1
|
Jiang T, Feng Y, Gao C, Jiang J, Chen B, Liu S, Du D, Ding M, Rong J, Liao Z, Li W, Wilson DA, Tu Y, Song S, Wang Y, Peng F. Microenvironment Mechanical Torque from ZnFe 2O 4 (ZFO) Micromotors Inhibiting Tumor Migration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2409769. [PMID: 39838745 DOI: 10.1002/adma.202409769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 01/01/2025] [Indexed: 01/23/2025]
Abstract
Mechanical force attracts booming attention with the potential to tune the tumor cell behavior, especially in cell migration. However, the current approach for introducing mechanical input is difficult to apply in vivo. How the mechanical force affects cell behavior in situ also remains unclear. In this work, an intelligent miniaturized platform is constructed with magnetic ZnFe2O4 (ZFO) micromotors. The wireless ZFO can self-assemble in situ and rotate to generate mechanical torque of biologically relevant piconewton-scale at the target tumor site. It is observed unexpectedly that enhanced in situ mechanical rotating torque from ZFO micromotors and the active fluid inhibit the migration of highly invasive A549 tumor cells. The down-regulation of the Piezo1 channel and the suppressed signaling of ROCK1 in mechano-adaptive tumor cells is found to be related to the inhibition effect. With effectiveness confirmed with the zebrafish xenograft model, this platform provides a valuable toolkit for mechanobiology and force-associated non-invasive tumor therapy.
Collapse
Affiliation(s)
- Tingting Jiang
- The Key Lab of Low-carbon Chemistry, School of Chemical Engineering and Technology, Sun Yat-sen University, Guangzhou, 510275, China
| | - Ye Feng
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Chao Gao
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Jiamiao Jiang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Bin Chen
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Suyi Liu
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Dailing Du
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Miaomiao Ding
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Jinghui Rong
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Zongzhen Liao
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory for Aquatic Economic Animals, Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Wensheng Li
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory for Aquatic Economic Animals, Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Daniela A Wilson
- Institute for Molecules and Materials, Radboud University, Nijmegen, 6525 AJ, The Netherlands
| | - Yingfeng Tu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shuqin Song
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yi Wang
- The Key Lab of Low-carbon Chemistry, School of Chemical Engineering and Technology, Sun Yat-sen University, Guangzhou, 510275, China
| | - Fei Peng
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| |
Collapse
|
2
|
Iriondo C, Koornneef S, Skarp KP, Buscop-van Kempen M, Boerema-de Munck A, Rottier RJ. Simple-Flow: A 3D-Printed Multiwell Flow Plate to Coculture Primary Human Lung Cells at the Air-Liquid Interface. ACS Biomater Sci Eng 2025; 11:451-462. [PMID: 39719361 PMCID: PMC11734690 DOI: 10.1021/acsbiomaterials.4c01322] [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: 07/17/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 12/26/2024]
Abstract
Immortalized epithelial cell lines and animal models have been used in fundamental and preclinical research to study pulmonary diseases. However valuable, though, these models incompletely recapitulate the in vivo human lung, which leads to low predictive outcomes in potential respiratory treatments. Advanced technology and cell culture techniques stimulate the development of improved models that more closely mimic the physiology of the human lung. Nonetheless, most of these models are technically demanding and have a low throughput and reproducibility. Here, we describe a robust fluidic device consisting of a biocompatible and customizable 3D-printed cell culture plate, the Simple-Flow, which has medium throughput, is simple to manufacture, and is easy to set up. As a proof of principle, human primary bronchial epithelial cells (hPBECs) and human pulmonary microvascular endothelial cells (hMVECs) were cocultured on the apical and basolateral sides of the inset membranes, respectively. While hPBECs were cultured at the air-liquid interface to induce mucociliary differentiation, hMVECs were exposed to flow medium for up to 2 weeks. We show the versatility of 3D-printing technology in designing in vitro models for cell culturing applications, such as pediatric lung diseases or other pulmonary disorders.
Collapse
Affiliation(s)
- Cinta Iriondo
- Department
of Pediatric Surgery, Sophia Children’s Hospital, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
- Department
of Cell Biology, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
| | - Sem Koornneef
- Department
of Pediatric Surgery, Sophia Children’s Hospital, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
- Department
of Cell Biology, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
| | - Kari-Pekka Skarp
- Department
of Pediatric Surgery, Sophia Children’s Hospital, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
- Department
of Cell Biology, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
| | - Marjon Buscop-van Kempen
- Department
of Pediatric Surgery, Sophia Children’s Hospital, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
- Department
of Cell Biology, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
| | - Anne Boerema-de Munck
- Department
of Pediatric Surgery, Sophia Children’s Hospital, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
- Department
of Cell Biology, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
| | - Robbert J. Rottier
- Department
of Pediatric Surgery, Sophia Children’s Hospital, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
- Department
of Cell Biology, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
| |
Collapse
|
3
|
Feng P, He C, Li G, Li J, Luo Y, Chen Y, Tang Y, Ma J, Ke C. A light-cured injectable composite hydrogel based on chitosan and decellularized matrix modulates stem cell aggregation behavior for accelerating cartilage defect repair. Int J Biol Macromol 2025; 295:139711. [PMID: 39798753 DOI: 10.1016/j.ijbiomac.2025.139711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Cartilage repair remains a formidable challenge because of its limited regenerative capacity. Construction of a biomimetic hydrogel matrix that can induce cell aggregation is a promising therapeutic option. Cell aggregates are more beneficial than dissociated cells for improving survival and chondrogenic differentiation, thereby facilitating cartilage repair. Herein, we report a light-cured injectable composite hydrogel with cellular aggregation properties for cartilage defect repair that combines methacrylated chitosan (CSMA) and a methacrylate-modified decellularized extracellular matrix (dECMMA). The CSMA promotes cell aggregate formation by enhancing cadherin expression. The dECMMA retains many inherent components and bioactive factors, thereby providing a more natural microenvironment for cell proliferation and differentiation. By precisely adjusting the compositions of the CSMA and dECMMA, it was possible to fine-tune their physicochemical properties and biochemical cues. The results showed that a composite hydrogel composed of 2 % (m/v) CSMA and 5 % (m/v) dECMMA exhibited good injectability, appropriate degradability and mechanical properties, and regulated the formation of bone marrow mesenchymal stem cell aggregates. Transplantation of the composite hydrogel loaded with bone marrow mesenchymal stem cells (BMSCs) into a cartilage defect model demonstrated effective hyaline cartilage repair. Thus, light-cured injectable composite hydrogels embedded with BMSCs holds clinical promise for cartilage defect repair.
Collapse
Affiliation(s)
- Peipei Feng
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo 315040, China
| | - Chaonan He
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo 315040, China
| | - Guanrong Li
- Research Institute of Smart Medicine and Biological Engineering, Ningbo University, Ningbo 315211, China
| | - Jin Li
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo 315040, China
| | - Yang Luo
- Research Institute of Smart Medicine and Biological Engineering, Ningbo University, Ningbo 315211, China
| | - Yaoqi Chen
- Research Institute of Smart Medicine and Biological Engineering, Ningbo University, Ningbo 315211, China
| | - Yun Tang
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo 315040, China
| | - Jingyun Ma
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo 315040, China.
| | - Chunhai Ke
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo 315040, China.
| |
Collapse
|
4
|
Keijzer KAE, Tsingos E, Merks RMH. How cells align to structured collagen fibrils: a hybrid cellular Potts and molecular dynamics model with dynamic mechanosensitive focal adhesions. Front Cell Dev Biol 2025; 12:1462277. [PMID: 39834385 PMCID: PMC11743931 DOI: 10.3389/fcell.2024.1462277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
Many mammalian cells, including endothelial cells and fibroblasts, align and elongate along the orientation of extracellular matrix (ECM) fibers in a gel when cultured in vitro. During cell elongation, clusters of focal adhesions (FAs) form near the poles of the elongating cells. FAs are mechanosensitive clusters of adhesions that grow under mechanical tension exerted by the cells' pulling on the ECM and shrink when the tension is released. In this study, we use mathematical modeling to study the hypothesis that mechanical reciprocity between cells and the ECM is sufficient for directing cell shape changes and orientation. We show that FAs are preferentially stabilized along the orientation of ECM fibers, where cells can generate higher tension than in directions perpendicular to the ECM fibers. We present a hybrid computational model coupling three mathematical approaches: first, the cellular Potts model (CPM) describes an individual contractile cell; second, molecular dynamics (MD) represent the ECM as a network of cross-linked, deformable fibers; third, a set of ordinary differential equations (ODEs) describes the dynamics of the cell's FAs, in terms of a balance between assembly and a mechanoresponsive disassembly. The resulting computational model shows that mechanical reciprocity suffices for stiffness-dependent cell spreading, local ECM remodeling, and ECM-alignment-dependent cell elongation. These combined effects are sufficient to explain how cell morphology is influenced by the local ECM structure and mechanics.
Collapse
Affiliation(s)
- Koen A. E. Keijzer
- Mathematical Institute, Faculty of Science, Leiden University, Leiden, Netherlands
| | - Erika Tsingos
- Mathematical Institute, Faculty of Science, Leiden University, Leiden, Netherlands
- Institute of Biology Leiden, Faculty of Science, Leiden University, Leiden, Netherlands
| | - Roeland M. H. Merks
- Mathematical Institute, Faculty of Science, Leiden University, Leiden, Netherlands
- Institute of Biology Leiden, Faculty of Science, Leiden University, Leiden, Netherlands
| |
Collapse
|
5
|
da Silva RLCG, Bezjak D, Corrales TP, Kappl M, Petri DFS. Chitosan/vanillin/polydimethylsiloxane scaffolds with tunable stiffness for muscle cell proliferation. Int J Biol Macromol 2025; 286:138445. [PMID: 39645124 DOI: 10.1016/j.ijbiomac.2024.138445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 11/18/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
The mechanical properties of scaffolds can significantly influence cell behavior. We propose a methodology for producing chitosan and vanillin-crosslinked chitosan films with tunable mechanical properties to be applied as scaffolds for C2C12 myoblasts. In this approach, aqueous polydimethylsiloxane (PDMS) elastomeric dispersions were prepared using polysorbate 20 as emulsifier. These dispersions were then cured and incorporated into chitosan or vanillin-crosslinked chitosan polymeric dispersions at two different volume fractions (1 % and 10 %), followed by casting into films. Atomic force microscopy in force spectroscopy mode was used to characterize the mechanical properties of the swollen systems in PBS buffer. The mechanical properties of the chitosan and vanillin-crosslinked chitosan scaffolds were modulated by the incorporation of the elastomer. The elastic modulus (E) of chitosan-based scaffolds varied from 60 to 200 kPa, while for vanillin-based scaffolds, it ranged from 200 to 600 kPa with the addition of PDMS elastomers. A general trend observed was that the softest scaffolds exhibited the highest swelling degree and the lowest gel content. After 24 h, good cell viability was observed for chitosan and chitosan-PDMS scaffolds, whereas vanillin-based scaffolds showed borderline cytotoxicity (∼70 %). C2C12 cells demonstrated good adhesion on scaffolds with E values ranging from 114 to 568 kPa.
Collapse
Affiliation(s)
| | - Dragica Bezjak
- Centro de Biotecnología, Universidad Técnica Federico Santa María, Valparaíso, Chile
| | - Tomas P Corrales
- Centro de Biotecnología, Universidad Técnica Federico Santa María, Valparaíso, Chile; Departamento de Física, Universidad Técnica Federico Santa María, Valparaíso, Chile; Millenium Nucleus in NanoBioPhysics (NNBP), Valparaíso, Chile
| | - Michael Kappl
- Max Planck Institute for Polymer Research, Mainz, Germany.
| | - Denise F S Petri
- Fundamental Chemistry Department, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil.
| |
Collapse
|
6
|
Braxton T, Lim K, Alcala-Orozco C, Joukhdar H, Rnjak-Kovacina J, Iqbal N, Woodfield T, Wood D, Brockett C, Yang X. Mechanical and Physical Characterization of a Biphasic 3D Printed Silk-Infilled Scaffold for Osteochondral Tissue Engineering. ACS Biomater Sci Eng 2024; 10:7606-7618. [PMID: 39589862 PMCID: PMC11632666 DOI: 10.1021/acsbiomaterials.4c01865] [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: 10/07/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024]
Abstract
Osteochondral tissue damage is a serious concern, with even minor cartilage damage dramatically increasing an individual's risk of osteoarthritis. Therefore, there is a need for an early intervention for osteochondral tissue regeneration. 3D printing is an exciting method for developing novel scaffolds, especially for creating biological scaffolds for osteochondral tissue engineering. However, many 3D printing techniques rely on creating a lattice structure, which often demonstrates poor cell bridging between filaments due to its large pore size, reducing regenerative speed and capacity. To tackle this issue, a novel biphasic scaffold was developed by a combination of 3D printed poly(ethylene glycol)-terephthalate-poly(butylene-terephthalate) (PEGT/PBT) lattice infilled with a porous silk scaffold (derived from Bombyx mori silk fibroin) to make up a bone phase, which continued to a seamless silk top layer, representing a cartilage phase. Compression testing showed scaffolds had Young's modulus, ultimate compressive strength, and fatigue resistance that would allow for their theoretical survival during implantation and joint articulation without stress-shielding mechanosensitive cells. Fluorescent microscopy showed biphasic scaffolds could support the attachment and spreading of human mesenchymal stem cells from bone marrow (hMSC-BM). These promising results highlight the potential utilization of this novel scaffold for osteochondral tissue regeneration as well as highlighting the potential of infilling silk materials within 3D printed scaffolds to further increase their versatility.
Collapse
Affiliation(s)
- T. Braxton
- School
of Mechanical Engineering, University of
Leeds, Leeds LS2 9JT, U.K.
| | - K. Lim
- CReaTE
Group, Department of Orthopaedic Surgery, University of Otago Christchurch, Christchurch 8140, New Zealand
| | - C. Alcala-Orozco
- CReaTE
Group, Department of Orthopaedic Surgery, University of Otago Christchurch, Christchurch 8140, New Zealand
| | - H. Joukhdar
- Graduate
School of Biomedical Engineering, UNSW Sydney, Sydney, New South Wales 2052, Australia
| | - J. Rnjak-Kovacina
- Graduate
School of Biomedical Engineering, UNSW Sydney, Sydney, New South Wales 2052, Australia
| | - N. Iqbal
- Chemical
and Process Engineering, University of Leeds, Leeds LS2 9JT, U.K.
| | - T. Woodfield
- CReaTE
Group, Department of Orthopaedic Surgery, University of Otago Christchurch, Christchurch 8140, New Zealand
| | - D. Wood
- Biomaterials
and Tissue Engineering Group, Department of Oral Biology, University of Leeds, WTBB, St. James’s University
Hospital, Leeds LS9 7TF, U.K.
| | - C. Brockett
- School
of Mechanical Engineering, University of
Leeds, Leeds LS2 9JT, U.K.
| | - X.B. Yang
- Biomaterials
and Tissue Engineering Group, Department of Oral Biology, University of Leeds, WTBB, St. James’s University
Hospital, Leeds LS9 7TF, U.K.
| |
Collapse
|
7
|
Li CX, Zhao ZX, Su DB, Yin DC, Ye YJ. In vitro regulation of collective cell migration: Understanding the role of physical and chemical microenvironments. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 195:23-40. [PMID: 39612952 DOI: 10.1016/j.pbiomolbio.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/22/2024] [Accepted: 11/24/2024] [Indexed: 12/01/2024]
Abstract
Collective cell migration is the primary mode of cellular movement during embryonic morphogenesis, tissue repair and regeneration, and cancer invasion. Distinct from single-cell migration, collective cell migration involves complex intercellular signaling cascades and force transmission. Consequently, cell collectives exhibit intricate and diverse migration patterns under the influence of the microenvironment in vivo. Investigating the patterns and mechanisms of collective cell migration within complex environmental factors in vitro is essential for elucidating collective cell migration in vivo. This review elucidates the influence of physical and chemical factors in vitro microenvironment on the migration patterns and efficiency of cell collectives, thereby enhancing our comprehension of the phenomenon. Furthermore, we concisely present the effects of characteristic properties of common biomaterials on collective cell migration during tissue repair and regeneration, as well as the features and applications of tumor models of different dimensions (2D substrate or 3D substrate) in vitro. Finally, we highlight the challenges facing the research of collective cell migration behaviors in vitro microenvironment and propose that modulating collective cell migration may represent a potential strategy to promote tissue repair and regeneration and to control tumor invasion and metastasis.
Collapse
Affiliation(s)
- Chang-Xing Li
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Zi-Xu Zhao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Dan-Bo Su
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Da-Chuan Yin
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Ya-Jing Ye
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China.
| |
Collapse
|
8
|
Jang Y, Lee D, Oh J. Fast Autograft Generation Using Transferable 3D Keratinocyte Cell Sheet on PEDOT:PSS Composite PDMS Membrane for Enhancing Wound Healing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2406529. [PMID: 39588867 DOI: 10.1002/smll.202406529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/20/2024] [Indexed: 11/27/2024]
Abstract
The application of cell sheet technology for wound healing preserves dense cell tissue and the natural extracellular matrix (ECM), contributing to disease prevention. Despite the effectiveness of autologous and allograft cell sheets for wound healing, conventional cell sheets, although stable, may experience necrosis in their middle layers due to a lack of nutrients or oxygen. To address these issues, a novel approach is proposed to create cell sheets using mechanical and electrical stimulation. This method not only facilitates the transfer of cell sheets but also enhances cell bioactivity. The performance of the proposed membrane, with a mechanically controlled microstructure under electrical stimulation, is validated in both in vitro and in vivo models. The micro-structured membrane allows for diverse electrical stimulation compared to flat membranes, which accelerates the detachment of cell sheets and promotes angiogenesis and re-epithelialization. These findings indicate that the innovative cell sheet technology could significantly enhance rapid wound healing in regenerative medicine.
Collapse
Affiliation(s)
- Yeongseok Jang
- Department of Mechanical Design Engineering, Jeonbuk National University, Jeonju-si, Jeollabuk-do, 54896, Republic of Korea
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Dongwon Lee
- Department of Polymer Nano Engineering, Jeonbuk National University, Jeonju-si, Jeollabuk-do, 54896, Republic of Korea
| | - Jonghyun Oh
- Department of Nano-Bio Mechanical System Engineering, Jeonbuk National University, Jeonju-si, Jeollabuk-do, 54896, Republic of Korea
| |
Collapse
|
9
|
Liu X, Zhou C, Xie Q, Xia L, Liu L, Bao W, Lin H, Xiong X, Zhang H, Zheng Z, Zhao J, Liang W. Recent advances in layer-by-layer assembly scaffolds for co-delivery of bioactive molecules for bone regeneration: an updated review. J Transl Med 2024; 22:1001. [PMID: 39501263 PMCID: PMC11539823 DOI: 10.1186/s12967-024-05809-0] [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: 06/07/2024] [Accepted: 10/25/2024] [Indexed: 11/08/2024] Open
Abstract
Orthopedic implants have faced challenges in treating bone defects due to various factors, including inadequate osseointegration, oxidative stress, bacterial infection, immunological rejection, and poor individualized treatment. These challenges profoundly affect both the results of treatment and patients' daily lives. There is great promise for the layer-by-layer (LbL) assembly method in tissue engineering. The method primarily relies on electrostatic attraction and entails the consecutive deposition of electrolyte complexes with opposite charges onto a substrate, leading to the formation of homogeneous single layers that can be quickly deposited to produce nanolayer films. LbL has attracted considerable interest as a coating technology because of its ease of production, cost-effectiveness, and capability to apply diverse biomaterial coatings without compromising the primary bio-functional properties of the substrate materials. This review will look into the fundamentals and evolution of LbL in orthopedics, provide an analysis of the chemical strategy used to prepare bone implants with LbL and introduce the application of LbL bone implants in orthopedics over recent years. Among the many potential uses of LbL, such as the implementation of sustained-release and programmed drug delivery, which in turn promotes the osseointegration and the development of new blood vessels, as well as antibacterial, antioxidant, and other similar applications. In addition, we offer a thorough examination of cell behavior and biomaterial interaction to facilitate the advancement of next-generation LbL films for tissue engineering.
Collapse
Affiliation(s)
- Xiankun Liu
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, 355 Xinqiao Road, Dinghai District, Zhoushan, 316000, Zhejiang, People's Republic of China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua Hospital, Zhoushan, 316000, Zhejiang, China
| | - Qiong Xie
- Department of Orthopedics, Zhoushan Guanghua Hospital, Zhoushan, 316000, Zhejiang, China
| | - Linying Xia
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Lu Liu
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Wenwen Bao
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Hongming Lin
- Department of Orthopedics, Zhoushan Guanghua Hospital, Zhoushan, 316000, Zhejiang, China
| | - Xiaochun Xiong
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, 355 Xinqiao Road, Dinghai District, Zhoushan, 316000, Zhejiang, People's Republic of China
| | - Hao Zhang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, 355 Xinqiao Road, Dinghai District, Zhoushan, 316000, Zhejiang, People's Republic of China
| | - Zeping Zheng
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, 355 Xinqiao Road, Dinghai District, Zhoushan, 316000, Zhejiang, People's Republic of China
| | - Jiayi Zhao
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, 355 Xinqiao Road, Dinghai District, Zhoushan, 316000, Zhejiang, People's Republic of China.
| | - Wenqing Liang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, 355 Xinqiao Road, Dinghai District, Zhoushan, 316000, Zhejiang, People's Republic of China.
| |
Collapse
|
10
|
Wang X, Chen S, Chen X, Wu J, Huang Z, Wang J, Chen F, Liu C. Biomimetic multi-channel nerve conduits with micro/nanostructures for rapid nerve repair. Bioact Mater 2024; 41:577-596. [PMID: 39257673 PMCID: PMC11384339 DOI: 10.1016/j.bioactmat.2024.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/13/2024] [Accepted: 07/13/2024] [Indexed: 09/12/2024] Open
Abstract
Peripheral nervous system (PNS) injuries often lead to significant sensory and motor impairments. Traditional artificial nerve conduits, lacking anisotropic structures, have been associated with prolonged repair time and failures in nerve regeneration. This study aimed to address these challenges by developing a novel approach for rapid repair of peripheral nerve injuries (PNI). A 3D oriented fibers scaffold featuring distinct radial (RFs) and longitudinal (LFs) fibers orientations was engineered using coaxial electrospinning and gas directional foaming techniques. This scaffold was then integrated with a shape memory conduit to form a directional multi-channel nerve conduit with micro/nanostructures. The results revealed that the grooved surface of the fibers significantly improved cellular directional guidance, effectively facilitating the migration of SCs from the periphery towards the center and from the base to the apex of the scaffold. In a rat model with a 10 mm nerve defect, the ND-PLATMC/LF ND-PCL scaffold significantly enhanced nerve regeneration and motor function recovery within 4 weeks. These results suggest the potential of this innovative scaffold for efficient repair of the nerve injuries.
Collapse
Affiliation(s)
- Xinqing Wang
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Shuo Chen
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Xiaolei Chen
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Juan Wu
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Zhenhua Huang
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Jing Wang
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Fangping Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| |
Collapse
|
11
|
Luo Y, Hu Z, Ni R, Xu R, Zhao J, Feng P, Zhu T, Chen Y, Yao J, Yao Y, Yang L, Zhang H, Zhu Y. Fabrication of 3D Biomimetic Smooth Muscle Using Magnetic Induction and Bioprinting for Tissue Regeneration. Biomater Res 2024; 28:0076. [PMID: 39253032 PMCID: PMC11382380 DOI: 10.34133/bmr.0076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/18/2024] [Accepted: 08/10/2024] [Indexed: 09/11/2024] Open
Abstract
Smooth muscles play a vital role in peristalsis, tissue constriction, and relaxation but lack adequate self-repair capability for addressing extensive muscle defects. Engineering scaffolds have been broadly proposed to repair the muscle tissue. However, efforts to date have shown that those engineered scaffolds focus on cell alignment in 2-dimension (2D) and fail to direct muscle cells to align in 3D area, which is irresolvable to remodel the muscle architecture and restore the muscle functions like contraction and relaxation. Herein, we introduced an iron oxide (Fe3O4) filament-embedded gelatin (Gel)-silk fibroin composite hydrogel in which the oriented Fe3O4 self-assembled and functioned as micro/nanoscale geometric cues to induce cell alignment growth. The hydrogel scaffold can be designed to fabricate aligned or anisotropic muscle by combining embedded 3D bioprinting with magnetic induction to accommodate special architectures of muscular tissues in the body. Particularly, the bioprinted muscle-like matrices effectively promote the self-organization of smooth muscle cells (SMCs) and the directional differentiation of bone marrow mesenchymal stem cells (BMSCs) into SMCs. This biomimetic muscle accelerated tissue regeneration, enhancing intercellular connectivity within the muscular tissue, and the deposition of fibronectin and collagen I. This work provides a novel approach for constructing engineered biomimetic muscles, holding significant promise for clinical treatment of muscle-related diseases in the future.
Collapse
Affiliation(s)
- Yang Luo
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Zeming Hu
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Renhao Ni
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Rong Xu
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Jianmin Zhao
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Peipei Feng
- Ningbo Institute of Innovation for Combined Medicine and Engineering, The Affiliated Lihuili Hospital of Ningbo University, Ningbo 315046, China
| | - Tong Zhu
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Yaoqi Chen
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Jie Yao
- The First Affiliated Hospital of Ningbo University, Ningbo 315010, China
- Research Institute of Smart Medicine and Biological Engineering, Ningbo University, Ningbo 315211, China
| | - Yudong Yao
- Health Science Center, Ningbo University, Ningbo 315211, China
- Research Institute of Smart Medicine and Biological Engineering, Ningbo University, Ningbo 315211, China
| | - Lu Yang
- The First Affiliated Hospital of Ningbo University, Ningbo 315010, China
| | - Hua Zhang
- Health Science Center, Ningbo University, Ningbo 315211, China
- Research Institute of Smart Medicine and Biological Engineering, Ningbo University, Ningbo 315211, China
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200438, China
| | - Yabin Zhu
- Health Science Center, Ningbo University, Ningbo 315211, China
| |
Collapse
|
12
|
Kim D, Lee MJ, Arai Y, Ahn J, Lee GW, Lee SH. Ultrasound-triggered three dimensional hyaluronic acid hydrogel promotes in vitro and in vivo reprogramming into induced pluripotent stem cells. Bioact Mater 2024; 38:331-345. [PMID: 38764447 PMCID: PMC11101682 DOI: 10.1016/j.bioactmat.2024.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/12/2024] [Accepted: 05/05/2024] [Indexed: 05/21/2024] Open
Abstract
Cellular reprogramming technologies have been developed with different physicochemical factors to improve the reprogramming efficiencies of induced pluripotent stem cells (iPSCs). Ultrasound is a clinically applied noncontact biophysical factor known for regulating various cellular behaviors but remains uninvestigated for cellular reprogramming. Here, we present a new reprogramming strategy using low-intensity ultrasound (LIUS) to improve cellular reprogramming of iPSCs in vitro and in vivo. Under 3D microenvironment conditions, increased LIUS stimulation shows enhanced cellular reprogramming of the iPSCs. The cellular reprogramming process facilitated by LIUS is accompanied by increased mesenchymal to epithelial transition and histone modification. LIUS stimulation transiently modulates the cytoskeletal rearrangement, along with increased membrane fluidity and mobility to increase HA/CD44 interactions. Furthermore, LIUS stimulation with HA hydrogel can be utilized in application of both human cells and in vivo environment, for enhanced reprogrammed cells into iPSCs. Thus, LIUS stimulation with a combinatorial 3D microenvironment system can improve cellular reprogramming in vitro and in vivo environments, which can be applied in various biomedical fields.
Collapse
Affiliation(s)
| | | | - Yoshie Arai
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620, Seoul, South Korea
| | - Jinsung Ahn
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620, Seoul, South Korea
| | - Gun Woo Lee
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620, Seoul, South Korea
| | - Soo-Hong Lee
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620, Seoul, South Korea
| |
Collapse
|
13
|
Xie B, Xie H. Application of stimuli-responsive hydrogel in brain disease treatment. Front Bioeng Biotechnol 2024; 12:1450267. [PMID: 39091971 PMCID: PMC11291207 DOI: 10.3389/fbioe.2024.1450267] [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: 06/17/2024] [Accepted: 06/26/2024] [Indexed: 08/04/2024] Open
Abstract
Treating brain diseases presents significant challenges due to neuronal degeneration, inflammation, and the intricate nature of the brain. Stimuli-responsive hydrogels, designed to closely resemble the brain's extracellular matrix, have emerged as promising candidates for controlled drug delivery and tissue engineering. These hydrogels have the unique ability to encapsulate therapeutic agents and release them in a controlled manner when triggered by environmental stimuli. This property makes them particularly suitable for delivering drugs precisely to targeted areas of the brain, while minimizing collateral damage to healthy tissue. Their preclinical success in treating various brain diseases in animal studies underscores their translational potential for human brain disease treatment. However, a deeper understanding of their long-term behavior, biodistribution, and biocompatibility within the brain remains crucial. Furthermore, exploring novel hydrogel systems and therapeutic combinations is paramount for advancing towards more effective treatments. This review summarizes the latest advancements in this field over the past 5 years, specifically highlighting preclinical progress with novel stimuli-responsive hydrogels for treating brain diseases.
Collapse
Affiliation(s)
- Bingqing Xie
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, Sichuan, China
| | - Huangfan Xie
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
14
|
Choi SY, Kim HJ, Hwang S, Park J, Park J, Lee JW, Son KH. The Modulation of Respiratory Epithelial Cell Differentiation by the Thickness of an Electrospun Poly-ε-Carprolactone Mesh Mimicking the Basement Membrane. Int J Mol Sci 2024; 25:6650. [PMID: 38928356 PMCID: PMC11203971 DOI: 10.3390/ijms25126650] [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: 04/24/2024] [Revised: 06/10/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
The topology of the basement membrane (BM) affects cell physiology and pathology, and BM thickening is associated with various chronic lung diseases. In addition, the topology of commercially available poly (ethylene terephthalate) (PET) membranes, which are used in preclinical in vitro models, differs from that of the human BM, which has a fibrous and elastic structure. In this study, we verified the effect of BM thickness on the differentiation of normal human bronchial epithelial (NHBE) cells. To evaluate whether the thickness of poly-ε-carprolactone (PCL) mesh affects the differentiation of NHBE cells, cells were grown on thin- (6-layer) and thick-layer (80-layer) meshes consisting of electrospun PCL nanofibers using an air-liquid interface (ALI) cell culture system. It was found that the NHBE cells formed a normal pseudostratified epithelium composed of ciliated, goblet, and basal cells on the thin-layer PCL mesh; however, goblet cell hyperplasia was observed on the thick-layer PCL mesh. Differentiated NHBE cells cultured on the thick-layer PCL mesh also demonstrated increased epithelial-mesenchymal transition (EMT) compared to those cultured on the thin-layer PCL mesh. In addition, expression of Sox9, nuclear factor (NF)-κB, and oxidative stress-related markers, which are also associated with goblet cell hyperplasia, was increased in the differentiated NHBE cells cultured on the thick-layer PCL mesh. Thus, the use of thick electrospun PCL mesh led to NHBE cells differentiating into hyperplastic goblet cells via EMT and the oxidative stress-related signaling pathway. Therefore, the topology of the BM, for example, thickness, may affect the differentiation direction of human bronchial epithelial cells.
Collapse
Affiliation(s)
- Seon Young Choi
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, College of Medicine, Gachon University, Incheon 21565, Republic of Korea; (S.Y.C.); (H.J.K.); (S.H.)
| | - Hyun Joo Kim
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, College of Medicine, Gachon University, Incheon 21565, Republic of Korea; (S.Y.C.); (H.J.K.); (S.H.)
| | - Soyoung Hwang
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, College of Medicine, Gachon University, Incheon 21565, Republic of Korea; (S.Y.C.); (H.J.K.); (S.H.)
| | - Jangho Park
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon 21999, Republic of Korea; (J.P.); (J.P.)
| | - Jungkyu Park
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon 21999, Republic of Korea; (J.P.); (J.P.)
| | - Jin Woo Lee
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon 21999, Republic of Korea; (J.P.); (J.P.)
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences & Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
| | - Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, College of Medicine, Gachon University, Incheon 21565, Republic of Korea; (S.Y.C.); (H.J.K.); (S.H.)
| |
Collapse
|
15
|
Nejati S, Mongeau L. In Vitro Investigation of Vocal Fold Cellular Response to Variations in Hydrogel Porosity and Elasticity. ACS Biomater Sci Eng 2024; 10:3909-3922. [PMID: 38783819 DOI: 10.1021/acsbiomaterials.4c00197] [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] [Indexed: 05/25/2024]
Abstract
Tissue regeneration is intricately influenced by the dynamic interplay between the physical attributes of tissue engineering scaffolds and the resulting biological responses. A tunable microporous hydrogel system was engineered using gelatin methacryloyl (GelMA) and polyethylene glycol diacrylate (PEGDA), with polyethylene glycol (PEG) serving as a porogen. Through systematic variation of PEGDA molecular weights, hydrogels with varying mechanical and architectural properties were obtained. The objective of the present study was to elucidate the impact of substrate mechanics and architecture on the immunological and reparative activities of vocal fold tissues. Mechanical characterization of the hydrogels was performed using tensile strength measurements and rheometry. Their morphological properties were investigated using scanning electron microscopy (SEM) and confocal microscopy. A series of biological assays were conducted. Cellular morphology, differentiation, and collagen synthesis of human vocal fold fibroblasts (hVFFs) were evaluated using immunostaining. Fibroblast proliferation was studied using the WST-1 assay, and cell migration was investigated via the Boyden chamber assay. Macrophage polarization and secretions were also examined using immunostaining and ELISA. The results revealed that increasing the molecular weight of PEGDA from 700 Da to 10,000 Da resulted in decreased hydrogel stiffness, from 62.6 to 8.8 kPa, and increased pore dimensions from approximately 64.9 to 137.4 μm. Biological evaluations revealed that hydrogels with a higher stiffness promoted fibroblast proliferation and spreading, albeit with an increased propensity for fibrosis, as indicated by a surge in myofibroblast differentiation and collagen synthesis. In contrast, hydrogels with greater molecular weights had a softer matrix with expanded pores, enhancing cellular migration and promoting an M2 macrophage phenotype conducive to tissue healing. The findings show that the hydrogels formulated with a PEGDA molecular weight of 6000 Da are best among the hydrogels considered for vocal fold repair. The microporous hydrogels could be tuned to serve in other tissue engineering applications.
Collapse
Affiliation(s)
- Sara Nejati
- Department of Mechanical Engineering, McGill University, Montreal H3A 0C3, Canada
| | - Luc Mongeau
- Department of Mechanical Engineering, McGill University, Montreal H3A 0C3, Canada
| |
Collapse
|
16
|
Rasoulian B, Sheikholislam Z, Houshdar Tehrani MH, Chegeni S, Hoveizi E, Rezayat SM, Tavakol S. Unveiling the superior function of RADA in bone regeneration compared to KSL as two critical cores within self-assembling peptide nanofibers: Insights from in vitro and in vivo studies. Regen Ther 2024; 26:999-1009. [PMID: 39553539 PMCID: PMC11564076 DOI: 10.1016/j.reth.2024.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/12/2024] [Accepted: 09/25/2024] [Indexed: 11/19/2024] Open
Abstract
Introduction Self-assembling peptide nanofibers have emerged as promising biomaterials in the realm of bone tissue engineering due to their biocompatibility, biodegradability, and ability to mimic the native extracellular matrix. This study delved into the comparative efficacy of two distinct self-assembling peptide nanofibers, RADA-BMHP1 and KSL-BMHP1, both incorporating the biological motif of BMHP1, but differing in their core peptide sequences. Methods Cell viability and osteogenic differentiation in rat mesenchymal stem cells (rMSCs), and bone regeneration in rat were compared. Results In vitro assays revealed that KSL-BMHP1 promoted enhanced cell viability, and nitric oxide production than RADA-BMHP1, an effect potentially attributable to its lower hydrophobicity and higher net charge at physiological pH. Conversely, RADA-BMHP1 induced superior osteogenic differentiation, evidenced by upregulation of key osteogenic genes, increased alkaline phosphatase activity (ALP), and enhanced matrix mineralization which may be attributed to its higher protein-binding potential and grand hydropathy, facilitating interactions between the peptide nanofibers and proteins involved in osteogenesis. In vivo experiments utilizing a rat bone defect model demonstrated that both peptide nanofibers improved bone regeneration at the genes level and ALP activity, with RADA-BMHP1 exhibiting a more pronounced increase in bone formation compared to KSL-BMHP1. Histological evaluation using H&E, Masson's trichrome and Wright-Giemsa staining confirmed the biocompatibility of both nanofibers. Conclusion These findings underscore the pivotal role of the core structure of self-assembling peptide nanofibers, beyond their biological motif, in the fate of tissue regeneration. Further research is warranted to optimize the physicochemical properties and functionalization of these nanofibers to enhance their efficacy in bone regeneration applications.
Collapse
Affiliation(s)
- Bita Rasoulian
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- School of Biomedical Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Zahra Sheikholislam
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Solmaz Chegeni
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Hoveizi
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Seyed Mahdi Rezayat
- Department of Medical Nanotechnology, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Research and Development, Tavakol BioMimetic Technologies Company, Tehran, Iran
| |
Collapse
|
17
|
Farsheed AC, Zevallos-Delgado C, Yu LT, Saeidifard S, Swain JWR, Makhoul JT, Thomas AJ, Cole CC, Garcia Huitron E, Grande-Allen KJ, Singh M, Larin KV, Hartgerink JD. Tunable Macroscopic Alignment of Self-Assembling Peptide Nanofibers. ACS NANO 2024; 18:12477-12488. [PMID: 38699877 PMCID: PMC11285723 DOI: 10.1021/acsnano.4c02030] [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] [Indexed: 05/05/2024]
Abstract
Progress in the design and synthesis of nanostructured self-assembling systems has facilitated the realization of numerous nanoscale geometries, including fibers, ribbons, and sheets. A key challenge has been achieving control across multiple length scales and creating macroscopic structures with nanoscale organization. Here, we present a facile extrusion-based fabrication method to produce anisotropic, nanofibrous hydrogels using self-assembling peptides. The application of shear force coinciding with ion-triggered gelation is used to kinetically trap supramolecular nanofibers into aligned, hierarchical macrostructures. Further, we demonstrate the ability to tune the nanostructure of macroscopic hydrogels through modulating phosphate buffer concentration during peptide self-assembly. In addition, increases in the nanostructural anisotropy of fabricated hydrogels are found to enhance their strength and stiffness under hydrated conditions. To demonstrate their utility as an extracellular matrix-mimetic biomaterial, aligned nanofibrous hydrogels are used to guide directional spreading of multiple cell types, but strikingly, increased matrix alignment is not always correlated with increased cellular alignment. Nanoscale observations reveal differences in cell-matrix interactions between variably aligned scaffolds and implicate the need for mechanical coupling for cells to understand nanofibrous alignment cues. In total, innovations in the supramolecular engineering of self-assembling peptides allow us to decouple nanostructure from macrostructure and generate a gradient of anisotropic nanofibrous hydrogels. We anticipate that control of architecture at multiple length scales will be critical for a variety of applications, including the bottom-up tissue engineering explored here.
Collapse
Affiliation(s)
- Adam C Farsheed
- Department of Bioengineering, Rice University, Houston, Texas 77005, United States
| | | | - Le Tracy Yu
- Department of Chemistry, Rice University, Houston, Texas 77005, United States
| | - Sajede Saeidifard
- Department of Biomedical Engineering, University of Houston, Houston, Texas 77204, United States
| | - Joseph W R Swain
- Department of Chemistry, Rice University, Houston, Texas 77005, United States
| | - Jonathan T Makhoul
- Department of Bioengineering, Rice University, Houston, Texas 77005, United States
- Department of Chemistry, Rice University, Houston, Texas 77005, United States
| | - Adam J Thomas
- Department of Chemistry, Rice University, Houston, Texas 77005, United States
| | - Carson C Cole
- Department of Chemistry, Rice University, Houston, Texas 77005, United States
| | - Eric Garcia Huitron
- Department of Bioengineering, Rice University, Houston, Texas 77005, United States
| | | | - Manmohan Singh
- Department of Biomedical Engineering, University of Houston, Houston, Texas 77204, United States
| | - Kirill V Larin
- Department of Biomedical Engineering, University of Houston, Houston, Texas 77204, United States
| | - Jeffrey D Hartgerink
- Department of Bioengineering, Rice University, Houston, Texas 77005, United States
- Department of Chemistry, Rice University, Houston, Texas 77005, United States
| |
Collapse
|
18
|
Jerka D, Bonowicz K, Piekarska K, Gokyer S, Derici US, Hindy OA, Altunay BB, Yazgan I, Steinbrink K, Kleszczyński K, Yilgor P, Gagat M. Unraveling Endothelial Cell Migration: Insights into Fundamental Forces, Inflammation, Biomaterial Applications, and Tissue Regeneration Strategies. ACS APPLIED BIO MATERIALS 2024; 7:2054-2069. [PMID: 38520346 PMCID: PMC11022177 DOI: 10.1021/acsabm.3c01227] [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: 12/12/2023] [Revised: 03/07/2024] [Accepted: 03/11/2024] [Indexed: 03/25/2024]
Abstract
Cell migration is vital for many fundamental biological processes and human pathologies throughout our life. Dynamic molecular changes in the tissue microenvironment determine modifications of cell movement, which can be reflected either individually or collectively. Endothelial cell (EC) migratory adaptation occurs during several events and phenomena, such as endothelial injury, vasculogenesis, and angiogenesis, under both normal and highly inflammatory conditions. Several advantageous processes can be supported by biomaterials. Endothelial cells are used in combination with various types of biomaterials to design scaffolds promoting the formation of mature blood vessels within tissue engineered structures. Appropriate selection, in terms of scaffolding properties, can promote desirable cell behavior to varying degrees. An increasing amount of research could lead to the creation of the perfect biomaterial for regenerative medicine applications. In this review, we summarize the state of knowledge regarding the possible systems by which inflammation may influence endothelial cell migration. We also describe the fundamental forces governing cell motility with a specific focus on ECs. Additionally, we discuss the biomaterials used for EC culture, which serve to enhance the proliferative, proangiogenic, and promigratory potential of cells. Moreover, we introduce the mechanisms of cell movement and highlight the significance of understanding these mechanisms in the context of designing scaffolds that promote tissue regeneration.
Collapse
Affiliation(s)
- Dominika Jerka
- Department
of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
| | - Klaudia Bonowicz
- Department
of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
- Faculty
of Medicine, Collegium Medicum, Mazovian
Academy in Płock, 09-402 Płock, Poland
| | - Klaudia Piekarska
- Department
of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
| | - Seyda Gokyer
- Department
of Biomedical Engineering, Faculty of Engineering, Ankara University, Ankara 06100, Turkey
| | - Utku Serhat Derici
- Department
of Biomedical Engineering, Faculty of Engineering, Ankara University, Ankara 06100, Turkey
| | - Osama Ali Hindy
- Department
of Biomedical Engineering, Faculty of Engineering, Ankara University, Ankara 06100, Turkey
| | - Baris Burak Altunay
- Department
of Biomedical Engineering, Faculty of Engineering, Ankara University, Ankara 06100, Turkey
| | - Işıl Yazgan
- Department
of Biomedical Engineering, Faculty of Engineering, Ankara University, Ankara 06100, Turkey
| | - Kerstin Steinbrink
- Department
of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany
| | - Konrad Kleszczyński
- Department
of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany
| | - Pinar Yilgor
- Department
of Biomedical Engineering, Faculty of Engineering, Ankara University, Ankara 06100, Turkey
| | - Maciej Gagat
- Department
of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
- Faculty
of Medicine, Collegium Medicum, Mazovian
Academy in Płock, 09-402 Płock, Poland
| |
Collapse
|
19
|
Ripamonti U, Duarte R. Mechanistic insights into the spontaneous induction of bone formation. BIOMATERIALS ADVANCES 2024; 158:213795. [PMID: 38335762 DOI: 10.1016/j.bioadv.2024.213795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/19/2023] [Accepted: 02/01/2024] [Indexed: 02/12/2024]
Abstract
The grand discovery of morphogens, or "form-generating substances", revealed that tissue morphogenesis is initiated by soluble molecular signals or morphogens primarily belonging to the transforming growth factor-β (TGF-β) supergene family. The regenerative potential of bone rests on its extracellular matrix, which is the repository of several morphogens that tightly control cellular differentiating pathways, cellular matrix deposition and remodeling. Alluringly, the matrix also contains specific factors transferred from the heterotopic implanted bone matrices initiating "Tissue Induction", as provocatively described in Nature in 1945. Later, it was found that selected genes and gene products of the TGF-β supergene family singly, synchronously, and synergistically mastermind the induction of bone formation. This review describes the phenomenon of the spontaneous and/or intrinsic osteoinductivity of calcium phosphate-based biomaterials and titanium' constructs without the applications of soluble osteogenetic molecular signals. The review shows the spontaneous induction of bone formation initiated by Ca++ activating stem cell differentiation and up-regulation of bone morphogenetic proteins genes. Expressed gene products are embedded into the concavities of the calcium phosphate-based substrata, initiating bone formation as a secondary response. Pure titanium's substrata do not initiate the spontaneous induction of bone formation. The induction of bone is solely dependent on acid, alkali and heat treatments to form apatite layers on the treated titanium surfaces. The induction of bone formation is achieved exclusively by apatite-based biomaterial surfaces. The hydroxyapatite, in its various forms and geometric configurations, finely tunes the induction of bone formation in heterotopic sites. Cellular differentiation by fine-tuning of the cellular molecular machinery is initiated by specific geometric modularity of the hydroxyapatite substrata that push cellular buttons that start the ripple-like cascade of "Tissue Induction", generating newly formed ossicles with bone marrow in heterotopic extraskeletal sites. The highlighted mechanistic insights into the spontaneous induction of bone formation are a research platform invocating selected molecular elements to construct the induction of bone formation.
Collapse
Affiliation(s)
- Ugo Ripamonti
- Bone Research Laboratory, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - Raquel Duarte
- Bone Research Laboratory, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; Internal Medicine Research Laboratory, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
20
|
Shim HW, Kurian AG, Lee J, Lee SC, Kim HW, Singh RK, Lee JH. Surface-Engineered Titanium with Nanoceria to Enhance Soft Tissue Integration Via Reactive Oxygen Species Modulation and Nanotopographical Sensing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:13622-13639. [PMID: 38466038 DOI: 10.1021/acsami.4c02119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
The design of implantable biomaterials involves precise tuning of surface features because the early cellular fate on such engineered surfaces is highly influenced by many physicochemical factors [roughness, hydrophilicity, reactive oxygen species (ROS) responsiveness, etc.]. Herein, to enhance soft tissue integration for successful implantation, Ti substrates decorated with uniform layers of nanoceria (Ce), called Ti@Ce, were optimally developed by a simple and cost-effective in situ immersion coating technique. The characterization of Ti@Ce shows a uniform Ce distribution with enhanced roughness (∼3-fold increase) and hydrophilicity (∼4-fold increase) and adopted ROS-scavenging capacity by nanoceria coating. When human gingival fibroblasts were seeded on Ti@Ce under oxidative stress conditions, Ti@Ce supported cellular adhesion, spreading, and survivability by its cellular ROS-scavenging capacity. Mechanistically, the unique nanocoating resulted in higher expression of amphiphysin (a nanotopology sensor), paxillin (a focal adhesion protein), and cell adhesive proteins (collagen-1 and fibronectin). Ti@Ce also led to global chromatin condensation by decreasing histone 3 acetylation as an early differentiation feature. Transcriptome analysis by RNA sequencing confirmed the chromatin remodeling, antiapoptosis, antioxidant, cell adhesion, and TGF-β signaling-related gene signatures in Ti@Ce. As key fibroblast transcription (co)factors, Ti@Ce promotes serum response factor and MRTF-α nucleus localization. Considering all of this, it is proposed that the surface engineering approach using Ce could improve the biological properties of Ti implants, supporting their functioning at soft tissue interfaces and utilization as a bioactive implant for clinical conditions such as peri-implantitis.
Collapse
Affiliation(s)
- Hye-Won Shim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Amal George Kurian
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Jiwon Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Sang-Cheol Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
| | - Rajendra K Singh
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
21
|
Redolfi Riva E, Özkan M, Contreras E, Pawar S, Zinno C, Escarda-Castro E, Kim J, Wieringa P, Stellacci F, Micera S, Navarro X. Beyond the limiting gap length: peripheral nerve regeneration through implantable nerve guidance conduits. Biomater Sci 2024; 12:1371-1404. [PMID: 38363090 DOI: 10.1039/d3bm01163a] [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: 02/17/2024]
Abstract
Peripheral nerve damage results in the loss of sensorimotor and autonomic functions, which is a significant burden to patients. Furthermore, nerve injuries greater than the limiting gap length require surgical repair. Although autografts are the preferred clinical choice, their usage is impeded by their limited availability, dimensional mismatch, and the sacrifice of another functional donor nerve. Accordingly, nerve guidance conduits, which are tubular scaffolds engineered to provide a biomimetic environment for nerve regeneration, have emerged as alternatives to autografts. Consequently, a few nerve guidance conduits have received clinical approval for the repair of short-mid nerve gaps but failed to regenerate limiting gap damage, which represents the bottleneck of this technology. Thus, it is still necessary to optimize the morphology and constituent materials of conduits. This review summarizes the recent advances in nerve conduit technology. Several manufacturing techniques and conduit designs are discussed, with emphasis on the structural improvement of simple hollow tubes, additive manufacturing techniques, and decellularized grafts. The main objective of this review is to provide a critical overview of nerve guidance conduit technology to support regeneration in long nerve defects, promote future developments, and speed up its clinical translation as a reliable alternative to autografts.
Collapse
Affiliation(s)
- Eugenio Redolfi Riva
- The Biorobotic Institute, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy.
- Department of Excellence in Robotics & AI, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy
| | - Melis Özkan
- Institute of Materials, école Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
- Bertarelli Foundation Chair in Translational Neural Engineering, Center for Neuroprosthetics and Institute of Bioengineering, école Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Estefania Contreras
- Integral Service for Laboratory Animals (SIAL), Faculty of Veterinary, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Spain.
| | - Sujeet Pawar
- Institute of Materials, école Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Ciro Zinno
- The Biorobotic Institute, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy.
- Department of Excellence in Robotics & AI, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy
| | - Enrique Escarda-Castro
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Jaehyeon Kim
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Paul Wieringa
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Francesco Stellacci
- Institute of Materials, école Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
- Institute of Materials, Department of Bioengineering and Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Station 12, CH-1015 Lausanne, Switzerland
| | - Silvestro Micera
- The Biorobotic Institute, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy.
- Department of Excellence in Robotics & AI, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy
- Bertarelli Foundation Chair in Translational Neural Engineering, Center for Neuroprosthetics and Institute of Bioengineering, école Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Xavier Navarro
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Institute Guttmann Foundation, Hospital of Neurorehabilitation, Badalona, Spain
| |
Collapse
|
22
|
Kim H, Kim S, Lim H, Chung AJ. Expanding CAR-T cell immunotherapy horizons through microfluidics. LAB ON A CHIP 2024; 24:1088-1120. [PMID: 38174732 DOI: 10.1039/d3lc00622k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Chimeric antigen receptor (CAR)-T cell therapies have revolutionized cancer treatment, particularly in hematological malignancies. However, their application to solid tumors is limited, and they face challenges in safety, scalability, and cost. To enhance current CAR-T cell therapies, the integration of microfluidic technologies, harnessing their inherent advantages, such as reduced sample consumption, simplicity in operation, cost-effectiveness, automation, and high scalability, has emerged as a powerful solution. This review provides a comprehensive overview of the step-by-step manufacturing process of CAR-T cells, identifies existing difficulties at each production stage, and discusses the successful implementation of microfluidics and related technologies in addressing these challenges. Furthermore, this review investigates the potential of microfluidics-based methodologies in advancing cell-based therapy across various applications, including solid tumors, next-generation CAR constructs, T-cell receptors, and the development of allogeneic "off-the-shelf" CAR products.
Collapse
Affiliation(s)
- Hyelee Kim
- Department of Bioengineering, Korea University, 02841 Seoul, Republic of Korea
- Interdisciplinary Program in Precision Public Health (PPH), Korea University, 02841 Seoul, Republic of Korea.
| | - Suyeon Kim
- Department of Bioengineering, Korea University, 02841 Seoul, Republic of Korea
- Interdisciplinary Program in Precision Public Health (PPH), Korea University, 02841 Seoul, Republic of Korea.
| | - Hyunjung Lim
- Interdisciplinary Program in Precision Public Health (PPH), Korea University, 02841 Seoul, Republic of Korea.
| | - Aram J Chung
- Department of Bioengineering, Korea University, 02841 Seoul, Republic of Korea
- Interdisciplinary Program in Precision Public Health (PPH), Korea University, 02841 Seoul, Republic of Korea.
- School of Biomedical Engineering, Korea University, 02841 Seoul, Republic of Korea.
- MxT Biotech, 04785 Seoul, Republic of Korea
| |
Collapse
|
23
|
Suh K, Cho YK, Breinyn IB, Cohen DJ. E-cadherin biomaterials reprogram collective cell migration and cell cycling by forcing homeostatic conditions. Cell Rep 2024; 43:113743. [PMID: 38358889 DOI: 10.1016/j.celrep.2024.113743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 01/02/2024] [Accepted: 01/18/2024] [Indexed: 02/17/2024] Open
Abstract
Cells attach to the world through either cell-extracellular matrix adhesion or cell-cell adhesion, and traditional biomaterials imitate the matrix for integrin-based adhesion. However, materials incorporating cadherin proteins that mimic cell-cell adhesion offer an alternative to program cell behavior and integrate into living tissues. We investigated how cadherin substrates affect collective cell migration and cell cycling in epithelia. Our approach involved biomaterials with matrix proteins on one-half and E-cadherin proteins on the other, forming a "Janus" interface across which we grew a single sheet of cells. Tissue regions over the matrix side exhibited normal collective dynamics, but an abrupt behavior shift occurred across the Janus boundary onto the E-cadherin side, where cells attached to the substrate via E-cadherin adhesions, resulting in stalled migration and slowing of the cell cycle. E-cadherin surfaces disrupted long-range mechanical coordination and nearly doubled the length of the G0/G1 phase of the cell cycle, linked to the lack of integrin focal adhesions on the E-cadherin surface.
Collapse
Affiliation(s)
- Kevin Suh
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Youn Kyoung Cho
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Isaac B Breinyn
- Department of Quantitative and Computational Biology, Princeton University, Princeton, NJ 08544, USA
| | - Daniel J Cohen
- Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
24
|
Jalloh US, Gsell A, Gultian KA, MacAulay J, Madden A, Smith J, Siri L, Vega SL. Synthesis and Photopatterning of Synthetic Thiol-Norbornene Hydrogels. Gels 2024; 10:164. [PMID: 38534582 DOI: 10.3390/gels10030164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 03/28/2024] Open
Abstract
Hydrogels are a class of soft biomaterials and the material of choice for a myriad of biomedical applications due to their biocompatibility and highly tunable mechanical and biochemical properties. Specifically, light-mediated thiol-norbornene click reactions between norbornene-modified macromers and di-thiolated crosslinkers can be used to form base hydrogels amenable to spatial biochemical modifications via subsequent light reactions between pendant norbornenes in the hydrogel network and thiolated peptides. Macromers derived from natural sources (e.g., hyaluronic acid, gelatin, alginate) can cause off-target cell signaling, and this has motivated the use of synthetic macromers such as poly(ethylene glycol) (PEG). In this study, commercially available 8-arm norbornene-modified PEG (PEG-Nor) macromers were reacted with di-thiolated crosslinkers (dithiothreitol, DTT) to form synthetic hydrogels. By varying the PEG-Nor weight percent or DTT concentration, hydrogels with a stiffness range of 3.3 kPa-31.3 kPa were formed. Pendant norbornene groups in these hydrogels were used for secondary reactions to either increase hydrogel stiffness (by reacting with DTT) or to tether mono-thiolated peptides to the hydrogel network. Peptide functionalization has no effect on bulk hydrogel mechanics, and this confirms that mechanical and biochemical signals can be independently controlled. Using photomasks, thiolated peptides can also be photopatterned onto base hydrogels, and mesenchymal stem cells (MSCs) attach and spread on RGD-functionalized PEG-Nor hydrogels. MSCs encapsulated in PEG-Nor hydrogels are also highly viable, demonstrating the ability of this platform to form biocompatible hydrogels for 2D and 3D cell culture with user-defined mechanical and biochemical properties.
Collapse
Affiliation(s)
- Umu S Jalloh
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - Arielle Gsell
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - Kirstene A Gultian
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - James MacAulay
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - Abigail Madden
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - Jillian Smith
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - Luke Siri
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - Sebastián L Vega
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
- Department of Orthopaedic Surgery, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| |
Collapse
|
25
|
Man K, Mazumder S, Dahotre NB, Yang Y. Surface Nanostructures Enhanced Biocompatibility and Osteoinductivity of Laser-Additively Manufactured CoCrMo Alloys. ACS OMEGA 2023; 8:47658-47666. [PMID: 38144145 PMCID: PMC10734289 DOI: 10.1021/acsomega.3c04305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/19/2023] [Indexed: 12/26/2023]
Abstract
Cobalt-chromium-molybdenum (CoCrMo) alloys are widely used in orthopedic implants due to their excellent corrosion and wear resistance and superior mechanical properties. However, their limited capability to promote cell adhesion and new bone tissue formation, poor blood compatibility, and risk of microbial infection can lead to implant failure or reduced implant lifespan. Surface structure modification has been used to improve the cytocompatibility and blood compatibility of implant materials and reduce the risk of infection. In this study, we prepared CoCrMo alloys with surface nanostructures of various aspect ratios (AR) using laser-directed energy deposition (L-DED) and biocorrosion. Our results showed that medium and high AR nanostructures reduced platelet adhesion, while all of the alloys demonstrated good blood compatibility and antibacterial properties. Moreover, the medium and high AR nanostructures promoted cell adhesion and spreading of both preosteoblast MC3T3 cells and human bone marrow mesenchymal stem cells (hMSCs). Furthermore, the nanostructure promoted the osteogenic differentiation of both cell types compared with the flat control surface, with a substantial enhancing effect for the medium and high ARs. Our study proposes a promising approach for developing implant materials with improved clinical outcomes.
Collapse
Affiliation(s)
- Kun Man
- Department
of Biomedical Engineering, University of
North Texas, Denton, Texas 76207, United States
- Center
for Agile and Adaptive Additive Manufacturing, University of North Texas, Denton, Texas 76207, United States
| | - Sangram Mazumder
- Center
for Agile and Adaptive Additive Manufacturing, University of North Texas, Denton, Texas 76207, United States
- Department
of Materials Science and Engineering, University
of North Texas, Denton, Texas 76207, United States
| | - Narendra B. Dahotre
- Center
for Agile and Adaptive Additive Manufacturing, University of North Texas, Denton, Texas 76207, United States
- Department
of Materials Science and Engineering, University
of North Texas, Denton, Texas 76207, United States
| | - Yong Yang
- Department
of Biomedical Engineering, University of
North Texas, Denton, Texas 76207, United States
| |
Collapse
|
26
|
Peussa H, Fedele C, Tran H, Marttinen M, Fadjukov J, Mäntylä E, Priimägi A, Nymark S, Ihalainen TO. Light-Induced Nanoscale Deformation in Azobenzene Thin Film Triggers Rapid Intracellular Ca 2+ Increase via Mechanosensitive Cation Channels. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206190. [PMID: 37946608 PMCID: PMC10724422 DOI: 10.1002/advs.202206190] [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: 10/24/2022] [Revised: 09/15/2023] [Indexed: 11/12/2023]
Abstract
Epithelial cells are in continuous dynamic biochemical and physical interaction with their extracellular environment. Ultimately, this interplay guides fundamental physiological processes. In these interactions, cells generate fast local and global transients of Ca2+ ions, which act as key intracellular messengers. However, the mechanical triggers initiating these responses have remained unclear. Light-responsive materials offer intriguing possibilities to dynamically modify the physical niche of the cells. Here, a light-sensitive azobenzene-based glassy material that can be micropatterned with visible light to undergo spatiotemporally controlled deformations is used. Real-time monitoring of consequential rapid intracellular Ca2+ signals reveals that the mechanosensitive cation channel Piezo1 has a major role in generating the Ca2+ transients after nanoscale mechanical deformation of the cell culture substrate. Furthermore, the studies indicate that Piezo1 preferably responds to shear deformation at the cell-material interphase rather than to absolute topographical change of the substrate. Finally, the experimentally verified computational model suggests that Na+ entering alongside Ca2+ through the mechanosensitive cation channels modulates the duration of Ca2+ transients, influencing differently the directly stimulated cells and their neighbors. This highlights the complexity of mechanical signaling in multicellular systems. These results give mechanistic understanding on how cells respond to rapid nanoscale material dynamics and deformations.
Collapse
Affiliation(s)
- Heidi Peussa
- BioMediTechFaculty of Medicine and Health TechnologyTampere UniversityArvo Ylpön katu 34Tampere33520Finland
| | - Chiara Fedele
- Faculty of Engineering and Natural SciencesTampere UniversityKorkeakoulunkatu 3Tampere33720Finland
| | - Huy Tran
- BioMediTechFaculty of Medicine and Health TechnologyTampere UniversityArvo Ylpön katu 34Tampere33520Finland
| | - Mikael Marttinen
- BioMediTechFaculty of Medicine and Health TechnologyTampere UniversityArvo Ylpön katu 34Tampere33520Finland
| | - Julia Fadjukov
- BioMediTechFaculty of Medicine and Health TechnologyTampere UniversityArvo Ylpön katu 34Tampere33520Finland
| | - Elina Mäntylä
- BioMediTechFaculty of Medicine and Health TechnologyTampere UniversityArvo Ylpön katu 34Tampere33520Finland
| | - Arri Priimägi
- Faculty of Engineering and Natural SciencesTampere UniversityKorkeakoulunkatu 3Tampere33720Finland
| | - Soile Nymark
- BioMediTechFaculty of Medicine and Health TechnologyTampere UniversityArvo Ylpön katu 34Tampere33520Finland
| | - Teemu O. Ihalainen
- BioMediTechFaculty of Medicine and Health TechnologyTampere UniversityArvo Ylpön katu 34Tampere33520Finland
- Tampere Institute for Advanced StudyTampere UniversityArvo Ylpön katu 34Tampere33520Finland
| |
Collapse
|
27
|
Tymetska S, Shymborska Y, Stetsyshyn Y, Budkowski A, Bernasik A, Awsiuk K, Donchak V, Raczkowska J. Thermoresponsive Smart Copolymer Coatings Based on P(NIPAM- co-HEMA) and P(OEGMA- co-HEMA) Brushes for Regenerative Medicine. ACS Biomater Sci Eng 2023; 9:6256-6272. [PMID: 37874897 PMCID: PMC10646826 DOI: 10.1021/acsbiomaterials.3c00917] [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: 07/08/2023] [Revised: 10/04/2023] [Accepted: 10/04/2023] [Indexed: 10/26/2023]
Abstract
The fabrication of multifunctional, thermoresponsive platforms for regenerative medicine based on polymers that can be easily functionalized is one of the most important challenges in modern biomaterials science. In this study, we utilized atom transfer radical polymerization (ATRP) to produce two series of novel smart copolymer brush coatings. These coatings were based on copolymerizing 2-hydroxyethyl methacrylate (HEMA) with either oligo(ethylene glycol) methyl ether methacrylate (OEGMA) or N-isopropylacrylamide (NIPAM). The chemical compositions of the resulting brush coatings, namely, poly(oligo(ethylene glycol) methyl ether methacrylate-co-2-hydroxyethyl methacrylate) (P(OEGMA-co-HEMA)) and poly(N-isopropylacrylamide-co-2-hydroxyethyl methacrylate) (P(NIPAM-co-HEMA)), were predicted using reactive ratios of the monomers. These predictions were then verified using time-of-flight-secondary ion mass spectrometry (ToF-SIMS) and X-ray photoelectron spectroscopy (XPS). The thermoresponsiveness of the coatings was examined through water contact angle (CA) measurements at different temperatures, revealing a transition driven by lower critical solution temperature (LCST) or upper critical solution temperature (UCST) or a vanishing transition. The type of transition observed depended on the chemical composition of the coatings. Furthermore, it was demonstrated that the transition temperature of the coatings could be easily adjusted by modifying their composition. The topography of the coatings was characterized using atomic force microscopy (AFM). To assess the biocompatibility of the coatings, dermal fibroblast cultures were employed, and the results indicated that none of the coatings exhibited cytotoxicity. However, the shape and arrangement of the cells were significantly influenced by the chemical structure of the coating. Additionally, the viability of the cells was correlated with the wettability and roughness of the coatings, which determined the initial adhesion of the cells. Lastly, the temperature-induced changes in the properties of the fabricated copolymer coatings effectively controlled cell morphology, adhesion, and spontaneous detachment in a noninvasive, enzyme-free manner that was confirmed using optical microscopy.
Collapse
Affiliation(s)
- Svitlana Tymetska
- Jagiellonian
University, Doctoral School of Exact and
Natural Sciences, Łojasiewicza
11, 30-348 Kraków, Poland
- Jagiellonian
University, Faculty of Physics, Astronomy
and Applied Computer Science, Smoluchowski Institute of Physics, Łojasiewicza 11, 30-348 Kraków, Poland
| | - Yana Shymborska
- Jagiellonian
University, Doctoral School of Exact and
Natural Sciences, Łojasiewicza
11, 30-348 Kraków, Poland
- Jagiellonian
University, Faculty of Physics, Astronomy
and Applied Computer Science, Smoluchowski Institute of Physics, Łojasiewicza 11, 30-348 Kraków, Poland
- Lviv
Polytechnic National University, St. George’s Square 2, 79013 Lviv, Ukraine
| | - Yurij Stetsyshyn
- Lviv
Polytechnic National University, St. George’s Square 2, 79013 Lviv, Ukraine
| | - Andrzej Budkowski
- Jagiellonian
University, Faculty of Physics, Astronomy
and Applied Computer Science, Smoluchowski Institute of Physics, Łojasiewicza 11, 30-348 Kraków, Poland
| | - Andrzej Bernasik
- Faculty
of Physics and Applied Computer Science, AGH - University of Science and Technology, al. Mickiewicza 30, 30-049 Kraków, Poland
| | - Kamil Awsiuk
- Jagiellonian
University, Faculty of Physics, Astronomy
and Applied Computer Science, Smoluchowski Institute of Physics, Łojasiewicza 11, 30-348 Kraków, Poland
| | - Volodymyr Donchak
- Lviv
Polytechnic National University, St. George’s Square 2, 79013 Lviv, Ukraine
| | - Joanna Raczkowska
- Jagiellonian
University, Faculty of Physics, Astronomy
and Applied Computer Science, Smoluchowski Institute of Physics, Łojasiewicza 11, 30-348 Kraków, Poland
| |
Collapse
|
28
|
Chen J, Chen X, Ma Y, Liu Y, Li J, Peng K, Dai Y, Chen X. Effect of Anisotropic Structural Depth on Orientation and Differentiation Behavior of Skeletal Muscle Cells. ACS OMEGA 2023; 8:41374-41382. [PMID: 37969971 PMCID: PMC10634202 DOI: 10.1021/acsomega.3c04981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/03/2023] [Indexed: 11/17/2023]
Abstract
Extensive research has been conducted to examine how substrate topological factors are involved in modulating the cell behavior. Among numerous topological factors, the vital influence of the touchable depth of substrates on cell behaviors has already been extensively characterized, but the response of cells to the topological structure at untouchable depth is still elusive. Herein, the influences of substrate depth on myoblast behaviors are systematically investigated using substrates with depths ranging from touchable depth (microgrooved) to untouchable depth (microbridges). The results show that an increase in microgroove depth is accompanied by an inhibited cell spreading, an enhanced elongation, and a more obvious orientation along microgrooves. Interestingly, myoblasts located on microbridges show a more pronounced elongation with increasing culture time but a position-dependent orientation. Myoblasts on the center and parallel boundary of microbridges orient along the bridges, while myoblasts on the vertical boundary align perpendicular to the microbridges. Moreover, the differentiation results of the myoblasts indicate that the differentiated myotubes can maintain this position-dependent orientation. The simulation of the stress field in cell monolayers suggests that the position-dependent orientation is caused by the comprehensive response of myoblasts to the substrate discontinuity and substrate depth. These findings provide valuable insights into the mechanism of cell depth sensing and could inform the design of tissue engineering scaffolds for skeletal muscle and biohybrid actuation.
Collapse
Affiliation(s)
- Jianfeng Chen
- School
of Advanced Manufacturing, Nanchang University, Nanchang 330031, Jiangxi, P. R. China
| | - Xuefei Chen
- School
of Advanced Manufacturing, Nanchang University, Nanchang 330031, Jiangxi, P. R. China
| | - Yihao Ma
- School
of Advanced Manufacturing, Nanchang University, Nanchang 330031, Jiangxi, P. R. China
| | - Yiran Liu
- School
of Advanced Manufacturing, Nanchang University, Nanchang 330031, Jiangxi, P. R. China
| | - Jin Li
- School
of Advanced Manufacturing, Nanchang University, Nanchang 330031, Jiangxi, P. R. China
| | - Kai Peng
- School
of Advanced Manufacturing, Nanchang University, Nanchang 330031, Jiangxi, P. R. China
| | - Yichuan Dai
- School
of Advanced Manufacturing, Nanchang University, Nanchang 330031, Jiangxi, P. R. China
| | - Xiaoxiao Chen
- School
of Advanced Manufacturing, Nanchang University, Nanchang 330031, Jiangxi, P. R. China
| |
Collapse
|
29
|
Wang K, Frey N, Garcia A, Man K, Yang Y, Gualerzi A, Clemens ZJ, Bedoni M, LeDuc PR, Ambrosio F. Nanotopographical Cues Tune the Therapeutic Potential of Extracellular Vesicles for the Treatment of Aged Skeletal Muscle Injuries. ACS NANO 2023; 17:19640-19651. [PMID: 37797946 PMCID: PMC10603813 DOI: 10.1021/acsnano.3c02269] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023]
Abstract
Skeletal muscle regeneration relies on the tightly temporally regulated lineage progression of muscle stem/progenitor cells (MPCs) from activation to proliferation and, finally, differentiation. However, with aging, MPC lineage progression is disrupted and delayed, ultimately causing impaired muscle regeneration. Extracellular vesicles (EVs) have attracted broad attention as next-generation therapeutics for promoting tissue regeneration. As a next step toward clinical translation, strategies to manipulate EV effects on downstream cellular targets are needed. Here, we developed an engineering strategy to tune the therapeutic potential of EVs using nanotopographical cues. We found that EVs released by young MPCs cultured on flat substrates (fEVs) promoted the proliferation of aged MPCs while EVs released by MPCs cultured on nanogratings (nEVs) promoted myogenic differentiation. We then employed a bioengineered 3D muscle aging model to optimize the administration protocol and test the therapeutic potential of fEVs and nEVs in a high-throughput manner. We found that the sequential administration first of fEVs during the phase of MPC proliferative expansion (i.e., 1 day after injury) followed by nEV administration at the stage of MPC differentiation (i.e., 3 days after injury) enhanced aged muscle regeneration to a significantly greater extent than fEVs and nEVs delivered either in isolation or mixed. The beneficial effects of the sequential EV treatment strategy were further validated in vivo, as evidenced by increased myofiber size and improved functional recovery. Collectively, our study demonstrates the ability of topographical cues to tune EV therapeutic potential and highlights the importance of optimizing the EV administration strategy to accelerate aged skeletal muscle regeneration.
Collapse
Affiliation(s)
- Kai Wang
- Discovery
Center for Musculoskeletal Recovery, Schoen
Adams Research Institute at Spaulding, Charlestown, Massachusetts 02129, United States
- Department
of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department
of Physical Medicine & Rehabilitation, Spaulding Rehabilitation Hospital, Charlestown, Massachusetts 02129, United States
- Department
of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| | - Nolan Frey
- Department
of Biological Sciences, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
| | - Andres Garcia
- Department
of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
| | - Kun Man
- Department
of Biomedical Engineering, University of
North Texas, Denton, Texas 76207, United States
| | - Yong Yang
- Department
of Biomedical Engineering, University of
North Texas, Denton, Texas 76207, United States
| | - Alice Gualerzi
- IRCCS
Fondazione Don Carlo Gnocchi ONLUS, Milan 20148, Italy
| | - Zachary J. Clemens
- Department
of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| | - Marzia Bedoni
- IRCCS
Fondazione Don Carlo Gnocchi ONLUS, Milan 20148, Italy
| | - Philip R. LeDuc
- Department
of Biological Sciences, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
- Department
of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
- Department
of Computational Biology, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
- Department
of Biomedical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
- Department
of Electrical and Computer Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Fabrisia Ambrosio
- Discovery
Center for Musculoskeletal Recovery, Schoen
Adams Research Institute at Spaulding, Charlestown, Massachusetts 02129, United States
- Department
of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department
of Physical Medicine & Rehabilitation, Spaulding Rehabilitation Hospital, Charlestown, Massachusetts 02129, United States
- Department
of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|
30
|
Turan Sorhun D, Kuşoğlu A, Öztürk E. Developing Bovine Brain-Derived Extracellular Matrix Hydrogels: a Screen of Decellularization Methods for Their Impact on Biochemical and Mechanical Properties. ACS OMEGA 2023; 8:36933-36947. [PMID: 37841171 PMCID: PMC10569007 DOI: 10.1021/acsomega.3c04064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 09/20/2023] [Indexed: 10/17/2023]
Abstract
Tissue models that recapitulate the key biochemical and physical aspects of the brain have been highly pursued in neural tissue engineering. Decellularization of native organs offers the advantage of preserving the composition of native extracellular matrix (ECM). Brain ECM has distinct features which play a major role in neural cell behavior. Cell instructive ligands and mechanical properties take part in the regulation of cellular processes in homeostasis and diseases. One of the main challenges in decellularization is maintaining mechanical integrity in reconstituted hydrogels and achieving physiologically relevant stiffness. The effect of the decellularization process on different mechanical aspects, particularly the viscoelasticity of brain-derived hydrogels, has not been addressed. In this study, we developed bovine brain-derived hydrogels for the first time. We pursued seven protocols for decellularization and screened their effect on biochemical content, hydrogel formation, and mechanical characteristics. We show that bovine brain offers an easily accessible alternative for in vitro brain tissue modeling. Our data demonstrate that the choice of decellularization method strongly alters gelation as well as the stiffness and viscoelasticity of the resulting hydrogels. Lastly, we investigated the cytocompatibility of brain ECM hydrogels and the effect of modulated mechanical properties on the growth and morphological features of neuroblastoma cells.
Collapse
Affiliation(s)
- Duygu Turan Sorhun
- Engineered
Cancer and Organ Models Laboratory, Koç
University, Istanbul 34450, Turkey
- Research
Center for Translational Medicine (KUTTAM), Koç University, Istanbul 34450, Turkey
| | - Alican Kuşoğlu
- Engineered
Cancer and Organ Models Laboratory, Koç
University, Istanbul 34450, Turkey
- Research
Center for Translational Medicine (KUTTAM), Koç University, Istanbul 34450, Turkey
| | - Ece Öztürk
- Engineered
Cancer and Organ Models Laboratory, Koç
University, Istanbul 34450, Turkey
- Research
Center for Translational Medicine (KUTTAM), Koç University, Istanbul 34450, Turkey
- Department
of Medical Biology, School of Medicine, Koç University, Istanbul 34450, Turkey
| |
Collapse
|
31
|
Zhou H, Ye S, Xu M, Hao L, Chen J, Fang Z, Guo K, Chen Y, Wang L. Dynamic surface adapts to multiple service stages by orchestrating responsive polymers and functional peptides. Biomaterials 2023; 301:122200. [PMID: 37423184 DOI: 10.1016/j.biomaterials.2023.122200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 07/11/2023]
Abstract
Control over the implant surface functions is highly desirable to enhance tissue healing outcomes but has remained unexplored to adapt to the different service stages. In the present study, we develop a smart titanium surface by orchestrating thermoresponsive polymer and antimicrobial peptide to enable dynamic adaptation to the implantation stage, normal physiological stage and bacterial infection stage. The optimized surface inhibited bacterial adhesion and biofilm formation during surgical implantation, while promoted osteogenesis in the physiological stage. The further temperature increase driven by bacterial infection induced polymer chain collapse to expose antimicrobial peptides by rupturing bacterial membranes, as well as protect the adhered cells from the hostile environment of infection and abnormal temperature. The engineered surface could inhibit infection and promote tissue healing in rabbit subcutaneous and bone defect infection models. This strategy enables the possibility to create a versatile surface platform to balance bacteria/cell-biomaterial interactions at different service stages of implants that has not been achieved before.
Collapse
Affiliation(s)
- Haiyan Zhou
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China
| | - Silin Ye
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China
| | - Mingjian Xu
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Lihui Hao
- Department of Stomatology, Xingtai Medical College, Xingtai 054000, China
| | - Junjian Chen
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China.
| | - Zhou Fang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Kunzhong Guo
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Yunhua Chen
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China.
| | - Lin Wang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China.
| |
Collapse
|
32
|
Shi G, Zhang P, Zhang X, Li J, Zheng X, Yan J, Zhang N, Yang H. The spatiotemporal heterogeneity of the biophysical microenvironment during hematopoietic stem cell development: from embryo to adult. Stem Cell Res Ther 2023; 14:251. [PMID: 37705072 PMCID: PMC10500792 DOI: 10.1186/s13287-023-03464-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 08/22/2023] [Indexed: 09/15/2023] Open
Abstract
Hematopoietic stem cells (HSCs) with the ability to self-renew and differentiate are responsible for maintaining the supply of all types of blood cells. The complex and delicate microenvironment surrounding HSCs is called the HSC niche and can provide physical, chemical, and biological stimuli to regulate the survival, maintenance, proliferation, and differentiation of HSCs. Currently, the exploration of the biophysical regulation of HSCs remains in its infancy. There is evidence that HSCs are susceptible to biophysical stimuli, suggesting that the construction of engineered niche biophysical microenvironments is a promising way to regulate the fate of HSCs in vitro and ultimately contribute to clinical applications. In this review, we introduced the spatiotemporal heterogeneous biophysical microenvironment during HSC development, homeostasis, and malignancy. Furthermore, we illustrated how these biophysical cues contribute to HSC behaviors, as well as the possible mechanotransduction mechanisms from the extracellular microenvironment into cells. Comprehending the important functions of these biophysical regulatory factors will provide novel approaches to resolve clinical problems.
Collapse
Affiliation(s)
- Guolin Shi
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, Shaanxi, China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Pan Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, Shaanxi, China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, China
- School of Food Science and Engineering, Shaanxi University of Science & Technology, Xi'an, China
| | - Xi Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, Shaanxi, China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Jing Li
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Xinmin Zheng
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, Shaanxi, China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Jinxiao Yan
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, Shaanxi, China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Nu Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, Shaanxi, China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Hui Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, Shaanxi, China.
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, China.
| |
Collapse
|
33
|
Verma BK, Chatterjee A, Kondaiah P, Gundiah N. Substrate Stiffness Modulates TGF-β Activation and ECM-Associated Gene Expression in Fibroblasts. Bioengineering (Basel) 2023; 10:998. [PMID: 37760100 PMCID: PMC10525202 DOI: 10.3390/bioengineering10090998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 06/19/2023] [Accepted: 07/06/2023] [Indexed: 09/29/2023] Open
Abstract
Transforming growth factor-β (TGF-β) is a multifunctional cytokine that regulates the expression of ECM-associated genes during early injury. Tissue fibrosis development is driven by synergistic cues between the evolving biochemical and mechanical milieu. Few studies have addressed the role of substrate stiffness on TGF-β activity and extracellular matrix (ECM)-associated genes. We used a commercial formulation of polydimethylsiloxane (PDMS) to fabricate substrates of 40 kPa, 300 kPa, and 1.5 MPa stiffness, and cultured the HMF3S fibroblasts on substrates. We quantified TGF-β protein secreted by HMF3S cells on different substrates using a TGF-β responsive promoter reporter assay. We also tested for variations in gene expression levels on the substrates using RT-PCR and Western blotting and determined the MMP-2 and MMP-9 activities with gelatin zymography. The results showed that TGF-β protein activation was significantly compromised at lower stiffnesses. The expression of integrin α5 decreased on lower stiffness substrates and correlated with inefficient TGF-β protein activation. Collagen I, collagen III, and MMP-2 expression levels were lower on softer substrates; there was little MMP-9 activity on all substrates. Cell and nuclear morphologies were more rounded on compliant substrates, correlating with increased tubulin expression. Proliferations were higher on stiffer substrates, whereas cells on softer substrates showed cell cycle arrest. These results demonstrated critical feedback mechanisms between substrate stiffness and ECM regulation by fibroblasts, relevant in fibrosis.
Collapse
Affiliation(s)
- Brijesh Kumar Verma
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India
| | - Aritra Chatterjee
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India
| | - Paturu Kondaiah
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru 560012, India
| | - Namrata Gundiah
- Department of Mechanical Engineering, Indian Institute of Science, Bengaluru 560012, India
| |
Collapse
|
34
|
Suh K, Cho YK, Breinyn IB, Cohen DJ. E-cadherin biointerfaces reprogram collective cell migration and cell cycling by forcing homeostatic conditions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.25.550505. [PMID: 37546933 PMCID: PMC10402016 DOI: 10.1101/2023.07.25.550505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Cells attach to the world around them in two ways-cell:extracellular-matrix adhesion and cell:cell adhesion-and conventional biomaterials are made to resemble the matrix to encourage integrin-based cell adhesion. However, interest is growing for cell-mimetic interfaces that mimic cell-cell interactions using cadherin proteins, as this offers a new way to program cell behavior and design synthetic implants and objects that can integrate directly into living tissues. Here, we explore how these cadherin-based materials affect collective cell behaviors, focusing specifically on collective migration and cell cycle regulation in cm-scale epithelia. We built culture substrates where half of the culture area was functionalized with matrix proteins and the contiguous half was functionalized with E-cadherin proteins, and we grew large epithelia across this 'Janus' interface. Parts of the tissues in contact with the matrix side of the Janus interface exhibited normal collective dynamics, but an abrupt shift in behaviors happened immediately across the Janus boundary onto the E-cadherin side, where cells formed hybrid E-cadherin junctions with the substrate, migration effectively froze in place, and cell-cycling significantly decreased. E-cadherin materials suppressed long-range mechanical correlations in the tissue and mechanical information reflected off the substrate interface. These effects could not be explained by conventional density, shape index, or contact inhibition explanations. E-cadherin surfaces nearly doubled the length of the G0/G1 phase of the cell cycle, which we ultimately connected to the exclusion of matrix focal adhesions induced by the E-cadherin culture surface.
Collapse
Affiliation(s)
- Kevin Suh
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA, 08544
| | - Youn Kyoung Cho
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA, 08544
| | - Isaac B Breinyn
- Department of Quantitative and Computational Biology, Princeton University, Princeton, NJ, USA, 08544
| | - Daniel J Cohen
- Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, NJ, USA, 08544
| |
Collapse
|
35
|
Shou Y, Teo XY, Wu KZ, Bai B, Kumar ARK, Low J, Le Z, Tay A. Dynamic Stimulations with Bioengineered Extracellular Matrix-Mimicking Hydrogels for Mechano Cell Reprogramming and Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300670. [PMID: 37119518 PMCID: PMC10375194 DOI: 10.1002/advs.202300670] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/10/2023] [Indexed: 06/19/2023]
Abstract
Cells interact with their surrounding environment through a combination of static and dynamic mechanical signals that vary over stimulus types, intensity, space, and time. Compared to static mechanical signals such as stiffness, porosity, and topography, the current understanding on the effects of dynamic mechanical stimulations on cells remains limited, attributing to a lack of access to devices, the complexity of experimental set-up, and data interpretation. Yet, in the pursuit of emerging translational applications (e.g., cell manufacturing for clinical treatment), it is crucial to understand how cells respond to a variety of dynamic forces that are omnipresent in vivo so that they can be exploited to enhance manufacturing and therapeutic outcomes. With a rising appreciation of the extracellular matrix (ECM) as a key regulator of biofunctions, researchers have bioengineered a suite of ECM-mimicking hydrogels, which can be fine-tuned with spatiotemporal mechanical cues to model complex static and dynamic mechanical profiles. This review first discusses how mechanical stimuli may impact different cellular components and the various mechanobiology pathways involved. Then, how hydrogels can be designed to incorporate static and dynamic mechanical parameters to influence cell behaviors are described. The Scopus database is also used to analyze the relative strength in evidence, ranging from strong to weak, based on number of published literatures, associated citations, and treatment significance. Additionally, the impacts of static and dynamic mechanical stimulations on clinically relevant cell types including mesenchymal stem cells, fibroblasts, and immune cells, are evaluated. The aim is to draw attention to the paucity of studies on the effects of dynamic mechanical stimuli on cells, as well as to highlight the potential of using a cocktail of various types and intensities of mechanical stimulations to influence cell fates (similar to the concept of biochemical cocktail to direct cell fate). It is envisioned that this progress report will inspire more exciting translational development of mechanoresponsive hydrogels for biomedical applications.
Collapse
Affiliation(s)
- Yufeng Shou
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
- Institute for Health Innovation and TechnologyNational University of SingaporeSingapore117599Singapore
| | - Xin Yong Teo
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
| | - Kenny Zhuoran Wu
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
| | - Bingyu Bai
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
| | - Arun R. K. Kumar
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
- Institute for Health Innovation and TechnologyNational University of SingaporeSingapore117599Singapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore117597Singapore
| | - Jessalyn Low
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
| | - Zhicheng Le
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
- Institute for Health Innovation and TechnologyNational University of SingaporeSingapore117599Singapore
| | - Andy Tay
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
- Institute for Health Innovation and TechnologyNational University of SingaporeSingapore117599Singapore
- NUS Tissue Engineering ProgramNational University of SingaporeSingapore117510Singapore
| |
Collapse
|
36
|
Antonelli F. 3D Cell Models in Radiobiology: Improving the Predictive Value of In Vitro Research. Int J Mol Sci 2023; 24:10620. [PMID: 37445795 DOI: 10.3390/ijms241310620] [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: 05/26/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Cancer is intrinsically complex, comprising both heterogeneous cellular composition and extracellular matrix. In vitro cancer research models have been widely used in the past to model and study cancer. Although two-dimensional (2D) cell culture models have traditionally been used for cancer research, they have many limitations, such as the disturbance of interactions between cellular and extracellular environments and changes in cell morphology, polarity, division mechanism, differentiation and cell motion. Moreover, 2D cell models are usually monotypic. This implies that 2D tumor models are ineffective at accurately recapitulating complex aspects of tumor cell growth, as well as their radiation responses. Over the past decade there has been significant uptake of three-dimensional (3D) in vitro models by cancer researchers, highlighting a complementary model for studies of radiation effects on tumors, especially in conjunction with chemotherapy. The introduction of 3D cell culture approaches aims to model in vivo tissue interactions with radiation by positioning itself halfway between 2D cell and animal models, and thus opening up new possibilities in the study of radiation response mechanisms of healthy and tumor tissues.
Collapse
Affiliation(s)
- Francesca Antonelli
- Laboratory of Biomedical Technologies, Division of Health Protection Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), 00123 Rome, Italy
| |
Collapse
|
37
|
Villanueva-Flores F, Garcia-Atutxa I, Santos A, Armendariz-Borunda J. Toward a New Generation of Bio-Scaffolds for Neural Tissue Engineering: Challenges and Perspectives. Pharmaceutics 2023; 15:1750. [PMID: 37376198 DOI: 10.3390/pharmaceutics15061750] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/04/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Neural tissue engineering presents a compelling technological breakthrough in restoring brain function, holding immense promise. However, the quest to develop implantable scaffolds for neural culture that fulfill all necessary criteria poses a remarkable challenge for material science. These materials must possess a host of desirable characteristics, including support for cellular survival, proliferation, and neuronal migration and the minimization of inflammatory responses. Moreover, they should facilitate electrochemical cell communication, display mechanical properties akin to the brain, emulate the intricate architecture of the extracellular matrix, and ideally allow the controlled release of substances. This comprehensive review delves into the primary requisites, limitations, and prospective avenues for scaffold design in brain tissue engineering. By offering a panoramic overview, our work aims to serve as an essential resource, guiding the creation of materials endowed with bio-mimetic properties, ultimately revolutionizing the treatment of neurological disorders by developing brain-implantable scaffolds.
Collapse
Affiliation(s)
- Francisca Villanueva-Flores
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Campus Chihuahua, Av. Heroico Colegio Militar 4700, Nombre de Dios, Chihuahua 31300, Chihuahua, Mexico
| | - Igor Garcia-Atutxa
- Máster en Bioinformática y Bioestadística, Universitat Oberta de Catalunya, Rambla del Poblenou, 156, 08018 Barcelona, Spain
| | - Arturo Santos
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Campus Guadalajara, Av. Gral Ramón Corona No 2514, Colonia Nuevo México, Zapopan 45201, Jalisco, Mexico
| | - Juan Armendariz-Borunda
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Campus Guadalajara, Av. Gral Ramón Corona No 2514, Colonia Nuevo México, Zapopan 45201, Jalisco, Mexico
- Instituto de Biología Molecular en Medicina y Terapia Génica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Independencia Oriente, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
38
|
Jain P, Rimal R, Möller M, Singh S. Topographical influence of electrospun basement membrane mimics on formation of cellular monolayer. Sci Rep 2023; 13:8382. [PMID: 37225757 DOI: 10.1038/s41598-023-34934-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 05/10/2023] [Indexed: 05/26/2023] Open
Abstract
Functional unit of many organs like lung, kidney, intestine, and eye have their endothelial and epithelial monolayers physically separated by a specialized extracellular matrix called the basement membrane. The intricate and complex topography of this matrix influences cell function, behavior and overall homeostasis. In vitro barrier function replication of such organs requires mimicking of these native features on an artificial scaffold system. Apart from chemical and mechanical features, the choice of nano-scale topography of the artificial scaffold is integral, however its influence on monolayer barrier formation is unclear. Though studies have reported improved single cell adhesion and proliferation in presence of pores or pitted topology, corresponding influence on confluent monolayer formation is not well reported. In this work, basement membrane mimic with secondary topographical cues is developed and its influence on single cells and their monolayers is investigated. We show that single cells cultured on fibers with secondary cues form stronger focal adhesions and undergo increased proliferation. Counterintuitively, absence of secondary cues promoted stronger cell-cell interaction in endothelial monolayers and promoted formation of integral tight barriers in alveolar epithelial monolayers. Overall, this work highlights the importance of choice of scaffold topology to develop basement barrier function in in vitro models.
Collapse
Affiliation(s)
- Puja Jain
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074, Aachen, Germany
| | - Rahul Rimal
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074, Aachen, Germany
- Max Planck Institute for Medical Research (MPImF), Jahnstrasse 29, 69120, Heidelberg, Germany
| | - Martin Möller
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074, Aachen, Germany
| | - Smriti Singh
- Max Planck Institute for Medical Research (MPImF), Jahnstrasse 29, 69120, Heidelberg, Germany.
| |
Collapse
|
39
|
Yang Q, Miao Y, Banerjee P, Hourwitz MJ, Hu M, Qing Q, Iglesias PA, Fourkas JT, Losert W, Devreotes PN. Nanotopography modulates intracellular excitable systems through cytoskeleton actuation. Proc Natl Acad Sci U S A 2023; 120:e2218906120. [PMID: 37126708 PMCID: PMC10175780 DOI: 10.1073/pnas.2218906120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 03/21/2023] [Indexed: 05/03/2023] Open
Abstract
Cellular sensing of most environmental cues involves receptors that affect a signal-transduction excitable network (STEN), which is coupled to a cytoskeletal excitable network (CEN). We show that the mechanism of sensing of nanoridges is fundamentally different. CEN activity occurs preferentially on nanoridges, whereas STEN activity is constrained between nanoridges. In the absence of STEN, waves disappear, but long-lasting F-actin puncta persist along the ridges. When CEN is suppressed, wave propagation is no longer constrained by nanoridges. A computational model reproduces these experimental observations. Our findings indicate that nanotopography is sensed directly by CEN, whereas STEN is only indirectly affected due to a CEN-STEN feedback loop. These results explain why texture sensing is robust and acts cooperatively with multiple other guidance cues in complex, in vivo microenvironments.
Collapse
Affiliation(s)
- Qixin Yang
- Department of Physics, University of Maryland, College Park, MD20742
- Institute of Physical Science and Technology, University of Maryland, College Park, MD20742
| | - Yuchuan Miao
- Department of Cell Biology, Johns Hopkins University, Baltimore, MD21205
| | - Parijat Banerjee
- Department of Physics & Astronomy, Johns Hopkins University, Baltimore, MD21218
| | - Matt J. Hourwitz
- Department of Chemistry & Biochemistry, University of Maryland, College Park, MD20742
| | - Minxi Hu
- School of Molecular Sciences, Arizona State University, Tempe, AZ85287
| | - Quan Qing
- Department of Physics, Arizona State University, Tempe, AZ85287
- Biodesign Institute, Arizona State University, Tempe, AZ85287
| | - Pablo A. Iglesias
- Department of Cell Biology, Johns Hopkins University, Baltimore, MD21205
- Department of Electrical & Computer Engineering, Johns Hopkins University, Baltimore, MD21218
| | - John T. Fourkas
- Institute of Physical Science and Technology, University of Maryland, College Park, MD20742
- Department of Chemistry & Biochemistry, University of Maryland, College Park, MD20742
| | - Wolfgang Losert
- Department of Physics, University of Maryland, College Park, MD20742
- Institute of Physical Science and Technology, University of Maryland, College Park, MD20742
| | - Peter N. Devreotes
- Department of Cell Biology, Johns Hopkins University, Baltimore, MD21205
| |
Collapse
|
40
|
Miyoshi H, Yamazaki M, Fujie H, Kidoaki S. Guideline for design of substrate stiffness for mesenchymal stem cell culture based on heterogeneity of YAP and RUNX2 responses. Biophys Physicobiol 2023; 20:e200018. [PMID: 38496240 PMCID: PMC10941962 DOI: 10.2142/biophysico.bppb-v20.0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 04/17/2023] [Indexed: 03/19/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have the potential for self-renewal and multipotency to differentiate into various lineages. Thus, they are of great interest in regenerative medicine as a cell source for tissue engineering. Substrate stiffness is one of the most extensively studied exogenous physical factors; however, consistent results have not always been reported for controlling MSCs. Conventionally used stiff culture substrates, such as tissue-culture polystyrene and glass, enhance nuclear localization of a mechanotransducer YAP and a pre-osteogenic transcription factor RUNX2, and bias MSCs towards the osteogenic lineage, even without osteogenic-inducing soluble factors. The mechanosensitive nature and intrinsic heterogeneity present challenges for obtaining reproducible results. This review summarizes the heterogeneity in human MSC response, specifically, nuclear/cytoplasmic localization changes in the mechanotransducer yes-associated protein (YAP) and the osteogenic transcription factor RUNX2, in response to substrate stiffness. In addition, a perspective on the intracellular factors attributed to response heterogeneity is discussed. The optimal range of stiffness parameters, Young's modulus, for MSC expansion culture to suppress osteogenic differentiation bias through the suppression of YAP and RUNX2 nuclear localization, and cell cycle progression is likely to be surprisingly narrow for a cell population from an identical donor and vary among cell populations from different donors. We believe that characterization of the heterogeneity of MSCs and understanding their biological meaning is an exciting research direction to establish guidelines for the design of culture substrates for the sophisticated control of MSC properties.
Collapse
Affiliation(s)
- Hiromi Miyoshi
- Department of Mechanical Systems Engineering, Graduate School of Systems Design, Tokyo Metropolitan University, Hachioji, Tokyo 192-0397, Japan
| | - Masashi Yamazaki
- Department of Mechanical Systems Engineering, Graduate School of Systems Design, Tokyo Metropolitan University, Hachioji, Tokyo 192-0397, Japan
| | - Hiromichi Fujie
- Department of Mechanical Systems Engineering, Graduate School of Systems Design, Tokyo Metropolitan University, Hachioji, Tokyo 192-0397, Japan
| | - Satoru Kidoaki
- Institute for Materials Chemistry and Engineering, Kyushu University, Fukuoka 819-0395, Japan
| |
Collapse
|
41
|
Pramotton FM, Cousin L, Roy T, Giampietro C, Cecchini M, Masciullo C, Ferrari A, Poulikakos D. Accelerated epithelial layer healing induced by tactile anisotropy in surface topography. SCIENCE ADVANCES 2023; 9:eadd1581. [PMID: 37027475 PMCID: PMC10081848 DOI: 10.1126/sciadv.add1581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 03/03/2023] [Indexed: 06/19/2023]
Abstract
Mammalian cells respond to tactile cues from topographic elements presented by the substrate. Among these, anisotropic features distributed in an ordered manner give directionality. In the extracellular matrix, this ordering is embedded in a noisy environment altering the contact guidance effect. To date, it is unclear how cells respond to topographical signals in a noisy environment. Here, using rationally designed substrates, we report morphotaxis, a guidance mechanism enabling fibroblasts and epithelial cells to move along gradients of topographic order distortion. Isolated cells and cell ensembles perform morphotaxis in response to gradients of different strength and directionality, with mature epithelia integrating variations of topographic order over hundreds of micrometers. The level of topographic order controls cell cycle progression, locally delaying or promoting cell proliferation. In mature epithelia, the combination of morphotaxis and noise-dependent distributed proliferation provides a strategy to enhance wound healing as confirmed by a mathematical model capturing key elements of the process.
Collapse
Affiliation(s)
- Francesca Michela Pramotton
- Experimental Continuum Mechanics Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Zurich 8092, Switzerland
- EMPA, Swiss Federal Laboratories for Material Science and Technologies, Überlandstrasse 129, Dübendorf 8600, Switzerland
| | - Lucien Cousin
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Tamal Roy
- Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, Zurich CH-8092, Switzerland
| | - Costanza Giampietro
- Experimental Continuum Mechanics Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Zurich 8092, Switzerland
- EMPA, Swiss Federal Laboratories for Material Science and Technologies, Überlandstrasse 129, Dübendorf 8600, Switzerland
| | - Marco Cecchini
- NEST, Istituto Nanoscienze CNR and Scuola Normale Superiore, Pisa 56127, Italy
| | - Cecilia Masciullo
- NEST, Istituto Nanoscienze CNR and Scuola Normale Superiore, Pisa 56127, Italy
| | - Aldo Ferrari
- EMPA, Swiss Federal Laboratories for Material Science and Technologies, Überlandstrasse 129, Dübendorf 8600, Switzerland
- Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, Zurich CH-8092, Switzerland
| | - Dimos Poulikakos
- Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, Zurich CH-8092, Switzerland
| |
Collapse
|
42
|
Wang J, Li H, Fu S, Su Y. Porous BCP ceramics with nanoscale whisker structure accelerate bone regeneration by regulating inflammatory response. BIOMATERIALS ADVANCES 2023; 147:213313. [PMID: 36753873 DOI: 10.1016/j.bioadv.2023.213313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 01/07/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023]
Abstract
Inflammation-induced by biomaterials is a critical event to determine the success and efficiency of tissue repair. Macrophages are a major population that participates the biomaterial induced inflammation. The response of macrophages depends on the characteristics of biomaterials, thus causing a cascade reaction in subsequent biological processes. In this study, porous biphase calcium phosphate (BCP) ceramics with the different surface structures were constructed to compare the effect of surface structure on bone generation potential, and further reveal the inflammation-involved mechanism. Our results demonstrated that macrophages on three ceramics showed distinct morphologies and spreading areas. The nanoscale whisker structure did induce more bone generation in the mice thigh muscle. The in vitro result revealed that nanoscale whisker structure could drive macrophage polarization towards M1-like phenotype, indicated by a higher expression of pro-inflammatory specific markers (iNOS and CCR7), and mass secretion of TNF-α. Further research indicated that additional TNF-α could promote the osteogenic differentiation of mesenchymal stem cells (MSCs). However, excess addition of TNF-α showed an opposite effect on the osteogenic differentiation of MSCs by initiating the NF-κB signaling pathway, which suppresses the osteogenesis process.
Collapse
Affiliation(s)
- Jing Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Huishan Li
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Shijia Fu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yangyang Su
- State Key Laboratory of Solidification Processing, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
43
|
Wang K, Smith SH, Iijima H, Hettinger ZR, Mallepally A, Shroff SG, Ambrosio F. Bioengineered 3D Skeletal Muscle Model Reveals Complement 4b as a Cell-Autonomous Mechanism of Impaired Regeneration with Aging. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2207443. [PMID: 36650030 DOI: 10.1002/adma.202207443] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 12/11/2022] [Indexed: 05/17/2023]
Abstract
A mechanistic understanding of cell-autonomous skeletal muscle changes after injury can lead to novel interventions to improve functional recovery in an aged population. However, major knowledge gaps persist owing to limitations of traditional biological aging models. 2D cell culture represents an artificial environment, while aging mammalian models are contaminated by influences from non-muscle cells and other organs. Here, a 3D muscle aging system is created to overcome the limitations of these traditional platforms. It is shown that old muscle constructs (OMC) manifest a sarcopenic phenotype, as evidenced by hypotrophic myotubes, reduced contractile function, and decreased regenerative capacity compared to young muscle constructs. OMC also phenocopy the regenerative responses of aged muscle to two interventions, pharmacological and biological. Interrogation of muscle cell-specific mechanisms that contribute to impaired regeneration over time further reveals that an aging-induced increase of complement component 4b (C4b) delays muscle progenitor cell amplification and impairs functional recovery. However, administration of complement factor I, a C4b inactivator, improves muscle regeneration in vitro and in vivo, indicating that C4b inhibition may be a novel approach to enhance aged muscle repair. Collectively, the model herein exhibits capabilities to study cell-autonomous changes in skeletal muscle during aging, regeneration, and intervention.
Collapse
Affiliation(s)
- Kai Wang
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Charlestown, MA, 02129, USA
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, 02115, USA
| | - Stephen H Smith
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Hirotaka Iijima
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Zachary R Hettinger
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Charlestown, MA, 02129, USA
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, 02115, USA
- Department of Medicine, Division of Geriatric Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Adarsh Mallepally
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Sanjeev G Shroff
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Fabrisia Ambrosio
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Charlestown, MA, 02129, USA
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, 02115, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| |
Collapse
|
44
|
Cheng X, Zhang M, Xie W, Ma X, Yang X, Cai Y. Well-aligned three-dimensional silk fibroin protein scaffold for orientation regulation of cells. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2023:1-17. [PMID: 36745185 DOI: 10.1080/09205063.2023.2177828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The similar characteristics of biomaterials to the extracellular matrix are essential for efficient tissue repair through dictating cell behaviors. But the scaffold fabrication with complex shapes and controlled alignment have proven to be a difficult task. Herein, a well-designed three-dimensional silk fibroin scaffold is fabricated through ice template technology. The effect of the silk fibroin protein concentration and the freezing temperature on the microstructure and mechanical properties of scaffolds are investigated systematically. Cells behavior mediated by the obtained silk fibroin scaffolds is detected. The results show that the protein concentration plays a vital role in microstructure and scaffold strength. A well-aligned scaffold can be obtained when silk fibroin solution is kept at 12 wt%, which holds the highest mechanical properties. The pore size can be further adjusted in the range of 5-80 µm by changing the freezing temperature from -60 to -196 °C. The well-oriented scaffold with the appropriate pore size of 10-20 µm has the best ability to guide cell alignment. The resulting scaffolds provide an excellent matrix to guide cells behaviors and have a potential application in tissue engineering.
Collapse
Affiliation(s)
- Xiuwen Cheng
- The Key Laboratory of Advanced Textile Materials and Manufacturing Technology of Ministry of Education, National Engineering Lab for Textile Fiber Materials and Processing Technology, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, China
| | - Minghao Zhang
- The Key Laboratory of Advanced Textile Materials and Manufacturing Technology of Ministry of Education, National Engineering Lab for Textile Fiber Materials and Processing Technology, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, China
| | - Wenjiao Xie
- The Key Laboratory of Advanced Textile Materials and Manufacturing Technology of Ministry of Education, National Engineering Lab for Textile Fiber Materials and Processing Technology, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiaoyu Ma
- The Key Laboratory of Advanced Textile Materials and Manufacturing Technology of Ministry of Education, National Engineering Lab for Textile Fiber Materials and Processing Technology, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiaogang Yang
- Academy of Science and Technology, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yurong Cai
- The Key Laboratory of Advanced Textile Materials and Manufacturing Technology of Ministry of Education, National Engineering Lab for Textile Fiber Materials and Processing Technology, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
45
|
Wang K, Man K, Liu J, Meckes B, Yang Y. Dissecting Physical and Biochemical Effects in Nanotopographical Regulation of Cell Behavior. ACS NANO 2023; 17:2124-2133. [PMID: 36668987 DOI: 10.1021/acsnano.2c08075] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Regulating cell behavior using nanotopography has been widely implemented. To facilitate cell adhesion, physical nanotopography is usually coated with adhesive proteins such as fibronectin (FN). However, the confounding effects of physical and biochemical cues of nanotopography hinder the understanding of nanotopography in regulating cell behavior, which ultimately limits the biomedical applications of nanotopography. To delineate the roles of the physical and biochemical cues in cell regulation, we fabricate substrates that have either the same physical nanotopography but different biochemical (FN) nanopatterns or identical FN nanopatterns but different physical nanotopographies. We then examine the influences of physical and biochemical cues of nanotopography on spreading, nuclear deformation, mechanotransduction, and function of human mesenchymal stem cells (hMSCs). Our results reveal that physical topographies, especially nanogratings, dominantly control cell spreading, YAP localization, proliferation, and differentiation of hMSCs. However, biochemical FN nanopatterns affect hMSC elongation, YAP intracellular localization, and lamin a/c (LAMAC) expression. Furthermore, we find that physical nanogratings induce nanoscale curvature of nuclei at the basal side, which attenuates the osteogenic differentiation of hMSCs. Collectively, our study highlights the dominant effect of physical nanotopography in regulating stem cell functions, while suggesting that fine-tuning of cell behavior can be achieved through altering the presentation of biochemical cues on substrate surfaces.
Collapse
Affiliation(s)
- Kai Wang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Kun Man
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Jiafeng Liu
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Brian Meckes
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| |
Collapse
|
46
|
Cheng Y, Pang SW. Effects of nanopillars and surface coating on dynamic traction force. MICROSYSTEMS & NANOENGINEERING 2023; 9:6. [PMID: 36620393 PMCID: PMC9814462 DOI: 10.1038/s41378-022-00473-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 10/11/2022] [Accepted: 11/04/2022] [Indexed: 06/17/2023]
Abstract
The extracellular matrix serves as structural support for cells and provides biophysical and biochemical cues for cell migration. Topography, material, and surface energy can regulate cell migration behaviors. Here, the responses of MC3T3-E1 cells, including migration speed, morphology, and spreading on various platform surfaces, were investigated. Polydimethylsiloxane (PDMS) micropost sensing platforms with nanopillars, silicon oxide, and titanium oxide on top of the microposts were fabricated, and the dynamic cell traction force during migration was monitored. The relationships between various platform surfaces, migration behaviors, and cell traction forces were studied. Compared with the flat PDMS surface, cells on silicon oxide and titanium oxide surfaces showed reduced mobility and less elongation. On the other hand, cells on the nanopillar surface showed more elongation and a higher migration speed than cells on silicon oxide and titanium oxide surfaces. MC3T3-E1 cells on microposts with nanopillars exerted a larger traction force than those on flat PDMS microposts and had more filopodia and long protrusions. Understanding the relationships between platform surface condition, migration behavior, and cell traction force can potentially lead to better control of cell migration in biomaterials capable of promoting tissue repair and regeneration.
Collapse
Affiliation(s)
- Yijun Cheng
- Department of Electrical Engineering, City University of Hong Kong, Kowloon, Hong Kong, China
- Centre for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Stella W. Pang
- Department of Electrical Engineering, City University of Hong Kong, Kowloon, Hong Kong, China
- Centre for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong, China
| |
Collapse
|
47
|
Cheng K, Huang Z, Wang P, Sun L, Ghasemi H, Ardebili H, Karim A. Antibacterial flexible triboelectric nanogenerator via capillary force lithography. J Colloid Interface Sci 2023; 630:611-622. [DOI: 10.1016/j.jcis.2022.10.129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/03/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
|
48
|
Kumari S, Katiyar S, Darshna, Anand A, Singh D, Singh BN, Mallick SP, Mishra A, Srivastava P. Design strategies for composite matrix and multifunctional polymeric scaffolds with enhanced bioactivity for bone tissue engineering. Front Chem 2022; 10:1051678. [PMID: 36518978 PMCID: PMC9742444 DOI: 10.3389/fchem.2022.1051678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/14/2022] [Indexed: 09/19/2023] Open
Abstract
Over the past few decades, various bioactive material-based scaffolds were investigated and researchers across the globe are actively involved in establishing a potential state-of-the-art for bone tissue engineering applications, wherein several disciplines like clinical medicine, materials science, and biotechnology are involved. The present review article's main aim is to focus on repairing and restoring bone tissue defects by enhancing the bioactivity of fabricated bone tissue scaffolds and providing a suitable microenvironment for the bone cells to fasten the healing process. It deals with the various surface modification strategies and smart composite materials development that are involved in the treatment of bone tissue defects. Orthopaedic researchers and clinicians constantly focus on developing strategies that can naturally imitate not only the bone tissue architecture but also its functional properties to modulate cellular behaviour to facilitate bridging, callus formation and osteogenesis at critical bone defects. This review summarizes the currently available polymeric composite matrices and the methods to improve their bioactivity for bone tissue regeneration effectively.
Collapse
Affiliation(s)
- Shikha Kumari
- School of Biochemical Engineering, IIT BHU, Varanasi, India
| | - Soumya Katiyar
- School of Biochemical Engineering, IIT BHU, Varanasi, India
| | - Darshna
- School of Biochemical Engineering, IIT BHU, Varanasi, India
| | - Aditya Anand
- School of Biochemical Engineering, IIT BHU, Varanasi, India
| | - Divakar Singh
- School of Biochemical Engineering, IIT BHU, Varanasi, India
| | - Bhisham Narayan Singh
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sarada Prasanna Mallick
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation, Vaddeswaram, Andhra Pradesh, India
| | - Abha Mishra
- School of Biochemical Engineering, IIT BHU, Varanasi, India
| | | |
Collapse
|
49
|
Effect of Piezoelectric BaTiO 3 Filler on Mechanical and Magnetoelectric Properties of Zn 0.25Co 0.75Fe 2O 4/PVDF-TrFE Composites. Polymers (Basel) 2022; 14:polym14224807. [PMID: 36432934 PMCID: PMC9695481 DOI: 10.3390/polym14224807] [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: 10/12/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022] Open
Abstract
Polymer-based multiferroics, combining magnetic and piezoelectric properties, are studied experimentally-from synthesis to multi-parameter characterization-in view of their prospects for fabricating biocompatible scaffolds. The main advantage of these systems is facile generation of mechanical deformations and electric signals in response to external magnetic fields. Herein, we address the composites based on PVDF-TrFE polymer matrices filled with a combination of piezoelectric (BaTiO3, BTO) and/or ferrimagnetic (Zn0.25Co0.75Fe2O4, ZCFO) particles. It is shown that the presence of BTO micron-size particles favors stripe-type structuring of the ZCFO filler and enhances the magnetoelectric response of the sample up to 18.6 mV/(cm∙Oe). Besides that, the admixing of BTO particles is crucial because the mechanical properties of the composite filled with only ZCFO is much less efficient in transforming magnetic excitations into the mechanical and electric responses. Attention is focused on the local surfacial mechanical properties since those, to a great extent, determine the fate of stem cells cultivated on these surfaces. The nano-indentation tests are accomplished with the aid of scanning probe microscopy technique. With their proven suitable mechanical properties, a high level of magnetoelectric conversion and also biocompatibility, the composites of the considered type are enticing as the materials for multiferroic-based polymer scaffolds.
Collapse
|
50
|
Schmitz C, Pepelanova I, Ude C, Lavrentieva A. Studies on oxygen availability and the creation of natural and artificial oxygen gradients in gelatin-methacryloyl hydrogel 3D cell culture. J Tissue Eng Regen Med 2022; 16:977-986. [PMID: 35962761 DOI: 10.1002/term.3344] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 06/19/2022] [Accepted: 07/26/2022] [Indexed: 12/15/2022]
Abstract
Three-dimensional (3D) cultivation platforms allow the creation of cell models, which more closely resemble in vivo-like cell behavior. Therefore, 3D cell culture platforms have started to replace conventional two-dimensional (2D) cultivation techniques in many fields. Besides the advantages of 3D culture, there are also some challenges: cultivation in 3D often results in an inhomogeneous microenvironment and therefore unique cultivation conditions for each cell inside the construct. As a result, the analysis and precise control over the singular cell state is limited in 3D. In this work, we address these challenges by exploring ways to monitor oxygen concentrations in gelatin methacryloyl (GelMA) 3D hydrogel culture at the cellular level using hypoxia reporter cells and deep within the construct using a non-invasive optical oxygen sensing spot. We could show that the appearance of oxygen limitations is more prominent in softer GelMA-hydrogels, which enable better cell spreading. Beyond demonstrating novel or space-resolved techniques of visualizing oxygen availability in hydrogel constructs, we also describe a method to create a stable and controlled oxygen gradient throughout the construct using a 3D printed flow-through chamber.
Collapse
Affiliation(s)
- Carola Schmitz
- Institute of Technical Chemistry, Gottfried Wilhelm Leibniz University Hannover, Hannover, Germany
| | - Iliyana Pepelanova
- Institute of Technical Chemistry, Gottfried Wilhelm Leibniz University Hannover, Hannover, Germany
| | - Christian Ude
- Institute of Technical Chemistry, Gottfried Wilhelm Leibniz University Hannover, Hannover, Germany
| | - Antonina Lavrentieva
- Institute of Technical Chemistry, Gottfried Wilhelm Leibniz University Hannover, Hannover, Germany
| |
Collapse
|