cues to encapsulated cells; however, the impact of changing hydrogel
stiffness on the diffusivity of solutes to and from encapsulated cells
has received only limited attention. This is particularly true in
selectively cross-linked “tetra-PEG” hydrogels, whose
design limits network inhomogeneities. Here, we used a combination
of theoretical calculations, predictive modeling, and experimental
measurements of hydrogel swelling, rheological behavior, and diffusion
kinetics to characterize tetra-PEG hydrogels’ permissiveness
to the diffusion of molecules of biologically relevant size as we
changed polymer concentration, and thus hydrogel mechanical strength.
Our models predict that hydrogel mesh size has little effect on the
diffusivity of model molecules and instead predicts that diffusion
rates are more highly dependent on solute size. Indeed, our model
predicts that changes in hydrogel mesh size only begin to have a non-negligible
impact on the concentration of a solute that diffuses out of hydrogels
for the smallest mesh sizes and largest diffusing solutes. Experimental
measurements characterizing the diffusion of fluorescein isothiocyanate
(FITC)-labeled dextran molecules of known size aligned well with modeling
predictions and suggest that doubling the polymer concentration from
2.5% (w/v) to 5% produces stiffer gels with faster gelling kinetics
without affecting the diffusivity of solutes of biologically relevant
size but that 10% hydrogels can slow their diffusion. Our findings
provide confidence that the stiffness of tetra-PEG hydrogels can be
modulated over a physiological range without significantly impacting
the transport rates of solutes to and from encapsulated cells.
Collapse
Affiliation(s)
- Suzette T Lust
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, United Kingdom.,School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, United Kingdom
| | - Dominique Hoogland
- Department of Chemistry, King's College London, London SE1 1DB, United Kingdom
| | - Michael D A Norman
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, United Kingdom
| | - Caoimhe Kerins
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, United Kingdom
| | - Jasmin Omar
- Institute of Pharmaceutical Science, King's College London, Franklin-Wilkins Building, London SE1 9NH, United Kingdom
| | - Geraldine M Jowett
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, United Kingdom
| | - Tracy T L Yu
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, United Kingdom
| | - Ziqian Yan
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, United Kingdom
| | - Jessie Z Xu
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, United Kingdom
| | - Daniele Marciano
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, United Kingdom
| | - Ricardo M P da Silva
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, United Kingdom
| | - Cécile A Dreiss
- Institute of Pharmaceutical Science, King's College London, Franklin-Wilkins Building, London SE1 9NH, United Kingdom
| | - Pablo Lamata
- School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, United Kingdom
| | - Rebecca J Shipley
- Institute of Healthcare Engineering and Department of Mechanical Engineering, University College London, London WC1E 7JE, United Kingdom
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, United Kingdom
| |
Collapse
|
49
|
Hajiali H, Ouyang L, Llopis-Hernandez V, Dobre O, Rose FRAJ. Review of emerging nanotechnology in bone regeneration: progress, challenges, and perspectives. NANOSCALE 2021; 13:10266-10280. [PMID: 34085085 DOI: 10.1039/d1nr01371h] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
The application of nanotechnology to regenerative medicine has increased over recent decades. The development of materials that can influence biology at the nanoscale has gained interest as our understanding of the interactions between cells and biomaterials at the nanoscale has grown. Materials that are either nanostructured or influence the nanostructure of the cellular microenvironment have been developed and shown to have advantages over their microscale counterparts. There are several reviews which have been published that discuss how nanomaterials have been used in regenerative medicine, particularly in bone regeneration. Most of these studies have explored this concept in specific areas, such as the application of glass-based nanocomposites, nanotechnology for targeted drug delivery to stimulate bone repair, and the progress in nanotechnology for the treatment of osteoporosis. In this review paper, the impact of nanotechnology in biomaterials development for bone regeneration will be discussed highlighting specifically, nanostructured materials that influence mechanical properties, biocompatibility, and osteoinductivity.
Collapse
Affiliation(s)
- Hadi Hajiali
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, University Park, University of Nottingham, NG7 2RD, UK.
| | - Liliang Ouyang
- Department of Materials, Imperial College London, London, SW7 2AZ, UK and Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | | | - Oana Dobre
- Centre for the Cellular Microenvironment, University of Glasgow, Glasgow, G12 8LT, UK
| | - Felicity R A J Rose
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, University Park, University of Nottingham, NG7 2RD, UK.
| |
Collapse
|
50
|
Yang B, Wei K, Loebel C, Zhang K, Feng Q, Li R, Wong SHD, Xu X, Lau C, Chen X, Zhao P, Yin C, Burdick JA, Wang Y, Bian L. Enhanced mechanosensing of cells in synthetic 3D matrix with controlled biophysical dynamics. Nat Commun 2021; 12:3514. [PMID: 34112772 PMCID: PMC8192531 DOI: 10.1038/s41467-021-23120-0] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 03/19/2021] [Indexed: 01/08/2023] Open
Abstract
3D culture of cells in designer biomaterial matrices provides a biomimetic cellular microenvironment and can yield critical insights into cellular behaviours not available from conventional 2D cultures. Hydrogels with dynamic properties, achieved by incorporating either degradable structural components or reversible dynamic crosslinks, enable efficient cell adaptation of the matrix and support associated cellular functions. Herein we demonstrate that given similar equilibrium binding constants, hydrogels containing dynamic crosslinks with a large dissociation rate constant enable cell force-induced network reorganization, which results in rapid stellate spreading, assembly, mechanosensing, and differentiation of encapsulated stem cells when compared to similar hydrogels containing dynamic crosslinks with a low dissociation rate constant. Furthermore, the static and precise conjugation of cell adhesive ligands to the hydrogel subnetwork connected by such fast-dissociating crosslinks is also required for ultra-rapid stellate spreading (within 18 h post-encapsulation) and enhanced mechanosensing of stem cells in 3D. This work reveals the correlation between microscopic cell behaviours and the molecular level binding kinetics in hydrogel networks. Our findings provide valuable guidance to the design and evaluation of supramolecular biomaterials with cell-adaptable properties for studying cells in 3D cultures. 3D culture systems can provide critical insights into cellular behaviour. Here, the authors study the binding timescale of dynamic crosslinks and the conjugation stability of cell-adhesive ligands in cell–hydrogel network interactions to evaluate the impact on stem cell behaviour, mechanosensing and differentiation.
Collapse
Affiliation(s)
- Boguang Yang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Kongchang Wei
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China.,Empa, Swiss Federal Laboratories for Materials Science and Technology, Laboratory for Biomimetic Membranes and Textiles, St. Gallen, Switzerland
| | - Claudia Loebel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Kunyu Zhang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Qian Feng
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China.,Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, China
| | - Rui Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Siu Hong Dexter Wong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China.,Department of Biomedical Engineering, The Hong Kong Polytechnic University, HongKong, China
| | - Xiayi Xu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Chunhon Lau
- Department of Physics, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoyu Chen
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China.,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Pengchao Zhao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Chao Yin
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Yi Wang
- Department of Physics, The Chinese University of Hong Kong, Hong Kong, China.
| | - Liming Bian
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China. .,Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China. .,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang, China.
| |
Collapse
|