1
|
Gao L, Dai X, Wu Y, Wang Y, Cheng L, Yan LT. Self-Assembly at Curved Biointerfaces. ACS NANO 2024; 18:30184-30210. [PMID: 39453716 DOI: 10.1021/acsnano.4c09675] [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: 10/27/2024]
Abstract
Most of the biological interfaces are curved. Understanding the organizational structures and interaction patterns at such curved biointerfaces is therefore crucial not only for deepening our comprehension of the principles that govern life processes but also for designing and developing targeted drugs aimed at diseased cells and tissues. Despite the considerable efforts dedicated to this area of research, our understanding of curved biological interfaces is still limited. Many aspects of these interfaces remain elusive, presenting both challenges and opportunities for further exploration. In this review, we summarize the structural characteristics of biological interfaces found in nature, the current research status of materials associated with curved biointerfaces, and the theoretical advancements achieved to date. Finally, we outline future trends and challenges in the theoretical and technological development of curved biointerfaces. By addressing these challenges, people could bridge the knowledge gap and unlock the full potential of curved biointerfaces for scientific and technological advancements, ultimately benefiting various fields and improving human health and well-being.
Collapse
Affiliation(s)
- Lijuan Gao
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Xiaobin Dai
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Yibo Wu
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Yuming Wang
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Linghe Cheng
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Li-Tang Yan
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| |
Collapse
|
2
|
Vishwakarma A, Sinha N. Determination of the Optimum Architecture of Additively Manufactured Magnetic Bioactive Glass Scaffolds for Bone Tissue Engineering and Drug-Delivery Applications. ACS APPLIED BIO MATERIALS 2024; 7:6847-6864. [PMID: 39382202 DOI: 10.1021/acsabm.4c00995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
For better bone regeneration, precise control over the architecture of the scaffolds is necessary. Because the shape of the pore may affect the bone regeneration, therefore, additive manufacturing has been used in this study to fabricate magnetic bioactive glass (MBG) scaffolds with three different architectures, namely, grid, gyroid, and Schwarz D surface with 15 × 15 × 15 mm3 dimensions and 70% porosity. These scaffolds have been fabricated using an in-house-developed material-extrusion-based additive manufacturing system. The composition of bioactive glass was selected as 45% SiO2, 20% Na2O, 23% CaO, 6% P2O5, 2.5% B2O3, 1% ZnO, 2% MgO, and 0.5% CaF2 (wt %), and additionally 0.4 wt % of iron carbide nanoparticles were incorporated. Afterward, MBG powder was mixed with a 25% (w/v) Pluronic F-127 solution to prepare a slurry for fabricating scaffolds at 23% relative humidity. The morphological characterization using microcomputed tomography revealed the appropriate pore size distribution and interconnectivity of the scaffolds. The compressive strengths of the fabricated grid, gyroid, and Schwarz D scaffolds were found to be 14.01 ± 1.01, 10.78 ± 1.5, and 12.57 ± 1.2 MPa, respectively. The in vitro study was done by immersing the MBG scaffolds in simulated body fluid for 1, 3, 7, and 14 days. Darcy's law, which describes the flow through porous media, was used to evaluate the permeability of the scaffolds. Furthermore, an anticancer drug (Mitomycin C) was loaded onto these scaffolds, wherein these scaffolds depicted good release behavior. Overall, gyroid-structured scaffolds were found to be the most suitable among the three scaffolds considered in this study for bone tissue engineering and drug-delivery applications.
Collapse
Affiliation(s)
- Ashok Vishwakarma
- Department of Mechanical Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Niraj Sinha
- Department of Mechanical Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| |
Collapse
|
3
|
Roy A, Zhang Z, Eiken MK, Shi A, Pena-Francesch A, Loebel C. Programmable Tissue Folding Patterns in Structured Hydrogels. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2300017. [PMID: 36961361 PMCID: PMC10518030 DOI: 10.1002/adma.202300017] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 02/24/2023] [Indexed: 05/17/2023]
Abstract
Folding of mucosal tissues, such as the tissue within the epithelium of the upper respiratory airways, is critical for organ function. Studying the influence of folded tissue patterns on cellular function is challenging mainly due to the lack of suitable cell culture platforms that can recreate dynamic tissue folding in vitro. Here, a bilayer hydrogel folding system, composed of alginate/polyacrylamide double-network (DN) and hyaluronic acid (HA) hydrogels, to generate static folding patterns based on mechanical instabilities, is described. By encapsulating human fibroblasts into patterned HA hydrogels, human bronchial epithelial cells form a folded pseudostratified monolayer. Using magnetic microparticles, DN hydrogels reversibly fold into pre-defined patterns and enable programmable on-demand folding of cell-laden hydrogel systems upon applying a magnetic field. This hydrogel construction provides a dynamic culture system for mimicking tissue folding in vitro, which is extendable to other cell types and organ systems.
Collapse
Affiliation(s)
- Avinava Roy
- Department of Materials Science & Engineering, University of Michigan, North Campus Research Complex, 2800 Plymouth Rd, Ann Arbor, MI, 48109, USA
| | - Zenghao Zhang
- Department of Materials Science & Engineering, University of Michigan, North Campus Research Complex, 2800 Plymouth Rd, Ann Arbor, MI, 48109, USA
| | - Madeline K Eiken
- Department of Biomedical Engineering, University of Michigan, Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI, 48109, USA
| | - Alan Shi
- Department of Materials Science & Engineering, University of Michigan, North Campus Research Complex, 2800 Plymouth Rd, Ann Arbor, MI, 48109, USA
| | - Abdon Pena-Francesch
- Department of Materials Science & Engineering, University of Michigan, North Campus Research Complex, 2800 Plymouth Rd, Ann Arbor, MI, 48109, USA
| | - Claudia Loebel
- Department of Materials Science & Engineering, University of Michigan, North Campus Research Complex, 2800 Plymouth Rd, Ann Arbor, MI, 48109, USA
- Department of Biomedical Engineering, University of Michigan, Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI, 48109, USA
| |
Collapse
|
4
|
Tang RC, Shang L, Scumpia PO, Di Carlo D. Injectable Microporous Annealed Crescent-Shaped (MAC) Particle Hydrogel Scaffold for Enhanced Cell Infiltration. Adv Healthc Mater 2024; 13:e2302477. [PMID: 37985462 PMCID: PMC11102933 DOI: 10.1002/adhm.202302477] [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: 07/31/2023] [Revised: 11/10/2023] [Indexed: 11/22/2023]
Abstract
Hydrogels are widely used for tissue engineering applications to support cellular growth, yet the tightly woven structure often restricts cell infiltration and expansion. Consequently, granular hydrogels with microporous architectures have emerged as a new class of biomaterial. Particularly, the development of microporous annealed particle (MAP) hydrogel scaffolds has shown improved stability and integration with host tissue. However, the predominant use of spherically shaped particles limits scaffold porosity, potentially limiting the level of cell infiltration. Here, a novel microporous annealed crescent-shaped particle (MAC) scaffold that is predicted to have improved porosity and pore interconnectivity in silico is presented. With microfluidic fabrication, tunable cavity sizes that optimize interstitial void space features are achieved. In vitro, cells incorporated into MAC scaffolds form extensive 3D multicellular networks. In vivo, the injectable MAC scaffold significantly enhances cell infiltration compared to spherical MAP scaffolds, resulting in increased numbers of myofibroblasts and leukocytes present within the gel without relying on external biomolecular chemoattractants. The results shed light on the critical role of particle shape in cell recruitment, laying the foundation for MAC scaffolds as a next-generation granular hydrogel for diverse tissue engineering applications.
Collapse
Affiliation(s)
- Rui-Chian Tang
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Lily Shang
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Philip O Scumpia
- Division of Dermatology, Department of Medicine David Geffen School of Medicine University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Dermatology, VA Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA
- Jonsson Comprehensive Cancer Center University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Dino Di Carlo
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Mechanical and Aerospace Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- California Nano Systems Institute (CNSI) University of California Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
5
|
Saporito S, Panzetta V, Netti PA. Time and space modulation of substrate curvature to regulate cell mechanical identity. Acta Biomater 2024; 186:300-315. [PMID: 39127326 DOI: 10.1016/j.actbio.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/08/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Recently, a variety of microenvironmental biophysical stimuli have been proved to play a crucial role in regulating cell functions. Among them, morpho-physical cues, like curvature, are emerging as key regulators of cellular behavior. Changes in substrate curvature have been shown to impact the arrangement of Focal Adhesions (FAs), influencing the direction and intensity of cytoskeleton generated forces and resulting in an overall alteration of cell mechanical identity. In their native environment, cells encounter varying degrees of substrate curvature, and in specific organs, they are exposed to dynamic changes of curvature due to periodic tissue deformation. However, the mechanism by which cells perceive substrate curvature remains poorly understood. To this aim, a micro-pneumatic device was designed and implemented. This device enables the controlled application of substrate curvature, both statically and dynamically. Employing a combined experimental and simulative approach, human adipose-derived stem cells were exposed to controlled curvature intensity and frequency. During this exposure, measurements were taken on FAs extension and orientation, cytoskeleton organization and cellular/nuclear alignment. The data clearly indicated a significant influence of the substrate curvature on cell adhesion processes. These findings contribute to a better understanding of the mechanisms through which cells perceive and respond to substrate curvature signals. STATEMENT OF SIGNIFICANCE: This work is our contribution to the comprehension of substrate curvature's function as a crucial regulator of cell adhesion at the scale of focal adhesions and cell mechanical identity. In recent years, a large body of knowledge is continuously growing providing comprehension of the role of various microenvironmental biophysical stimuli in regulating cell functions. Nevertheless, little is known about the role of substrate curvature, in particular, when cells are exposed to this stimulus in a dynamic manner. To address the role of substrate curvature on cellular behavior, a micro-pneumatic device was designed and implemented. This device enables the controlled application of substrate curvature, both statically and dynamically. The experiment data made it abundantly evident that the substrate curvature had a major impact on the mechanisms involved in cell adhesion.
Collapse
Affiliation(s)
- Stefania Saporito
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Italy; Center for Advanced Biomaterials for Healthcare@CRIB, Istituto Italiano Di Tecnologia, Italy
| | - Valeria Panzetta
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Italy; Center for Advanced Biomaterials for Healthcare@CRIB, Istituto Italiano Di Tecnologia, Italy; Interdisciplinary research Center on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy
| | - Paolo Antonio Netti
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Italy; Center for Advanced Biomaterials for Healthcare@CRIB, Istituto Italiano Di Tecnologia, Italy; Interdisciplinary research Center on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy.
| |
Collapse
|
6
|
Wu Y, Liu P, Feng C, Cao Q, Xu X, Liu Y, Li X, Zhu X, Zhang X. 3D printing calcium phosphate ceramics with high osteoinductivity through pore architecture optimization. Acta Biomater 2024; 185:111-125. [PMID: 39002921 DOI: 10.1016/j.actbio.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
The osteoinductivity of 3D printed calcium phosphate (CaP) ceramics has a large gap compared with those prepared by conventional foaming methods, and improving the osteoinductivity of 3D printing CaP ceramics is crucial for successful application in bone regeneration. Pore architecture plays a critical role in osteoinductivity. In this study, CaP ceramics with a hexagonal close-packed (HCP) spherical pore structure were successfully fabricated using DLP printing technology. Additionally, octahedral (Octahedral), diamond (Diamond), and helical (Gyroid) structures were constructed with similar porosity and macropore diameter. CaP ceramics with the HCP structure exhibited higher compression strength (8.39 ± 1.82 MPa) and lower permeability (6.41 × 10-11 m2) compared to the Octahedral, Diamond, and Gyroid structures. In vitro cellular responses indicated that the macropore architecture strongly influenced the local growth rate of osteoblast-formed cell tissue; cells grew uniformly and formed circular rings in the HCP group. Furthermore, the HCP group promoted the expression of osteogenic genes and proteins more effectively than the other three groups. The outstanding osteoinductivity of the HCP group was confirmed in canine intramuscular implantation studies, where the new bone area reached up to 8.02 ± 1.94 % after a 10-week implantation. Additionally, the HCP group showed effective bone regeneration in repairing femoral condyle defects. Therefore, our findings suggest that 3D printed CaP bioceramics with an HCP structure promote osteoinductivity and can be considered as candidates for personalized precise treatment of bone defects in clinical applications. STATEMENT OF SIGNIFICANCE: 1. 3D printing BCP ceramics with high osteoinductivity were constructed through pore architecture optimization. 2. BCP ceramics with HCP structure exhibited relatively higher mechanical strength and lower permeability than those with Octahedral, Diamond and Gyroid structures. 3. BCP ceramics with HCP structure could promote the osteogenic differentiation of MC3T3-E1, and showed the superior in-vivo osteoinductivity and bone regeneration comparing with the other structures.
Collapse
Affiliation(s)
- Yonghao Wu
- National Engineering Research Center for Biomaterials, Med-X Center for Materials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Puxin Liu
- National Engineering Research Center for Biomaterials, Med-X Center for Materials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Cong Feng
- National Engineering Research Center for Biomaterials, Med-X Center for Materials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Quanle Cao
- National Engineering Research Center for Biomaterials, Med-X Center for Materials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xiujuan Xu
- National Engineering Research Center for Biomaterials, Med-X Center for Materials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China; Provincial Engineering Research Center for Biomaterials Genome of Sichuan, Sichuan University, Chengdu 610064, China
| | - Yunyi Liu
- National Engineering Research Center for Biomaterials, Med-X Center for Materials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xiangfeng Li
- National Engineering Research Center for Biomaterials, Med-X Center for Materials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China; Provincial Engineering Research Center for Biomaterials Genome of Sichuan, Sichuan University, Chengdu 610064, China.
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, Med-X Center for Materials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China; Provincial Engineering Research Center for Biomaterials Genome of Sichuan, Sichuan University, Chengdu 610064, China.
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Med-X Center for Materials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China; Provincial Engineering Research Center for Biomaterials Genome of Sichuan, Sichuan University, Chengdu 610064, China
| |
Collapse
|
7
|
Liao J, Timoshenko AB, Cordova DJ, Astudillo Potes MD, Gaihre B, Liu X, Elder BD, Lu L, Tilton M. Propelling Minimally Invasive Tissue Regeneration With Next-Era Injectable Pre-Formed Scaffolds. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2400700. [PMID: 38842622 DOI: 10.1002/adma.202400700] [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: 01/14/2024] [Revised: 05/12/2024] [Indexed: 06/07/2024]
Abstract
The growing aging population, with its associated chronic diseases, underscores the urgency for effective tissue regeneration strategies. Biomaterials play a pivotal role in the realm of tissue reconstruction and regeneration, with a distinct shift toward minimally invasive (MI) treatments. This transition, fueled by engineered biomaterials, steers away from invasive surgical procedures to embrace approaches offering reduced trauma, accelerated recovery, and cost-effectiveness. In the realm of MI tissue repair and cargo delivery, various techniques are explored. While in situ polymerization is prominent, it is not without its challenges. This narrative review explores diverse biomaterials, fabrication methods, and biofunctionalization for injectable pre-formed scaffolds, focusing on their unique advantages. The injectable pre-formed scaffolds, exhibiting compressibility, controlled injection, and maintained mechanical integrity, emerge as promising alternative solutions to in situ polymerization challenges. The conclusion of this review emphasizes the importance of interdisciplinary design facilitated by synergizing fields of materials science, advanced 3D biomanufacturing, mechanobiological studies, and innovative approaches for effective MI tissue regeneration.
Collapse
Affiliation(s)
- Junhan Liao
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Anastasia B Timoshenko
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Domenic J Cordova
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | | | - Bipin Gaihre
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | - Xifeng Liu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | - Benjamin D Elder
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Lichun Lu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | - Maryam Tilton
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
8
|
Josephson TO, Morgan EF. Mechanobiological optimization of scaffolds for bone tissue engineering. Biomech Model Mechanobiol 2024:10.1007/s10237-024-01880-0. [PMID: 39060881 DOI: 10.1007/s10237-024-01880-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024]
Abstract
Synthetic bone graft scaffolds aim to generate new bone tissue and alleviate the limitations of autografts and allografts. To meet that aim, it is essential to have a design approach able to generate scaffold architectures that will promote bone formation. Here, we present a topology-varying design optimization method, the "mixed-topology" approach, that generates new designs from a set of starting structures. This approach was used with objective functions focusing on improving the scaffold's local mechanical microenvironments to mechanobiologically promote bone formation within the scaffold and constraints to ensure manufacturability and achieve desired macroscale properties. The results demonstrate that this approach can successfully generate scaffold designs with improved microenvironments, taking into account different combinations of relevant stimuli and constraints.
Collapse
Affiliation(s)
- Timothy O Josephson
- Biomedical Engineering, Boston University, Boston, MA, USA.
- Center for Multiscale and Translational Mechanobiology, Boston University, Boston, MA, USA.
| | - Elise F Morgan
- Biomedical Engineering, Boston University, Boston, MA, USA
- Center for Multiscale and Translational Mechanobiology, Boston University, Boston, MA, USA
- Mechanical Engineering, Boston University, Boston, MA, USA
| |
Collapse
|
9
|
Wang Y, Liu H, Wang H, Xie H, Zhou S. Micropatterned shape-memory polymer substrate containing hydrogen bonds creates a long-term dynamic microenvironment for regulating nerve-cell fate. J Mater Chem B 2024; 12:6690-6702. [PMID: 38895854 DOI: 10.1039/d4tb00593g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Peripheral nerve injuries (PNIs) caused by mechanical contusion are frequently encountered in clinical practice, using nerve guidance conduits (NGCs) is now a promising therapy. An NGC creates a microenvironment for cell growth and differentiation, thus understanding physical and biochemical cues that can affect nerve-cell fate is a prerequisite for rationally designing NGCs. However, most of the previous works were focused on some static cues, the dynamic nature of the nerve microenvironment has not yet been well captured. Herein, we develop a micropatterned shape-memory polymer as a programmable substrate for providing a dynamic cue for nerve-cell growth. The shape-memory properties enable temporal programming of the substrate, and a dynamic microenvironment is created during standard cell culturing at 37 °C. Unlike most of the biomedical shape-memory polymers that recover rapidly at 37 °C, the proposed substrate shows a slow recovery process lasting 3-4 days and creates a long-term dynamic microenvironment. Results demonstrate that the vertically programmed substrates provide the most suitable dynamic microenvironment for PC12 cells as both the differentiation and maturity are promoted. Overall, this work provides a strategy for creating a long-term dynamic microenvironment for regulating nerve-cell fate and will inspire the rational design of NGCs for the treatment of PNIs.
Collapse
Affiliation(s)
- Yilei Wang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.
| | - Hao Liu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.
| | - Huan Wang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education of China, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Hui Xie
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.
| | - Shaobing Zhou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.
| |
Collapse
|
10
|
Yin S, Wu H, Huang Y, Lu C, Cui J, Li Y, Xue B, Wu J, Jiang C, Gu X, Wang W, Cao Y. Structurally and mechanically tuned macroporous hydrogels for scalable mesenchymal stem cell-extracellular matrix spheroid production. Proc Natl Acad Sci U S A 2024; 121:e2404210121. [PMID: 38954541 PMCID: PMC11253011 DOI: 10.1073/pnas.2404210121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/01/2024] [Indexed: 07/04/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are essential in regenerative medicine. However, conventional expansion and harvesting methods often fail to maintain the essential extracellular matrix (ECM) components, which are crucial for their functionality and efficacy in therapeutic applications. Here, we introduce a bone marrow-inspired macroporous hydrogel designed for the large-scale production of MSC-ECM spheroids. Through a soft-templating approach leveraging liquid-liquid phase separation, we engineer macroporous hydrogels with customizable features, including pore size, stiffness, bioactive ligand distribution, and enzyme-responsive degradability. These tailored environments are conducive to optimal MSC proliferation and ease of harvesting. We find that soft hydrogels enhance mechanotransduction in MSCs, establishing a standard for hydrogel-based 3D cell culture. Within these hydrogels, MSCs exist as both cohesive spheroids, preserving their innate vitality, and as migrating entities that actively secrete functional ECM proteins. Additionally, we also introduce a gentle, enzymatic harvesting method that breaks down the hydrogels, allowing MSCs and secreted ECM to naturally form MSC-ECM spheroids. These spheroids display heightened stemness and differentiation capacity, mirroring the benefits of a native ECM milieu. Our research underscores the significance of sophisticated materials design in nurturing distinct MSC subpopulations, facilitating the generation of MSC-ECM spheroids with enhanced therapeutic potential.
Collapse
Affiliation(s)
- Sheng Yin
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing210093, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan250021, China
| | - Haipeng Wu
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing210093, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan250021, China
| | - Yaying Huang
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing210093, China
| | - Chenjing Lu
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing210093, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan250021, China
| | - Jian Cui
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing210093, China
| | - Ying Li
- Institute of Advanced Materials and Flexible Electronics, School of Chemistry and Materials Science, Nanjing University of Information Science and Technology, Nanjing210044, China
| | - Bin Xue
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing210093, China
| | - Junhua Wu
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan250021, China
- Medical School, Nanjing University, Nanjing210093, China
| | - Chunping Jiang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan250021, China
- Medical School, Nanjing University, Nanjing210093, China
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing210008, China
| | - Xiaosong Gu
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan250021, China
| | - Wei Wang
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing210093, China
- Institute for Brain Sciences, Nanjing University, Nanjing210093, China
| | - Yi Cao
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing210093, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan250021, China
- Institute for Brain Sciences, Nanjing University, Nanjing210093, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing210093, China
- Chemistry and Biomedicine Innovation Center, the Ministry of Education Key Laboratory of High Performance Polymer Materials and Technology, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing210023, China
| |
Collapse
|
11
|
Yang X, Sun Z, Hu Y, Mi C. Multi-parameter design of triply periodic minimal surface scaffolds: from geometry optimization to biomechanical simulation. Biomed Mater 2024; 19:055005. [PMID: 38917813 DOI: 10.1088/1748-605x/ad5ba8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/25/2024] [Indexed: 06/27/2024]
Abstract
This study introduces a multi-parameter design methodology to create triply periodic minimal surface (TPMS) scaffolds with predefined geometric characteristics. The level-set constant and unit cell lengths are systematically correlated with targeted porosity and minimum pore sizes. Network and sheet scaffolds featuring diamond, gyroid, and primitive level-set structures are generated. Three radially graded schemes are applied to each of the six scaffold type, accommodating radial variations in porosity and pore sizes. Computer simulations are conducted to assess the biomechanical performance of 18 scaffold models. Results disclose that diamond and gyroid scaffolds exhibit more expansive design ranges than primitive counterparts. While primitive scaffolds display the highest Young's modulus and permeability, their lower yield strength and mesenchymal stem cell (MSC) adhesion render them unsuitable for bone scaffolds. Gyroid scaffolds demonstrate superior mechanical and permeability performances, albeit with slightly lower MSC adhesion than diamond scaffolds. Sheet scaffolds, characterized by more uniform material distribution, exhibit superior mechanical performance in various directions, despite slightly lower permeability. The higher specific surface area of sheet scaffolds contributes to elevated MSC adhesion. The stimulus factor analysis also revealed the superior differentiation potential of sheet scaffolds over network ones. The diamond sheet type demonstrated the optimal differentiation. Introducing radial gradations enhances axial mechanical performance at the expense of radial mechanical performance. Radially decreasing porosity displays the highest permeability, MSC adhesion, and differentiation capability, aligning with the structural characteristics of human bones. This study underscores the crucial need to balance diverse biomechanical properties of TPMS scaffolds for bone tissue engineering.
Collapse
Affiliation(s)
- Xiaoshuai Yang
- Jiangsu Key Laboratory of Mechanical Analysis for Infrastructure and Advanced Equipment, School of Civil Engineering, Southeast University, Nanjing, Jiangsu 210096, People's Republic of China
| | - Zhongwei Sun
- Jiangsu Key Laboratory of Mechanical Analysis for Infrastructure and Advanced Equipment, School of Civil Engineering, Southeast University, Nanjing, Jiangsu 210096, People's Republic of China
| | - Yuanbin Hu
- Department of Orthopaedics, Yangzhou Hospital of TCM, Yangzhou, Jiangsu 225127, People's Republic of China
| | - Changwen Mi
- Jiangsu Key Laboratory of Mechanical Analysis for Infrastructure and Advanced Equipment, School of Civil Engineering, Southeast University, Nanjing, Jiangsu 210096, People's Republic of China
| |
Collapse
|
12
|
Krupczak B, Farruggio C, Van Vliet KJ. Manufacturing mesenchymal stromal cells in a microcarrier-microbioreactor platform can enhance cell yield and quality attributes: case study for acute respiratory distress syndrome. J Transl Med 2024; 22:614. [PMID: 38956643 PMCID: PMC11220991 DOI: 10.1186/s12967-024-05373-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/04/2024] [Indexed: 07/04/2024] Open
Abstract
Mesenchymal stem and stromal cells (MSCs) hold potential to treat a broad range of clinical indications, but clinical translation has been limited to date due in part to challenges with batch-to-batch reproducibility of potential critical quality attributes (pCQAs) that can predict potency/efficacy. Here, we designed and implemented a microcarrier-microbioreactor approach to cell therapy manufacturing, specific to anchorage-dependent cells such as MSCs. We sought to assess whether increased control of the biochemical and biophysical environment had the potential to create product with consistent presentation and elevated expression of pCQAs relative to established manufacturing approaches in tissue culture polystyrene (TCPS) flasks. First, we evaluated total cell yield harvested from dissolvable, gelatin microcarriers within a microbioreactor cassette (Mobius Breez) or a flask control with matched initial cell seeding density and culture duration. Next, we identified 24 genes implicated in a therapeutic role for a specific motivating indication, acute respiratory distress syndrome (ARDS); expression of these genes served as our pCQAs for initial in vitro evaluation of product potency. We evaluated mRNA expression for three distinct donors to assess inter-donor repeatability, as well as for one donor in three distinct batches to assess within-donor, inter-batch variability. Finally, we assessed gene expression at the protein level for a subset of the panel to confirm successful translation. Our results indicated that MSCs expanded with this microcarrier-microbioreactor approach exhibited reasonable donor-to-donor repeatability and reliable batch-to-batch reproducibility of pCQAs. Interestingly, the baseline conditions of this microcarrier-microbioreactor approach also significantly improved expression of several key pCQAs at the gene and protein expression levels and reduced total media consumption relative to TCPS culture. This proof-of-concept study illustrates key benefits of this approach to therapeutic cell process development for MSCs and other anchorage-dependent cells that are candidates for cell therapies.
Collapse
Affiliation(s)
- Brandon Krupczak
- Harvard-MIT Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
- Singapore-MIT Alliance for Research and Technology, Critical Analytics for Manufacturing Personalised-medicine, 1 Create Way, Singapore, 138602, Singapore
| | - Camille Farruggio
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, 02139, USA
- Singapore-MIT Alliance for Research and Technology, Critical Analytics for Manufacturing Personalised-medicine, 1 Create Way, Singapore, 138602, Singapore
| | - Krystyn J Van Vliet
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, 02139, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA.
- Singapore-MIT Alliance for Research and Technology, Critical Analytics for Manufacturing Personalised-medicine, 1 Create Way, Singapore, 138602, Singapore.
- Departments of Materials Science & Engineering and Biomedical Engineering, Cornell University, 144 Feeney Way, Ithaca, NY, 14853, USA.
| |
Collapse
|
13
|
Wang X, Yang Y, Wang Y, Lu C, Hu X, Kawazoe N, Yang Y, Chen G. Focal adhesion and actin orientation regulated by cellular geometry determine stem cell differentiation via mechanotransduction. Acta Biomater 2024; 182:81-92. [PMID: 38734287 DOI: 10.1016/j.actbio.2024.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 04/23/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Tuning cell adhesion geometry can affect cytoskeleton organization and the distribution of cytoskeleton forces, which play critical roles in controlling cell functions. To elucidate the geometrical relationship with cytoskeleton force distribution, it is necessary to control cell morphology. In this study, a series of dextral vortex micropatterns were prepared to precisely control cell morphology for investigating the influence of the curvature degree of adhesion curves on intracellular force distribution and stem cell differentiation at a sub-cellular level. Peripherial actin filaments of micropatterned cells were assembled along the adhesion curves and showed different orientations, filament thicknesses and densities. Focal adhesion and cytoskeleton force distribution were dependent on the curvature degree. Intracellular force distribution was also regulated by adhesion curves. The cytoskeleton and force distribution affected the osteogenic differentiation of mesenchymal stem cells through a YAP/TAZ-mediated mechanotransduction process. Thus, regulation of cell adhesion curvature, especially at cytoskeletal filament level, is critical for cell function manipulation. STATEMENT OF SIGNIFICANCE: In this study, a series of dextral micro-vortexes were prepared and used for the culture of human mesenchymal stem cells (hMSCs) to precisely control adhesive curvatures (0°, 30°, 60°, and 90°). The single MSCs on the micropatterns had the same size and shape but showed distinct focal adhesion (FA) and cytoskeleton orientations. Cellular nanomechanics were observed to be correlated with the curvature degrees, subsequently influencing nuclear morphological features. As a consequence, the localization of the mechanotransduction sensor and activator-YAP/TAZ was affected, influencing osteogenic differentiation. The results revealed the pivotal role of adhesive curvatures in the manipulation of stem cell differentiation via the machanotransduction process, which has rarely been investigated.
Collapse
Affiliation(s)
- Xinlong Wang
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
| | - Yingjun Yang
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
| | - Yongtao Wang
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
| | - Chengyu Lu
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
| | - Xiaohong Hu
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Graduate School of Life and Environmental Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
| | - Naoki Kawazoe
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Yingnan Yang
- Graduate School of Life and Environmental Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
| | - Guoping Chen
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan.
| |
Collapse
|
14
|
Ghezzi B, Meglioli M, Salvaterra Toffoli A, Mergoni G, Rossi F, Manfredi M, Lumetti S, Manfredi E. Polishing methods for composites restoration: the influence on human gingival fibroblasts behaviour. BMC Oral Health 2024; 24:651. [PMID: 38831398 PMCID: PMC11149303 DOI: 10.1186/s12903-024-04418-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/27/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Carious/Non-carious cervical lesions with gingival recessions may require both dental and periodontal reconstructive therapy, where flaps/grafts may be placed in contact with a dental filling material. Human Gingival Fibroblasts (HGF-1) response during the early phase of healing could vary according to the procedures employed to cure the dental composite. Moreover, oxygen diffusion into dental composite inhibits the polymerization reaction, creating an oxygen-inhibited layer (OIL) that presents residual unreacted monomers. The aim of this study was to assess the effect of different polishing techniques and OIL on HGF-1. METHODS Composite discs polished with different techniques (diamond rubber, abrasive discs and tungsten carbide burr) were used. An additional not polished smooth group obtained with and without OIL was used as control. Samples were physically characterized through the analysis of their hydrophilicity and surface topography through contact angle measurement and SEM, respectively; afterwards the biologic response of HGF-1 when cultured on the different substrates was analyzed in terms of cytotoxicity and gene expression. RESULTS The finishing systems caused alterations to the wettability, even if without a proportional relation towards the results of the proliferation essay, from which emerges a greater proliferation on surfaces polished with one-step diamond rubber and with abrasive discs as well as a direct effect of the glycerin layer, confirming that surface roughness can heavily influence the biological response of HGF-1. CONCLUSIONS Surfaces wettability as well as cellular behavior seem to be affected by the selection of the finishing system used to lastly shape the restoration. Especially, the presence of OIL act as a negative factor in the regards of human gingival fibroblasts. The present study may provide the first clinical instruction regarding the best polishing system of composite material when the restoration is placed directly in contact with soft tissue cells. Understanding HGF-1 behavior can help identifying the polishing treatment for direct restoration of carious/non-carious cervical lesions associated with gingival recessions.
Collapse
Affiliation(s)
- Benedetta Ghezzi
- Department of Medicine and Surgery, Centre for Dental Medicine, University of Parma, Via Gramsci 14, Parma, 43126, Italy.
- IMEM-CNR, Institute of Materials for Electronics and Magnetism, National Research Council, Parco Area delle Scienze 37/A, Parma, 43124, Italy.
| | - Matteo Meglioli
- Department of Medicine and Surgery, Centre for Dental Medicine, University of Parma, Via Gramsci 14, Parma, 43126, Italy
| | - Andrea Salvaterra Toffoli
- Department of Medicine and Surgery, Centre for Dental Medicine, University of Parma, Via Gramsci 14, Parma, 43126, Italy
| | - Giovanni Mergoni
- Department of Medicine and Surgery, Centre for Dental Medicine, University of Parma, Via Gramsci 14, Parma, 43126, Italy
| | - Francesca Rossi
- IMEM-CNR, Institute of Materials for Electronics and Magnetism, National Research Council, Parco Area delle Scienze 37/A, Parma, 43124, Italy
| | - Maddalena Manfredi
- Department of Medicine and Surgery, Centre for Dental Medicine, University of Parma, Via Gramsci 14, Parma, 43126, Italy
| | - Simone Lumetti
- Department of Medicine and Surgery, Centre for Dental Medicine, University of Parma, Via Gramsci 14, Parma, 43126, Italy
- IMEM-CNR, Institute of Materials for Electronics and Magnetism, National Research Council, Parco Area delle Scienze 37/A, Parma, 43124, Italy
| | - Edoardo Manfredi
- Department of Medicine and Surgery, Centre for Dental Medicine, University of Parma, Via Gramsci 14, Parma, 43126, Italy
| |
Collapse
|
15
|
Ghagre A, Delarue A, Srivastava LK, Koushki N, Ehrlicher A. Nuclear curvature determines Yes-associated protein localization and differentiation of mesenchymal stem cells. Biophys J 2024; 123:1222-1239. [PMID: 38605521 PMCID: PMC11140468 DOI: 10.1016/j.bpj.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 01/17/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024] Open
Abstract
Controlling mesenchymal stem cell (MSC) differentiation remains a critical challenge in MSCs' therapeutic application. Numerous biophysical and mechanical stimuli influence stem cell fate; however, their relative efficacy and specificity in mechanically directed differentiation remain unclear. Yes-associated protein (YAP) is one key mechanosensitive protein that controls MSC differentiation. Previous studies have related nuclear mechanics with YAP activity, but we still lack an understanding of what nuclear deformation specifically regulates YAP and its relationship with mechanical stimuli. Here, we report that maximum nuclear curvature is the most precise biophysical determinant for YAP mechanotransduction-mediated MSC differentiation and is a relevant parameter for stem cell-based therapies. We employed traction force microscopy and confocal microscopy to characterize the causal relationships between contractility and nuclear deformation in regulating YAP activity in MSCs. We observed that an increase in contractility compresses nuclei anisotropically, whereby the degree of asymmetric compression increased the bending curvature of the nuclear membrane. We then examined membrane curvature and tension using thin micropatterned adhesive substrate lines and an FRET-based tension sensor, revealing the direct role of curvature in YAP activity driven by both active and passive nuclear import. Finally, we employed micropatterned lines to control nuclear curvature and precisely direct MSC differentiation. This work illustrates that nuclear curvature subsumes other biophysical aspects to control YAP-mediated differentiation in MSCs and may provide a deterministic solution to some of the challenges in mesenchymal stem cell therapies.
Collapse
Affiliation(s)
- Ajinkya Ghagre
- Department of Bioengineering, McGill University, Montreal, Canada
| | - Alice Delarue
- Department of Bioengineering, McGill University, Montreal, Canada
| | | | - Newsha Koushki
- Department of Bioengineering, McGill University, Montreal, Canada
| | - Allen Ehrlicher
- Department of Bioengineering, McGill University, Montreal, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Canada; Department of Biomedical Engineering, McGill University, Montreal, Canada; Department of Mechanical Engineering, McGill University, Montreal, Canada; Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montreal, Canada; Centre for Structural Biology, McGill University, Montreal, Canada.
| |
Collapse
|
16
|
Schamberger B, Ehrig S, Dechat T, Spitzer S, Bidan CM, Fratzl P, Dunlop JWC, Roschger A. Twisted-plywood-like tissue formation in vitro. Does curvature do the twist? PNAS NEXUS 2024; 3:pgae121. [PMID: 38590971 PMCID: PMC10999733 DOI: 10.1093/pnasnexus/pgae121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 02/23/2024] [Indexed: 04/10/2024]
Abstract
Little is known about the contribution of 3D surface geometry to the development of multilayered tissues containing fibrous extracellular matrix components, such as those found in bone. In this study, we elucidate the role of curvature in the formation of chiral, twisted-plywood-like structures. Tissues consisting of murine preosteoblast cells (MC3T3-E1) were grown on 3D scaffolds with constant-mean curvature and negative Gaussian curvature for up to 32 days. Using 3D fluorescence microscopy, the influence of surface curvature on actin stress-fiber alignment and chirality was investigated. To gain mechanistic insights, we did experiments with MC3T3-E1 cells deficient in nuclear A-type lamins or treated with drugs targeting cytoskeleton proteins. We find that wild-type cells form a thick tissue with fibers predominantly aligned along directions of negative curvature, but exhibiting a twist in orientation with respect to older tissues. Fiber orientation is conserved below the tissue surface, thus creating a twisted-plywood-like material. We further show that this alignment pattern strongly depends on the structural components of the cells (A-type lamins, actin, and myosin), showing a role of mechanosensing on tissue organization. Our data indicate the importance of substrate curvature in the formation of 3D tissues and provide insights into the emergence of chirality.
Collapse
Affiliation(s)
- Barbara Schamberger
- Department of the Chemistry and Physics of Materials, Paris-Lodron University of Salzburg, 5020 Salzburg, Austria
| | - Sebastian Ehrig
- Laboratory of Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Hannoversche Str. 28, 10115 Berlin, Germany
| | - Thomas Dechat
- Ludwig Boltzmann Institute of Osteology of OEGK and AUVA Trauma Centre Meidling, 1st Medical Department, Hanusch Hospital, 1140 Vienna, Austria
| | - Silvia Spitzer
- Ludwig Boltzmann Institute of Osteology of OEGK and AUVA Trauma Centre Meidling, 1st Medical Department, Hanusch Hospital, 1140 Vienna, Austria
| | - Cécile M Bidan
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, 14476 Potsdam, Germany
| | - Peter Fratzl
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, 14476 Potsdam, Germany
| | - John W C Dunlop
- Department of the Chemistry and Physics of Materials, Paris-Lodron University of Salzburg, 5020 Salzburg, Austria
| | - Andreas Roschger
- Department of the Chemistry and Physics of Materials, Paris-Lodron University of Salzburg, 5020 Salzburg, Austria
| |
Collapse
|
17
|
Xiang Y, Yan J, Bao X, Gleadall A, Sun T. Investigation of cell infiltration and colonization in 3D porous scaffolds via integrated experimental and computational strategies. J Biotechnol 2024; 382:78-87. [PMID: 38307299 DOI: 10.1016/j.jbiotec.2024.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 01/29/2024] [Indexed: 02/04/2024]
Abstract
This study aimed to integrate experimental and computational methods to systematically investigate cell infiltration and colonization within porous scaffolds. Poly(lactic acid) discs (Diameter: 6 mm; Thickness: 500 µm) with open pores (Diameter: 400-1100 µm), corners (Angle: 30-120°) and gaps (Distance: 100-500 µm), and cellulosic scaffolds with irregular pores (Diameter: 50-300 µm) were situated in tissue culture plates and cultured with human dermal fibroblasts (HDFs). Both phase contrast and scanning electron microscopy revealed that HDFs initially proliferated on scaffold surfaces, then infiltrated into the porous structures via cell bridging and stacking strategies, which was affected by the initial cell seeding densities, porous structures and culture times. Based on the density-dependent cell growths in two-dimensional cell cultures, power law models were developed to quantitatively simulate cell growths on scaffold surfaces. Model analysis predicted the effect of cell seeding efficiency on cell infiltrations into the porous scaffolds, which was further validated via series cell seeding experiments. The novelty of this research lies in the incorporation of multiple experimental and computational strategies, which enables the mechanistic insights of cell invasion and colonization in porous scaffolds, also facilitates the development of suitable bioprocesses for cell seeding and tissue manufacturing in Tissue Engineering and Regenerative Medicine.
Collapse
Affiliation(s)
- Yu Xiang
- Department of Materials, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Jiongyi Yan
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Xujin Bao
- Department of Materials, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Andrew Gleadall
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Tao Sun
- Department of Chemical Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK.
| |
Collapse
|
18
|
Ozkendir O, Karaca I, Cullu S, Erdoğan OC, Yaşar HN, Dikici S, Owen R, Aldemir Dikici B. Engineering periodontal tissue interfaces using multiphasic scaffolds and membranes for guided bone and tissue regeneration. BIOMATERIALS ADVANCES 2024; 157:213732. [PMID: 38134730 DOI: 10.1016/j.bioadv.2023.213732] [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: 10/22/2023] [Revised: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023]
Abstract
Periodontal diseases are one of the greatest healthcare burdens worldwide. The periodontal tissue compartment is an anatomical tissue interface formed from the periodontal ligament, gingiva, cementum, and bone. This multifaceted composition makes tissue engineering strategies challenging to develop due to the interface of hard and soft tissues requiring multiphase scaffolds to recreate the native tissue architecture. Multilayer constructs can better mimic tissue interfaces due to the individually tuneable layers. They have different characteristics in each layer, with modulation of mechanical properties, material type, porosity, pore size, morphology, degradation properties, and drug-releasing profile all possible. The greatest challenge of multilayer constructs is to mechanically integrate consecutive layers to avoid delamination, especially when using multiple manufacturing processes. Here, we review the development of multilayer scaffolds that aim to recapitulate native periodontal tissue interfaces in terms of physical, chemical, and biological characteristics. Important properties of multiphasic biodegradable scaffolds are highlighted and summarised, with design requirements, biomaterials, and fabrication methods, as well as post-treatment and drug/growth factor incorporation discussed.
Collapse
Affiliation(s)
- Ozgu Ozkendir
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Ilayda Karaca
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Selin Cullu
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Oğul Can Erdoğan
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Hüsniye Nur Yaşar
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Serkan Dikici
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Robert Owen
- School of Pharmacy, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Betül Aldemir Dikici
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey.
| |
Collapse
|
19
|
Luciano M, Versaevel M, Kalukula Y, Gabriele S. Mechanoresponse of Curved Epithelial Monolayers Lining Bowl-Shaped 3D Microwells. Adv Healthc Mater 2024; 13:e2203377. [PMID: 37820698 DOI: 10.1002/adhm.202203377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Indexed: 10/13/2023]
Abstract
The optimal functioning of many organs relies on the curved architecture of their epithelial tissues. However, the mechanoresponse of epithelia to changes in curvature remains misunderstood. Here, bowl-shaped microwells in hydrogels are designed via photopolymerization to faithfully replicate the shape and dimensions of lobular structures. Leveraging these hydrogel-based microwells, curved epithelial monolayers are engineered, and how in-plane and Gaussian curvatures at the microwell entrance influence epithelial behavior is investigated. Cells and nuclei around the microwell edge display a more pronounced centripetal orientation as the in-plane curvature decreases, and enhanced cell straightness and speed. Moreover, cells reorganize their actin cytoskeleton by forming a supracellular actin cable at the microwell edge, with its size becoming more pronounced as the in-plane curvature decreases. The Gaussian curvature at the microwell entrance enhances the maturation of the supracellular actin cable architecture and leads to a vertical orientation of nuclei toward the bottom of the microwell. Increasing Gaussian curvature results in flattened and elongated nuclear morphologies characterized by highly compacted chromatin states. This approach provides better understanding of the mechanoresponse of curved epithelial monolayers curvatures lining lobular structures. In addition, bowl-shaped microwells offer a powerful platform to study curvature-dependent mechanotransduction pathways in anatomically relevant 3D structures.
Collapse
Affiliation(s)
- Marine Luciano
- Mechanobiology & Biomaterials Group, Research Institute for Biosciences, University of Mons, 20 Place du Parc, Mons, B-7000, Belgium
| | - Marie Versaevel
- Mechanobiology & Biomaterials Group, Research Institute for Biosciences, University of Mons, 20 Place du Parc, Mons, B-7000, Belgium
| | - Yohalie Kalukula
- Mechanobiology & Biomaterials Group, Research Institute for Biosciences, University of Mons, 20 Place du Parc, Mons, B-7000, Belgium
| | - Sylvain Gabriele
- Mechanobiology & Biomaterials Group, Research Institute for Biosciences, University of Mons, 20 Place du Parc, Mons, B-7000, Belgium
| |
Collapse
|
20
|
Mierke CT. Extracellular Matrix Cues Regulate Mechanosensing and Mechanotransduction of Cancer Cells. Cells 2024; 13:96. [PMID: 38201302 PMCID: PMC10777970 DOI: 10.3390/cells13010096] [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: 11/12/2023] [Revised: 12/29/2023] [Accepted: 01/01/2024] [Indexed: 01/12/2024] Open
Abstract
Extracellular biophysical properties have particular implications for a wide spectrum of cellular behaviors and functions, including growth, motility, differentiation, apoptosis, gene expression, cell-matrix and cell-cell adhesion, and signal transduction including mechanotransduction. Cells not only react to unambiguously mechanical cues from the extracellular matrix (ECM), but can occasionally manipulate the mechanical features of the matrix in parallel with biological characteristics, thus interfering with downstream matrix-based cues in both physiological and pathological processes. Bidirectional interactions between cells and (bio)materials in vitro can alter cell phenotype and mechanotransduction, as well as ECM structure, intentionally or unintentionally. Interactions between cell and matrix mechanics in vivo are of particular importance in a variety of diseases, including primarily cancer. Stiffness values between normal and cancerous tissue can range between 500 Pa (soft) and 48 kPa (stiff), respectively. Even the shear flow can increase from 0.1-1 dyn/cm2 (normal tissue) to 1-10 dyn/cm2 (cancerous tissue). There are currently many new areas of activity in tumor research on various biological length scales, which are highlighted in this review. Moreover, the complexity of interactions between ECM and cancer cells is reduced to common features of different tumors and the characteristics are highlighted to identify the main pathways of interaction. This all contributes to the standardization of mechanotransduction models and approaches, which, ultimately, increases the understanding of the complex interaction. Finally, both the in vitro and in vivo effects of this mechanics-biology pairing have key insights and implications for clinical practice in tumor treatment and, consequently, clinical translation.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Biological Physics Division, Peter Debye Institute of Soft Matter Physics, Faculty of Physics and Earth Science, Leipzig University, Linnéstraße 5, 04103 Leipzig, Germany
| |
Collapse
|
21
|
Ozanne H, Moubri L, Abou-Nassif L, Thoumire O, Echalard A, Morin-Grognet S, Atmani H, Ladam G, Labat B. Active Osteoblasts or Quiescent Bone Lining Cells? Preosteoblasts Fate Orchestrated by Curvature and Stiffness of an In Vitro 2.5D Biomimetic Culture System. Adv Healthc Mater 2024; 13:e2302222. [PMID: 37929897 DOI: 10.1002/adhm.202302222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Indexed: 11/07/2023]
Abstract
Biomimetic cell culture systems are required to provide more physiologically relevant microenvironments for bone cells. Here, a simple 2.5D culture platform is proposed, combining adjustable stiffness and surface features that mimic bone topography by using sandpaper grits as master molds with two stiffness formulations of polydimethylsiloxane (PDMS). The subsequent replicas perfectly conform the grits and reproduce the corresponding negative relief with cavities separated by convex edges. Biomimicry is also provided by an extracellular matrix (ECM)-like thin film coating, using the layer-by-layer (LbL) method. The topographical features, alternating concave, and convex structures drive preosteoblasts organization and morphology. Strikingly, curvature orchestrates the commitment of preosteoblasts, with i) maturation to active osteoblasts able to produce a dense collagenous matrix that ultimately mineralizes in the cavities, and ii) edges hosting quiescent cells that synthetize a very thin immature collagen layer with no mineralization. In summary, the present in vitro culture system model offers a cell-instructive 2.5D microenvironment that controls preosteoblasts fate, leading to two coexisting subpopulations: mature osteoblasts and bone lining cells (BLC). This promising culture system opens new avenues to advanced tissue-engineered modeling and can be applied to precellularized bone biomaterials.
Collapse
Affiliation(s)
- Hélène Ozanne
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, PBS UMR 6270, Evreux, F-27000, France
| | - Loïc Moubri
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, PBS UMR 6270, Evreux, F-27000, France
| | - Léa Abou-Nassif
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, PBS UMR 6270, Evreux, F-27000, France
| | - Olivier Thoumire
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, PBS UMR 6270, Evreux, F-27000, France
| | - Aline Echalard
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, PBS UMR 6270, Evreux, F-27000, France
| | | | - Hassan Atmani
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, PBS UMR 6270, Evreux, F-27000, France
| | - Guy Ladam
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, PBS UMR 6270, Evreux, F-27000, France
| | - Béatrice Labat
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, PBS UMR 6270, Evreux, F-27000, France
| |
Collapse
|
22
|
Miao Y, Liu X, Luo J, Yang Q, Chen Y, Wang Y. Double-Network DNA Macroporous Hydrogel Enables Aptamer-Directed Cell Recruitment to Accelerate Bone Healing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303637. [PMID: 37949678 PMCID: PMC10767401 DOI: 10.1002/advs.202303637] [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: 08/09/2023] [Revised: 09/20/2023] [Indexed: 11/12/2023]
Abstract
Recruiting endogenous bone marrow mesenchymal stem cells (BMSCs) in vivo to bone defect sites shows great promise in cell therapies for bone tissue engineering, which tackles the shortcomings of delivering exogenous stem cells, including limited sources, low retention, stemness loss, and immunogenicity. However, it remains challenging to efficiently recruit stem cells while simultaneously directing cell differentiation in the dynamic microenvironment and promoting neo-regenerated tissue ingrowth to achieve augmented bone regeneration. Herein, a synthetic macroporous double-network hydrogel presenting nucleic acid aptamer and nano-inducer enhances BMSCs recruitment, and osteogenic differentiation is demonstrated. An air-in-water template enables the rapid construction of highly interconnective macroporous structures, and the physical self-assembly of DNA strands and chemical cross-linking of gelatin chains synergistically generate a resilient double network. The aptamer Apt19S and black phosphorus nanosheets-specific macroporous hydrogel demonstrate highly efficient endogenous BMSCs recruitment, cell differentiation, and extracellular matrix mineralization. Notably, the enhanced calvarial bone healing with promising matrix mineralization and new bone formation is accompanied by adapting this engineered hydrogel to the bone defects. The findings suggest an appealing material approach overcoming the traditional limitations of cell-delivery therapy that can inspire the future design of next-generation hydrogel for enhanced bone tissue regeneration.
Collapse
Affiliation(s)
- Yali Miao
- School of Materials Science and EngineeringSouth China University of TechnologyGuangzhou510641China
- National Engineering Research Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou510006China
- Department of OrthopedicsGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhou510080China
- Guangdong Cardiovascular InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhou510080China
| | - Xiao Liu
- School of Materials Science and EngineeringSouth China University of TechnologyGuangzhou510641China
- National Engineering Research Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou510006China
| | - Jinshui Luo
- School of Materials Science and EngineeringSouth China University of TechnologyGuangzhou510641China
- National Engineering Research Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou510006China
| | - Qian Yang
- School of Materials Science and EngineeringSouth China University of TechnologyGuangzhou510641China
- National Engineering Research Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou510006China
| | - Yunhua Chen
- School of Materials Science and EngineeringSouth China University of TechnologyGuangzhou510641China
- National Engineering Research Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou510006China
- Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou510006China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of EducationSouth China University of TechnologyGuangzhou510006China
| | - Yingjun Wang
- School of Materials Science and EngineeringSouth China University of TechnologyGuangzhou510641China
- National Engineering Research Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou510006China
- Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou510006China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of EducationSouth China University of TechnologyGuangzhou510006China
| |
Collapse
|
23
|
Hayashi K, Kishida R, Tsuchiya A, Ishikawa K. Effects of Space Dimensionality within Scaffold for Bone Regeneration with Large and Oriented Blood Vessels. MATERIALS (BASEL, SWITZERLAND) 2023; 16:7518. [PMID: 38138660 PMCID: PMC10744811 DOI: 10.3390/ma16247518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023]
Abstract
The internal structure of the scaffolds is a key factor for bone regeneration. In this study, we focused on the space dimensionality within the scaffold that may control cell migration and evaluated the effects on the size and orientation of blood vessels and the amount of bone formation in the scaffold. The carbonate apatite scaffolds with intrascaffold space allowing one-dimensional (1D), two-dimensional (2D), or three-dimensional (3D) cell migration were fabricated by 3D printing. These scaffolds had the same space size, i.e., distances between the struts (~300 µm). The scaffolds were implanted into the medial condyle of rabbit femurs for four weeks. Both the size and orientation degree of the blood vessels formed in the scaffolds allowing 1D cell migration were 2.5- to 4.0-fold greater than those of the blood vessels formed in the scaffolds allowing 2D and 3D cell migration. Furthermore, the amount of bone formed in the scaffolds allowing 1D cell migration was 1.4-fold larger than that formed in the scaffolds allowing 2D and 3D cell migration. These are probably because the 1D space limited the direction of cell migration and prevented the branching of blood vessels, whereas 2D and 3D spaces provided the opportunity for random cell migration and blood vessel branching. Thus, scaffolds with 1D space are advantageous for inducing large and oriented blood vessels, resulting in a larger amount of bone formation.
Collapse
Affiliation(s)
- Koichiro Hayashi
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (R.K.); (A.T.); (K.I.)
| | | | | | | |
Collapse
|
24
|
Wang Z, Servio P, Rey AD. Geometry-structure models for liquid crystal interfaces, drops and membranes: wrinkling, shape selection and dissipative shape evolution. SOFT MATTER 2023. [PMID: 38031449 DOI: 10.1039/d3sm01164j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
We review our recent contributions to anisotropic soft matter models for liquid crystal interfaces, drops and membranes, emphasizing validations with experimental and biological data, and with related theory and simulation literature. The presentation aims to illustrate and characterize the rich output and future opportunities of using a methodology based on the liquid crystal-membrane shape equation applied to static and dynamic pattern formation phenomena. The geometry of static and kinetic shapes is usually described with dimensional curvatures that co-mingle shape and curvedness. In this review, we systematically show how the application of a novel decoupled shape-curvedness framework to practical and ubiquitous soft matter phenomena, such as the shape of drops and tactoids and bending of evolving membranes, leads to deeper quantitative insights than when using traditional dimensional mean and Gaussian curvatures. The review focuses only on (1) statics of wrinkling and shape selection in liquid crystal interfaces and membranes; (2) kinetics and dissipative dynamics of shape evolution in membranes; and (3) computational methods for shape selection and shape evolution; due to various limitations other important topics are excluded. Finally, the outlook follows a similar structure. The main results include: (1) single and multiple wavelength corrugations in liquid crystal interfaces appear naturally in the presence of surface splay and bend orientation distortions with scaling laws governed by ratios of anchoring-to-isotropic tension energy; adding membrane elasticity to liquid crystal anchoring generates multiple scales wrinkling as in tulips; drops of liquid crystals encapsulates in membranes can adopt, according to the ratios of anchoring/tension/bending, families of shapes as multilobal, tactoidal, and serrated as observed in biological cells. (2) Mapping the liquid crystal director to a membrane unit normal. The dissipative shape evolution model with irreversible thermodynamics for flows dominated by bending rates, yields new insights. The model explains the kinetic stability of cylinders, while spheres and saddles are attractors. The model also adds to the evolving understanding of outer hair cells in the inner ear. (3) Computational soft matter geometry includes solving shape equations, trajectories on energy and orientation landscapes, and shape-curvedness evolutions on entropy production landscape with efficient numerical methods and adaptive approaches.
Collapse
Affiliation(s)
- Ziheng Wang
- Department of Chemical Engineering, McGill University, 3610 University Street, Montréal, Québec, H3A 2B2, Canada.
| | - Phillip Servio
- Department of Chemical Engineering, McGill University, 3610 University Street, Montréal, Québec, H3A 2B2, Canada.
| | - Alejandro D Rey
- Department of Chemical Engineering, McGill University, 3610 University Street, Montréal, Québec, H3A 2B2, Canada.
| |
Collapse
|
25
|
Riley L, Cheng P, Segura T. Identification and analysis of 3D pores in packed particulate materials. NATURE COMPUTATIONAL SCIENCE 2023; 3:975-992. [PMID: 38177603 DOI: 10.1038/s43588-023-00551-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 10/09/2023] [Indexed: 01/06/2024]
Abstract
We took the classic 'guess the number of beans in a jar game' and amplified the research question. Rather than estimate the quantity of particles in the jar, we sought to characterize the spaces between them. Here we present an approach for delineating the pockets of empty space (three-dimensional pores) between packed particles, which are hotspots for activity in applications and natural phenomena that deal with particulate materials. We utilize techniques from graph theory to exploit information about particle configuration that allows us to locate important spatial landmarks within the void space. These landmarks are the basis for our pore segmentation, where we consider both interior pores as well as entrance and exit pores into and out of the structure. Our method is robust for particles of varying size, form, stiffness and configuration, which allows us to study and compare three-dimensional pores across a range of packed particle types. We report striking relationships between particles and pores that are described mathematically, and we offer a visual library of pore types. With a meaningful discretization of void space, we demonstrate that packed particles can be understood not by their solid space, but by their empty space.
Collapse
Affiliation(s)
- Lindsay Riley
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Tatiana Segura
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
- Department of Medicine, Neurology, Dermatology, Duke University, Durham, NC, USA.
| |
Collapse
|
26
|
Bril M, Saberi A, Jorba I, van Turnhout MC, Sahlgren CM, Bouten CV, Schenning AP, Kurniawan NA. Shape-Morphing Photoresponsive Hydrogels Reveal Dynamic Topographical Conditioning of Fibroblasts. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303136. [PMID: 37740666 PMCID: PMC10625123 DOI: 10.1002/advs.202303136] [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: 05/15/2023] [Revised: 08/22/2023] [Indexed: 09/25/2023]
Abstract
The extracellular environment defines a physical boundary condition with which cells interact. However, to date, cell response to geometrical environmental cues is largely studied in static settings, which fails to capture the spatiotemporally varying cues cells receive in native tissues. Here, a photoresponsive spiropyran-based hydrogel is presented as a dynamic, cell-compatible, and reconfigurable substrate. Local stimulation with blue light (455 nm) alters hydrogel swelling, resulting in on-demand reversible micrometer-scale changes in surface topography within 15 min, allowing investigation into cell response to controlled geometry actuations. At short term (1 h after actuation), fibroblasts respond to multiple rounds of recurring topographical changes by reorganizing their nucleus and focal adhesions (FA). FAs form primarily at the dynamic regions of the hydrogel; however, this propensity is abolished when the topography is reconfigured from grooves to pits, demonstrating that topographical changes dynamically condition fibroblasts. Further, this dynamic conditioning is found to be associated with long-term (72 h) maintenance of focal adhesions and epigenetic modifications. Overall, this study offers a new approach to dissect the dynamic interplay between cells and their microenvironment and shines a new light on the cell's ability to adapt to topographical changes through FA-based mechanotransduction.
Collapse
Affiliation(s)
- Maaike Bril
- Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
- Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Aref Saberi
- Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
- Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Ignasi Jorba
- Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
- Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Mark C. van Turnhout
- Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Cecilia M. Sahlgren
- Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
- Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
- Faculty of Science and EngineeringÅbo Akademi UniversityTurkuFI‐20520Finland
| | - Carlijn V.C. Bouten
- Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
- Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Albert P.H.J. Schenning
- Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
- Department of Chemical Engineering & ChemistryEindhoven University of TechnologyEindhoven5612 AEThe Netherlands
| | - Nicholas A. Kurniawan
- Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
- Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| |
Collapse
|
27
|
Valdés Gómez A, Sevilla FJ. Fractional and scaled Brownian motion on the sphere: The effects of long-time correlations on navigation strategies. Phys Rev E 2023; 108:054117. [PMID: 38115432 DOI: 10.1103/physreve.108.054117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 10/04/2023] [Indexed: 12/21/2023]
Abstract
We analyze fractional Brownian motion and scaled Brownian motion on the two-dimensional sphere S^{2}. We find that the intrinsic long-time correlations that characterize fractional Brownian motion collude with the specific dynamics (navigation strategies) carried out on the surface giving rise to rich transport properties. We focus our study on two classes of navigation strategies: one induced by a specific set of coordinates chosen for S^{2} (we have chosen the spherical ones in the present analysis), for which we find that contrary to what occurs in the absence of such long-time correlations, nonequilibrium stationary distributions are attained. These results resemble those reported in confined flat spaces in one and two dimensions [Guggenberger et al. New J. Phys. 21, 022002 (2019)1367-263010.1088/1367-2630/ab075f; Vojta et al. Phys. Rev. E 102, 032108 (2020)2470-004510.1103/PhysRevE.102.032108]; however, in the case analyzed here, there are no boundaries that affect the motion on the sphere. In contrast, when the navigation strategy chosen corresponds to a frame of reference moving with the particle (a Frenet-Serret reference system), then the equilibrium distribution on the sphere is recovered in the long-time limit. For both navigation strategies, the relaxation times toward the stationary distribution depend on the particular value of the Hurst parameter. We also show that on S^{2}, scaled Brownian motion, distinguished by a time-dependent diffusion coefficient with a power-scaling, is independent of the navigation strategy finding a good agreement between the analytical calculations obtained from the solution of a time-dependent diffusion equation on S^{2}, and the numerical results obtained from our numerical method to generate ensemble of trajectories.
Collapse
Affiliation(s)
- Adriano Valdés Gómez
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Alcaldía Coyoacán, C.P. 04510 Ciudad Universitaria, Ciudad de México, México
- BBVA AI Factory México
| | - Francisco J Sevilla
- Instituto de Física, Universidad Nacional Autónoma de México, Apdo. Postal 20-364, 01000, Ciudad de México, México
| |
Collapse
|
28
|
Yuan X, Wu T, Lu T, Ye J. Effects of Zinc and Strontium Doping on In Vitro Osteogenesis and Angiogenesis of Calcium Silicate/Calcium Phosphate Cement. ACS Biomater Sci Eng 2023; 9:5761-5771. [PMID: 37676927 DOI: 10.1021/acsbiomaterials.3c00193] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
Based on multiple biological functions (mainly osteogenesis and angiogenesis) of bioactive ions, Zn/Sr-doped calcium silicate/calcium phosphate cements (Zn/Sr-CS/CPCs, including 10Zn-CS/CPC, 20Sr-CS/CPC, and 10Zn/20Sr-CS/CPC) were prepared by the addition of Zn and Sr dual active ions into CS/CPC to further accelerate its bone regeneration in this study. The physicochemical and biological properties of the Zn/Sr-CS/CPCs were systematically investigated. The results showed that the setting time was slightly prolonged, the compressive strength and porosity did not change much, and all groups maintained good injectability after the doping of Zn and Sr. Besides, the doping of Zn and Sr had little effect on the phase and microstructure of hydrated products of CS/CPC. The degradation rate of Zn/Sr-CS/CPCs decreased after doping with Zn and Sr. In mouse bone marrow mesenchymal stem cells (mBMSC) experiments, all Zn/Sr-CS/CPCs stimulated the viability, adhesion, proliferation, and alkaline phosphatase (ALP) activity together with osteogenesis-related genes (ALP, Runx2, Col-I, OCN, and OPN). The further addition of Zn and Sr played better and synergistic roles in in vitro osteogenesis. Thereinto, 10Zn/20Sr-CS/CPC manifested the optimum in vitro osteogenic performance. As for human umbilical vein endothelial cell (HUVEC) experiments, the incorporation of CS doped with Zn and Sr into CPC possessed good vascularization properties of proliferation, NO secretion, tube formation, and the expression of angiogenesis-related genes (VEGF, bFGF, and eNOS). In conclusion, the doping of Zn and Sr into CS/CPC could exhibit excellent osteogenesis and good angiogenesis potentials and 10Zn/20Sr-CS/CPC could be considered as a promising candidate in bone repair.
Collapse
Affiliation(s)
- Xinyuan Yuan
- School of Materials Science and Engineering and Key Laboratory of Biomedical Materials of Ministry of Education, South China University of Technology, Guangzhou 510641, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou 510006, P. R. China
| | - Tingting Wu
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510536, P. R. China
| | - Teliang Lu
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, P. R. China
| | - Jiandong Ye
- School of Materials Science and Engineering and Key Laboratory of Biomedical Materials of Ministry of Education, South China University of Technology, Guangzhou 510641, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou 510006, P. R. China
| |
Collapse
|
29
|
Josephson TO, Morgan EF. Harnessing mechanical cues in the cellular microenvironment for bone regeneration. Front Physiol 2023; 14:1232698. [PMID: 37877097 PMCID: PMC10591087 DOI: 10.3389/fphys.2023.1232698] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/25/2023] [Indexed: 10/26/2023] Open
Abstract
At the macroscale, bones experience a variety of compressive and tensile loads, and these loads cause deformations of the cortical and trabecular microstructure. These deformations produce a variety of stimuli in the cellular microenvironment that can influence the differentiation of marrow stromal cells (MSCs) and the activity of cells of the MSC lineage, including osteoblasts, osteocytes, and chondrocytes. Mechanotransduction, or conversion of mechanical stimuli to biochemical and biological signals, is thus part of a multiscale mechanobiological process that drives bone modeling, remodeling, fracture healing, and implant osseointegration. Despite strong evidence of the influence of a variety of mechanical cues, and multiple paradigms proposed to explain the influence of these cues on tissue growth and differentiation, even a working understanding of how skeletal cells respond to the complex combinations of stimuli in their microenvironments remains elusive. This review covers the current understanding of what types of microenvironmental mechanical cues MSCs respond to and what is known about how they respond in the presence of multiple such cues. We argue that in order to realize the vast potential for harnessing the cellular microenvironment for the enhancement of bone regeneration, additional investigations of how combinations of mechanical cues influence bone regeneration are needed.
Collapse
Affiliation(s)
- Timothy O. Josephson
- Biomedical Engineering, Boston University, Boston, MA, United States
- Center for Multiscale and Translational Mechanobiology, Boston University, Boston, MA, United States
| | - Elise F. Morgan
- Biomedical Engineering, Boston University, Boston, MA, United States
- Center for Multiscale and Translational Mechanobiology, Boston University, Boston, MA, United States
- Mechanical Engineering, Boston University, Boston, MA, United States
| |
Collapse
|
30
|
de Lope-Planelles A, González-Novo R, Madrazo E, Peralta-Carrero G, Cruz Rodríguez MP, Zamora-Carreras H, Torrano V, López-Menéndez H, Roda-Navarro P, Monroy F, Redondo-Muñoz J. Mechanical stress confers nuclear and functional changes in derived leukemia cells from persistent confined migration. Cell Mol Life Sci 2023; 80:316. [PMID: 37801090 PMCID: PMC10558412 DOI: 10.1007/s00018-023-04968-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 08/07/2023] [Accepted: 09/14/2023] [Indexed: 10/07/2023]
Abstract
Nuclear deformability plays a critical role in cell migration. During this process, the remodeling of internal components of the nucleus has a direct impact on DNA damage and cell behavior; however, how persistent migration promotes nuclear changes leading to phenotypical and functional consequences remains poorly understood. Here, we described that the persistent migration through physical barriers was sufficient to promote permanent modifications in migratory-altered cells. We found that derived cells from confined migration showed changes in lamin B1 localization, cell morphology and transcription. Further analysis confirmed that migratory-altered cells showed functional differences in DNA repair, cell response to chemotherapy and cell migration in vivo homing experiments. Experimental modulation of actin polymerization affected the redistribution of lamin B1, and the basal levels of DNA damage in migratory-altered cells. Finally, since major nuclear changes were present in migratory-altered cells, we applied a multidisciplinary biochemical and biophysical approach to identify that confined conditions promoted a different biomechanical response of the nucleus in migratory-altered cells. Our observations suggest that mechanical compression during persistent cell migration has a role in stable nuclear and genomic alterations that might handle the genetic instability and cellular heterogeneity in aging diseases and cancer.
Collapse
Affiliation(s)
- Ana de Lope-Planelles
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - Raquel González-Novo
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - Elena Madrazo
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - Gracia Peralta-Carrero
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - María Pilar Cruz Rodríguez
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - Héctor Zamora-Carreras
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - Verónica Torrano
- Department of Biochemistry and Molecular Biology, University of the Basque Country, Leioa, Spain
| | - Horacio López-Menéndez
- Department of Physical Chemistry, Complutense University, Madrid, Spain
- Translational Biophysics, Hospital Doce de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Pedro Roda-Navarro
- Department of Immunology, School of Medicine, University Complutense de Madrid and 12 de Octubre Health Research Institute (Imas12) Madrid, Madrid, Spain
| | - Francisco Monroy
- Department of Physical Chemistry, Complutense University, Madrid, Spain
- Translational Biophysics, Hospital Doce de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Javier Redondo-Muñoz
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain.
| |
Collapse
|
31
|
Franca CM, Athirasala A, Subbiah R, Tahayeri A, Selvakumar P, Mansoorifar A, Horsophonphong S, Sercia A, Nih L, Bertassoni LE. High-Throughput Bioprinting of Geometrically-Controlled Pre-Vascularized Injectable Microgels for Accelerated Tissue Regeneration. Adv Healthc Mater 2023; 12:e2202840. [PMID: 37219011 PMCID: PMC10526736 DOI: 10.1002/adhm.202202840] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/01/2023] [Indexed: 05/24/2023]
Abstract
Successful integration of cell-laden tissue constructs with host vasculature depends on the presence of functional capillaries to provide oxygen and nutrients to the embedded cells. However, diffusion limitations of cell-laden biomaterials challenge regeneration of large tissue defects that require bulk-delivery of hydrogels and cells. Herein, a strategy to bioprint geometrically controlled, endothelial and stem-cell laden microgels in high-throughput is introduced, allowing these cells to form mature and functional pericyte-supported vascular capillaries in vitro, and then injecting these pre-vascularized constructs minimally invasively in-vivo. It is demonstrated that this approach offers both desired scalability for translational applications as well as unprecedented levels of control over multiple microgel parameters to design spatially-tailored microenvironments for better scaffold functionality and vasculature formation. As a proof-of-concept, the regenerative capacity of the bioprinted pre-vascularized microgels is compared with that of cell-laden monolithic hydrogels of the same cellular and matrix composition in hard-to-heal defects in vivo. The results demonstrate that the bioprinted microgels have faster and higher connective tissue formation, more vessels per area, and widespread presence of functional chimeric (human and murine) vascular capillaries across regenerated sites. The proposed strategy, therefore, addresses a significant issue in regenerative medicine, demonstrating a superior potential to facilitate translational regenerative efforts.
Collapse
Affiliation(s)
- Cristiane M Franca
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Portland, OR, 97201, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Portland, OR, 97201, USA
- Division of Biomaterial and Biosciences, Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health & Science University, 2730 S Moody Ave, Portland, OR, 97201, USA
| | - Avathamsa Athirasala
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Portland, OR, 97201, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Portland, OR, 97201, USA
- Division of Biomaterial and Biosciences, Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health & Science University, 2730 S Moody Ave, Portland, OR, 97201, USA
| | - Ramesh Subbiah
- Division of Biomaterial and Biosciences, Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health & Science University, 2730 S Moody Ave, Portland, OR, 97201, USA
| | - Anthony Tahayeri
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Portland, OR, 97201, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Portland, OR, 97201, USA
- Division of Biomaterial and Biosciences, Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health & Science University, 2730 S Moody Ave, Portland, OR, 97201, USA
| | - Prakash Selvakumar
- Division of Biomaterial and Biosciences, Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health & Science University, 2730 S Moody Ave, Portland, OR, 97201, USA
| | - Amin Mansoorifar
- Division of Biomaterial and Biosciences, Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health & Science University, 2730 S Moody Ave, Portland, OR, 97201, USA
| | - Sivaporn Horsophonphong
- Department of Pediatric Dentistry, School of Dentistry, Mahidol University, Bangkok, 73170, Thailand
| | - Ashley Sercia
- Division of Biomaterial and Biosciences, Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health & Science University, 2730 S Moody Ave, Portland, OR, 97201, USA
| | - Lina Nih
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
- David Geffen School of Medicine at University of California, Los Angeles, CA, 90095, USA
| | - Luiz E Bertassoni
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Portland, OR, 97201, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Portland, OR, 97201, USA
- Division of Biomaterial and Biosciences, Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health & Science University, 2730 S Moody Ave, Portland, OR, 97201, USA
- Division of Oncological Sciences, Knight Cancer Institute, Portland, OR, 97201, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR, 97201, USA
- Center for Regenerative Medicine, School of Medicine, Oregon Health & Science University, Portland, OR, 97201, USA
| |
Collapse
|
32
|
Patkar SS, Garcia Garcia C, Palmese LL, Kiick KL. Sequence-Encoded Differences in Phase Separation Enable Formation of Resilin-like Polypeptide-Based Microstructured Hydrogels. Biomacromolecules 2023; 24:3729-3741. [PMID: 37525441 DOI: 10.1021/acs.biomac.3c00418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Microstructured hydrogels are promising platforms to mimic structural and compositional heterogeneities of the native extracellular matrix (ECM). The current state-of-the-art soft matter patterning techniques for generating ECM mimics can be limited owing to their reliance on specialized equipment and multiple time- and energy-intensive steps. Here, a photocross-linking methodology that traps various morphologies of phase-separated multicomponent formulations of compositionally distinct resilin-like polypeptides (RLPs) is reported. Turbidimetry and quantitative 1H NMR spectroscopy were utilized to investigate the sequence-dependent liquid-liquid phase separation of multicomponent solutions of RLPs. Differences between the intermolecular interactions of two different photocross-linkable RLPs and a phase-separating templating RLP were exploited for producing microstructured hydrogels with tunable control over pore diameters (ranging from 1.5 to 150 μm) and shear storage moduli (ranging from 0.2 to 5 kPa). The culture of human mesenchymal stem cells demonstrated high viability and attachment on microstructured hydrogels, suggesting their potential for developing customizable platforms for regenerative medicine applications.
Collapse
Affiliation(s)
- Sai S Patkar
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Cristobal Garcia Garcia
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Luisa L Palmese
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Kristi L Kiick
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19713, United States
| |
Collapse
|
33
|
Li Y, Zhong Z, Xu C, Wu X, Li J, Tao W, Wang J, Du Y, Zhang S. 3D micropattern force triggers YAP nuclear entry by transport across nuclear pores and modulates stem cells paracrine. Natl Sci Rev 2023; 10:nwad165. [PMID: 37457331 PMCID: PMC10347367 DOI: 10.1093/nsr/nwad165] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 04/27/2023] [Accepted: 05/25/2023] [Indexed: 07/18/2023] Open
Abstract
Biophysical cues of the cellular microenvironment tremendously influence cell behavior by mechanotransduction. However, it is still unclear how cells sense and transduce the mechanical signals from 3D geometry to regulate cell function. Here, the mechanotransduction of human mesenchymal stem cells (MSCs) triggered by 3D micropatterns and its effect on the paracrine of MSCs are systematically investigated. Our findings show that 3D micropattern force could influence the spatial reorganization of the cytoskeleton, leading to different local forces which mediate nucleus alteration such as orientation, morphology, expression of Lamin A/C and chromatin condensation. Specifically, in the triangular prism and cuboid micropatterns, the ordered F-actin fibers are distributed over and fully transmit compressive forces to the nucleus, which results in nuclear flattening and stretching of nuclear pores, thus enhancing the nuclear import of YES-associated protein (YAP). Furthermore, the activation of YAP significantly enhances the paracrine of MSCs and upregulates the secretion of angiogenic growth factors. In contrast, the fewer compressive forces on the nucleus in cylinder and cube micropatterns cause less YAP entering the nucleus. The skin repair experiment provides the first in vivo evidence that enhanced MSCs paracrine by 3D geometry significantly promotes tissue regeneration. The current study contributes to understanding the in-depth mechanisms of mechanical signals affecting cell function and provides inspiration for innovative design of biomaterials.
Collapse
Affiliation(s)
| | | | - Cunjing Xu
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan430074, China
| | - Xiaodan Wu
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan430074, China
| | - Jiaqi Li
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan430074, China
| | - Weiyong Tao
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan430074, China
| | - Jianglin Wang
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan430074, China
| | | | | |
Collapse
|
34
|
Lu Q, Diao J, Wang Y, Feng J, Zeng F, Yang Y, Kuang Y, Zhao N, Wang Y. 3D printed pore morphology mediates bone marrow stem cell behaviors via RhoA/ROCK2 signaling pathway for accelerating bone regeneration. Bioact Mater 2023; 26:413-424. [PMID: 36969106 PMCID: PMC10036893 DOI: 10.1016/j.bioactmat.2023.02.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/10/2023] [Accepted: 02/22/2023] [Indexed: 03/28/2023] Open
Abstract
Bone bionics and structural engineering have sparked a broad interest in optimizing artificial scaffolds for better bone regeneration. However, the mechanism behind scaffold pore morphology-regulated bone regeneration remains unclear, making the structure design of scaffolds for bone repair challenging. To address this issue, we have carefully assessed diverse cell behaviors of bone mesenchymal stem cells (BMSCs) on the β-tricalcium phosphate (β-TCP) scaffolds with three representative pore morphologies (i.e., cross column, diamond, and gyroid pore unit, respectively). Among the scaffolds, BMSCs on the β-TCP scaffold with diamond pore unit (designated as D-scaffold) demonstrated enhanced cytoskeletal forces, elongated nucleus, faster cell mobility, and better osteogenic differentiation potential (for example, the alkaline phosphatase expression level in D-scaffold were 1.5-2 times higher than other groups). RNA-sequencing analysis and signaling pathway intervention revealed that Ras homolog gene family A (RhoA)/Rho-associated kinase-2 (ROCK2) has in-depth participated in the pore morphology-mediated BMSCs behaviors, indicating an important role of mechanical signaling transduction in scaffold-cell interactions. Finally, femoral condyle defect repair results showed that D-scaffold could effectively promote endogenous bone regeneration, of which the osteogenesis rate was 1.2-1.8 times higher than the other groups. Overall, this work provides insights into pore morphology-mediated bone regeneration mechanisms for developing novel bioadaptive scaffold designs.
Collapse
Affiliation(s)
- Qiji Lu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
- NMPA Key Laboratory for Research and Evaluation of Innovative Biomaterials for Medical Devices. Guangzhou, 510006, PR China
| | - Jingjing Diao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
- Medical Devices Research & Testing Center of SCUT, Guangzhou, 510006, PR China
| | - Yingqu Wang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
| | - Jianlang Feng
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
- NMPA Key Laboratory for Research and Evaluation of Innovative Biomaterials for Medical Devices. Guangzhou, 510006, PR China
| | - Fansen Zeng
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
- NMPA Key Laboratory for Research and Evaluation of Innovative Biomaterials for Medical Devices. Guangzhou, 510006, PR China
| | - Yan Yang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
- NMPA Key Laboratory for Research and Evaluation of Innovative Biomaterials for Medical Devices. Guangzhou, 510006, PR China
| | - Yudi Kuang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China
- Guangdong Institute of Advanced Biomaterials and Medical Devices, Guangzhou, 510535, PR China
- Corresponding author. National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China.
| | - Naru Zhao
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
- NMPA Key Laboratory for Research and Evaluation of Innovative Biomaterials for Medical Devices. Guangzhou, 510006, PR China
- Corresponding author. School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China.
| | - Yingjun Wang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
- NMPA Key Laboratory for Research and Evaluation of Innovative Biomaterials for Medical Devices. Guangzhou, 510006, PR China
- Guangdong Institute of Advanced Biomaterials and Medical Devices, Guangzhou, 510535, PR China
- Corresponding author. School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China.
| |
Collapse
|
35
|
Hayashi K, Kishida R, Tsuchiya A, Ishikawa K. Superiority of Triply Periodic Minimal Surface Gyroid Structure to Strut-Based Grid Structure in Both Strength and Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37433180 DOI: 10.1021/acsami.3c06263] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
The aging population has rapidly driven the demand for bone regeneration. The pore structure of a scaffold is a critical factor that affects its mechanical strength and bone regeneration. Triply periodic minimal surface gyroid structures similar to the trabecular bone structure are considered superior to strut-based lattice structures (e.g., grids) in terms of bone regeneration. However, at this stage, this is only a hypothesis and is not supported by evidence. In this study, we experimentally validated this hypothesis by comparing gyroid and grid scaffolds composed of carbonate apatite. The gyroid scaffolds possessed compressive strength approximately 1.6-fold higher than that of the grid scaffolds because the gyroid structure prevented stress concentration, whereas the grid structure could not. The porosity of gyroid scaffolds was higher than that of the grid scaffolds; however, porosity and compressive strength generally have a trade-off relationship. Moreover, the gyroid scaffolds formed more than twice the amount of bone as grid scaffolds in a critical-sized bone defect in rabbit femur condyles. This favorable bone regeneration using gyroid scaffolds was attributed to the high permeability (i.e., larger volume of macropores or porosity) and curvature profile of the gyroid structure. Thus, this study validated the conventional hypothesis using in vivo experiments and revealed factors that led to this hypothetical outcome. The findings of this study are expected to contribute to the development of scaffolds that can achieve early bone regeneration without sacrificing the mechanical strength.
Collapse
Affiliation(s)
- Koichiro Hayashi
- Department of Biomaterials, Faculty of Dental Science, Kyushu University 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Ryo Kishida
- Department of Biomaterials, Faculty of Dental Science, Kyushu University 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Akira Tsuchiya
- Department of Biomaterials, Faculty of Dental Science, Kyushu University 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kunio Ishikawa
- Department of Biomaterials, Faculty of Dental Science, Kyushu University 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
36
|
Shi H, Zhou K, Wang M, Wang N, Song Y, Xiong W, Guo S, Yi Z, Wang Q, Yang S. Integrating physicomechanical and biological strategies for BTE: biomaterials-induced osteogenic differentiation of MSCs. Theranostics 2023; 13:3245-3275. [PMID: 37351163 PMCID: PMC10283054 DOI: 10.7150/thno.84759] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/12/2023] [Indexed: 06/24/2023] Open
Abstract
Large bone defects are a major global health concern. Bone tissue engineering (BTE) is the most promising alternative to avoid the drawbacks of autograft and allograft bone. Nevertheless, how to precisely control stem cell osteogenic differentiation has been a long-standing puzzle. Compared with biochemical cues, physicomechanical stimuli have been widely studied for their biosafety and stability. The mechanical properties of various biomaterials (polymers, bioceramics, metal and alloys) become the main source of physicomechanical stimuli. By altering the stiffness, viscoelasticity, and topography of materials, mechanical stimuli with different strengths transmit into precise signals that mediate osteogenic differentiation. In addition, externally mechanical forces also play a critical role in promoting osteogenesis, such as compression stress, tensile stress, fluid shear stress and vibration, etc. When exposed to mechanical forces, mesenchymal stem cells (MSCs) differentiate into osteogenic lineages by sensing mechanical stimuli through mechanical sensors, including integrin and focal adhesions (FAs), cytoskeleton, primary cilium, ions channels, gap junction, and activating osteogenic-related mechanotransduction pathways, such as yes associated proteins (YAP)/TAZ, MAPK, Rho-GTPases, Wnt/β-catenin, TGFβ superfamily, Notch signaling. This review summarizes various biomaterials that transmit mechanical signals, physicomechanical stimuli that directly regulate MSCs differentiation, and the mechanical transduction mechanisms of MSCs. This review provides a deep and broad understanding of mechanical transduction mechanisms and discusses the challenges that remained in clinical translocation as well as the outlook for the future improvements.
Collapse
Affiliation(s)
- Huixin Shi
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Kaixuan Zhou
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| | - Mingfeng Wang
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| | - Ning Wang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yiping Song
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Wei Xiong
- Department of Plastic Surgery, The First Affiliated Hospital of Medical College of Shihezi University, Shihezi, Xinjiang 832008, China
| | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Zhe Yi
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| | - Qiang Wang
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| | - Shude Yang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| |
Collapse
|
37
|
Loewa A, Feng JJ, Hedtrich S. Human disease models in drug development. NATURE REVIEWS BIOENGINEERING 2023; 1:1-15. [PMID: 37359774 PMCID: PMC10173243 DOI: 10.1038/s44222-023-00063-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/30/2023] [Indexed: 06/20/2023]
Abstract
Biomedical research is undergoing a paradigm shift towards approaches centred on human disease models owing to the notoriously high failure rates of the current drug development process. Major drivers for this transition are the limitations of animal models, which, despite remaining the gold standard in basic and preclinical research, suffer from interspecies differences and poor prediction of human physiological and pathological conditions. To bridge this translational gap, bioengineered human disease models with high clinical mimicry are being developed. In this Review, we discuss preclinical and clinical studies that benefited from these models, focusing on organoids, bioengineered tissue models and organs-on-chips. Furthermore, we provide a high-level design framework to facilitate clinical translation and accelerate drug development using bioengineered human disease models.
Collapse
Affiliation(s)
- Anna Loewa
- Department of Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - James J. Feng
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC Canada
- Department of Mathematics, University of British Columbia, Vancouver, BC Canada
| | - Sarah Hedtrich
- Department of Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- Center of Biological Design, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC Canada
- Max-Delbrück Center for Molecular Medicine (MCD), Helmholtz Association, Berlin, Germany
| |
Collapse
|
38
|
Callens SJP, Fan D, van Hengel IAJ, Minneboo M, Díaz-Payno PJ, Stevens MM, Fratila-Apachitei LE, Zadpoor AA. Emergent collective organization of bone cells in complex curvature fields. Nat Commun 2023; 14:855. [PMID: 36869036 PMCID: PMC9984480 DOI: 10.1038/s41467-023-36436-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/31/2023] [Indexed: 03/05/2023] Open
Abstract
Individual cells and multicellular systems respond to cell-scale curvatures in their environments, guiding migration, orientation, and tissue formation. However, it remains largely unclear how cells collectively explore and pattern complex landscapes with curvature gradients across the Euclidean and non-Euclidean spectra. Here, we show that mathematically designed substrates with controlled curvature variations induce multicellular spatiotemporal organization of preosteoblasts. We quantify curvature-induced patterning and find that cells generally prefer regions with at least one negative principal curvature. However, we also show that the developing tissue can eventually cover unfavorably curved territories, can bridge large portions of the substrates, and is often characterized by collectively aligned stress fibers. We demonstrate that this is partly regulated by cellular contractility and extracellular matrix development, underscoring the mechanical nature of curvature guidance. Our findings offer a geometric perspective on cell-environment interactions that could be harnessed in tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Sebastien J P Callens
- Department of Biomechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft, 2628CD, The Netherlands. .,Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK.
| | - Daniel Fan
- Department of Precision and Microsystems Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Ingmar A J van Hengel
- Department of Biomechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Michelle Minneboo
- Department of Biomechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Pedro J Díaz-Payno
- Department of Biomechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft, 2628CD, The Netherlands.,Department of Orthopedics and Sports Medicine, Erasmus MC University Medical Center, Rotterdam, 3015GD, The Netherlands
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Lidy E Fratila-Apachitei
- Department of Biomechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Amir A Zadpoor
- Department of Biomechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft, 2628CD, The Netherlands
| |
Collapse
|
39
|
Schamberger B, Ziege R, Anselme K, Ben Amar M, Bykowski M, Castro APG, Cipitria A, Coles RA, Dimova R, Eder M, Ehrig S, Escudero LM, Evans ME, Fernandes PR, Fratzl P, Geris L, Gierlinger N, Hannezo E, Iglič A, Kirkensgaard JJK, Kollmannsberger P, Kowalewska Ł, Kurniawan NA, Papantoniou I, Pieuchot L, Pires THV, Renner LD, Sageman-Furnas AO, Schröder-Turk GE, Sengupta A, Sharma VR, Tagua A, Tomba C, Trepat X, Waters SL, Yeo EF, Roschger A, Bidan CM, Dunlop JWC. Curvature in Biological Systems: Its Quantification, Emergence, and Implications across the Scales. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2206110. [PMID: 36461812 DOI: 10.1002/adma.202206110] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/22/2022] [Indexed: 06/17/2023]
Abstract
Surface curvature both emerges from, and influences the behavior of, living objects at length scales ranging from cell membranes to single cells to tissues and organs. The relevance of surface curvature in biology is supported by numerous experimental and theoretical investigations in recent years. In this review, first, a brief introduction to the key ideas of surface curvature in the context of biological systems is given and the challenges that arise when measuring surface curvature are discussed. Giving an overview of the emergence of curvature in biological systems, its significance at different length scales becomes apparent. On the other hand, summarizing current findings also shows that both single cells and entire cell sheets, tissues or organisms respond to curvature by modulating their shape and their migration behavior. Finally, the interplay between the distribution of morphogens or micro-organisms and the emergence of curvature across length scales is addressed with examples demonstrating these key mechanistic principles of morphogenesis. Overall, this review highlights that curved interfaces are not merely a passive by-product of the chemical, biological, and mechanical processes but that curvature acts also as a signal that co-determines these processes.
Collapse
Affiliation(s)
- Barbara Schamberger
- Department of the Chemistry and Physics of Materials, Paris-Lodron University of Salzburg, 5020, Salzburg, Austria
| | - Ricardo Ziege
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, 14476, Potsdam, Germany
| | - Karine Anselme
- IS2M (CNRS - UMR 7361), Université de Haute-Alsace, F-68100, Mulhouse, France
- Université de Strasbourg, F-67081, Strasbourg, France
| | - Martine Ben Amar
- Department of Physics, Laboratoire de Physique de l'Ecole Normale Supérieure, 24 rue Lhomond, 75005, Paris, France
| | - Michał Bykowski
- Department of Plant Anatomy and Cytology, Faculty of Biology, University of Warsaw, 02-096, Warsaw, Poland
| | - André P G Castro
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, 1049-001, Lisboa, Portugal
- ESTS, Instituto Politécnico de Setúbal, 2914-761, Setúbal, Portugal
| | - Amaia Cipitria
- IS2M (CNRS - UMR 7361), Université de Haute-Alsace, F-68100, Mulhouse, France
- Group of Bioengineering in Regeneration and Cancer, Biodonostia Health Research Institute, 20014, San Sebastian, Spain
- IKERBASQUE, Basque Foundation for Science, 48009, Bilbao, Spain
| | - Rhoslyn A Coles
- Cluster of Excellence, Matters of Activity, Humboldt-Universität zu Berlin, 10178, Berlin, Germany
| | - Rumiana Dimova
- Department of Theory and Bio-Systems, Max Planck Institute of Colloids and Interfaces, 14476, Potsdam, Germany
| | - Michaela Eder
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, 14476, Potsdam, Germany
| | - Sebastian Ehrig
- Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
- Berlin Institute for Medical Systems Biology, 10115, Berlin, Germany
| | - Luis M Escudero
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla and Departamento de Biología Celular, Universidad de Sevilla, 41013, Seville, Spain
- Biomedical Network Research Centre on Neurodegenerative Diseases (CIBERNED), 28031, Madrid, Spain
| | - Myfanwy E Evans
- Institute for Mathematics, University of Potsdam, 14476, Potsdam, Germany
| | - Paulo R Fernandes
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, 1049-001, Lisboa, Portugal
| | - Peter Fratzl
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, 14476, Potsdam, Germany
| | - Liesbet Geris
- Biomechanics Research Unit, GIGA In Silico Medicine, University of Liège, 4000, Liège, Belgium
| | - Notburga Gierlinger
- Institute of Biophysics, Department of Nanobiotechnology, University of Natural Resources and Life Sciences Vienna (Boku), 1190, Vienna, Austria
| | - Edouard Hannezo
- Institute of Science and Technology Austria, 3400, Klosterneuburg, Austria
| | - Aleš Iglič
- Laboratory of Physics, Faculty of Electrical engineering, University of Ljubljana, Tržaška 25, SI-1000, Ljubljana, Slovenia
| | - Jacob J K Kirkensgaard
- Condensed Matter Physics, Niels Bohr Institute, University of Copenhagen, Universitetsparken 5, 2100, København Ø, Denmark
- Ingredients and Dairy Technology, Department of Food Science, University of Copenhagen, Rolighedsvej 26, 1958, Frederiksberg, Denmark
| | - Philip Kollmannsberger
- Center for Computational and Theoretical Biology, University of Würzburg, 97074, Würzburg, Germany
| | - Łucja Kowalewska
- Department of Plant Anatomy and Cytology, Faculty of Biology, University of Warsaw, 02-096, Warsaw, Poland
| | - Nicholas A Kurniawan
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB, Eindhoven, The Netherlands
| | - Ioannis Papantoniou
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, PB 813, 3000, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, O&N1, Herestraat 49, PB 813, 3000, Leuven, Belgium
- Institute of Chemical Engineering Sciences, Foundation for Research and Technology (FORTH), Stadiou Str., 26504, Patras, Greece
| | - Laurent Pieuchot
- IS2M (CNRS - UMR 7361), Université de Haute-Alsace, F-68100, Mulhouse, France
- Université de Strasbourg, F-67081, Strasbourg, France
| | - Tiago H V Pires
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, 1049-001, Lisboa, Portugal
| | - Lars D Renner
- Leibniz Institute of Polymer Research and the Max Bergmann Center of Biomaterials, 01069, Dresden, Germany
| | | | - Gerd E Schröder-Turk
- School of Physics, Chemistry and Mathematics, Murdoch University, 90 South St, Murdoch, WA, 6150, Australia
- Department of Materials Physics, Research School of Physics, The Australian National University, Canberra, ACT, 2600, Australia
| | - Anupam Sengupta
- Physics of Living Matter, Department of Physics and Materials Science, University of Luxembourg, L-1511, Luxembourg City, Grand Duchy of Luxembourg
| | - Vikas R Sharma
- Department of the Chemistry and Physics of Materials, Paris-Lodron University of Salzburg, 5020, Salzburg, Austria
| | - Antonio Tagua
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla and Departamento de Biología Celular, Universidad de Sevilla, 41013, Seville, Spain
- Biomedical Network Research Centre on Neurodegenerative Diseases (CIBERNED), 28031, Madrid, Spain
| | - Caterina Tomba
- Univ Lyon, CNRS, INSA Lyon, Ecole Centrale de Lyon, Université Claude Bernard Lyon 1, CPE Lyon, INL, UMR5270, 69622, Villeurbanne, France
| | - Xavier Trepat
- ICREA at the Institute for Bioengineering of Catalonia, The Barcelona Institute for Science and Technology, 08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 08028, Barcelona, Spain
| | - Sarah L Waters
- Mathematical Institute, University of Oxford, OX2 6GG, Oxford, UK
| | - Edwina F Yeo
- Mathematical Institute, University of Oxford, OX2 6GG, Oxford, UK
| | - Andreas Roschger
- Department of the Chemistry and Physics of Materials, Paris-Lodron University of Salzburg, 5020, Salzburg, Austria
| | - Cécile M Bidan
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, 14476, Potsdam, Germany
| | - John W C Dunlop
- Department of the Chemistry and Physics of Materials, Paris-Lodron University of Salzburg, 5020, Salzburg, Austria
| |
Collapse
|
40
|
Xie W, Wei X, Kang H, Jiang H, Chu Z, Lin Y, Hou Y, Wei Q. Static and Dynamic: Evolving Biomaterial Mechanical Properties to Control Cellular Mechanotransduction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204594. [PMID: 36658771 PMCID: PMC10037983 DOI: 10.1002/advs.202204594] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/28/2022] [Indexed: 06/17/2023]
Abstract
The extracellular matrix (ECM) is a highly dynamic system that constantly offers physical, biological, and chemical signals to embraced cells. Increasing evidence suggests that mechanical signals derived from the dynamic cellular microenvironment are essential controllers of cell behaviors. Conventional cell culture biomaterials, with static mechanical properties such as chemistry, topography, and stiffness, have offered a fundamental understanding of various vital biochemical and biophysical processes, such as cell adhesion, spreading, migration, growth, and differentiation. At present, novel biomaterials that can spatiotemporally impart biophysical cues to manipulate cell fate are emerging. The dynamic properties and adaptive traits of new materials endow them with the ability to adapt to cell requirements and enhance cell functions. In this review, an introductory overview of the key players essential to mechanobiology is provided. A biophysical perspective on the state-of-the-art manipulation techniques and novel materials in designing static and dynamic ECM-mimicking biomaterials is taken. In particular, different static and dynamic mechanical cues in regulating cellular mechanosensing and functions are compared. This review to benefit the development of engineering biomechanical systems regulating cell functions is expected.
Collapse
Affiliation(s)
- Wenyan Xie
- Department of BiotherapyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduSichuan610065China
| | - Xi Wei
- Department of Mechanical EngineeringThe University of Hong KongHong KongChina
| | - Heemin Kang
- Department of Materials Science and EngineeringKorea UniversitySeoul02841South Korea
| | - Hong Jiang
- Department of BiotherapyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduSichuan610065China
| | - Zhiqin Chu
- Department of Electrical and Electronic Engineering (Joint Appointment with School of Biomedical Sciences)The University of Hong KongHong KongChina
| | - Yuan Lin
- Department of Mechanical EngineeringThe University of Hong KongHong KongChina
| | - Yong Hou
- Department of Electrical and Electronic EngineeringThe University of Hong KongHong KongChina
- Institut für Chemie und BiochemieFreie Universität BerlinTakustrasse 314195BerlinGermany
| | - Qiang Wei
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials and EngineeringSichuan UniversityChengdu610065China
| |
Collapse
|
41
|
Li K, O'Dwyer R, Yang F, Cymerman J, Li J, Feldman JD, Simon M, Rafailovich M. Enhancement of acellular biomineralization, dental pulp stem cell migration, and differentiation by hybrid fibrin gelatin scaffolds. Dent Mater 2023; 39:305-319. [PMID: 36746694 DOI: 10.1016/j.dental.2023.01.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 01/16/2023] [Accepted: 01/31/2023] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The current in vitro study aims to evaluate cross-linked hydrogels with and without the addition of fibrin that could potentially be used in endodontic regeneration as a scaffold material. METHODS Synthesis of gelatin/fibrin scaffold, and performing nanoscale characterization using cryo-electron microscopy, dynamic rheology, and XRF for structure property relations; plating dental pulp stem cells and determining mineralization, migration, and differentiation using rt-PCR, XRF, and Raman spectroscopy. RESULTS Cryo electron imaging shows gelatin and fibrin, when gelled separately to form classical rectangular cross-linked networks, where the modulus scales inversely with the cube root of the mesh size. When gelled together, a network with a fundamentally different structure is formed, which has higher ductility and when placed as a scaffold in osteogenic media, produces twice the mineral content. Immunofluorescence, RT-PCR and Rahman Spectroscopy indicate that the hybrid gel enhances cell migration, induces odontogenic differentiation of dental pulp stem cells, and promotes formation of dentin. SIGNIFICANCE The mechanical properties of the hybrid gel scaffold enhance in-migration of stem cells and subsequent differentiation, which are critical for regenerative procedures. Under acellular conditions, placement of the hybrid gel enhances biomineralization, which would strengthen the root if used as a scaffold for endodontic regeneration. Our in vitro findings are consistent with previous in vivo studies which show improved mineralization when bleeding is induced into the canal, given that fibrin is a primary component in blood clotting. Therefore, insertion of the hybrid gelatin-fibrin scaffold could enable more reproducible and consistent outcomes if used for regenerative endodontic treatment (RET).
Collapse
Affiliation(s)
- Kao Li
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, NY 11794, USA; Shandong Institute of Petroleum and Chemical Technology, Doying, Shandong 257061, China
| | - Rita O'Dwyer
- Department of Periodontology, Division of Endodontics, School of Dental Medicine, Stony Brook University, NY 11794, USA; Department of Oral Biology and Pathology, Stony Brook University Medical Center, Stony Brook, NY 11794, USA
| | - Fan Yang
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jerome Cymerman
- Department of Periodontology, Division of Endodontics, School of Dental Medicine, Stony Brook University, NY 11794, USA; Department of Oral Biology and Pathology, Stony Brook University Medical Center, Stony Brook, NY 11794, USA
| | - Juyi Li
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jake D Feldman
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marcia Simon
- Department of Oral Biology and Pathology, Stony Brook University Medical Center, Stony Brook, NY 11794, USA
| | - Miriam Rafailovich
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, NY 11794, USA.
| |
Collapse
|
42
|
Li K, Lv C, Feng XQ. Curvature-dependent adhesion of vesicles. Phys Rev E 2023; 107:024405. [PMID: 36932565 DOI: 10.1103/physreve.107.024405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 01/09/2023] [Indexed: 02/10/2023]
Abstract
The morphology and motion behavior of a cell are highly influenced by its external biological, chemical, and physical stimuli, and geometric confinement. In this paper, it is revealed that the mean curvature of the substrate significantly influences the adhesion of vesicles. By employing the variational method and investigating the Helfrich free energy, the configuration of axisymmetric vesicles adhered to curved spherical substrates is obtained theoretically. Moreover, numerical simulations based on the finite element method are also carried out to investigate the adhesion of vesicles on curved substrates with complex shapes. It is found that for a fixed area of a vesicle, its total free energy depends mainly on the mean curvature of the adhesion region but is insensitive to the specific shape of the substrate, and the total free energy monotonically decreases with the increase in the mean curvature. In addition, possible biological significances of the curvature-dependent adhesion, such as the shape of the cell and antibiofouling, are discussed. This study may deepen our understanding of the underlying mechanisms of adhesion in cellular activities.
Collapse
Affiliation(s)
- Kun Li
- Institute of Biomechanics and Medical Engineering, Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Cunjing Lv
- Institute of Biomechanics and Medical Engineering, Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China.,Center for Nano and Micro Mechanics, Tsinghua University, Beijing 100084, China.,State Key Laboratory of Tribology in Advanced Equipment (SKLT), Tsinghua University, Beijing 100084, China
| | - Xi-Qiao Feng
- Institute of Biomechanics and Medical Engineering, Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China.,Center for Nano and Micro Mechanics, Tsinghua University, Beijing 100084, China.,Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing 100084, China
| |
Collapse
|
43
|
The Effect of Tortuosity on Permeability of Porous Scaffold. Biomedicines 2023; 11:biomedicines11020427. [PMID: 36830961 PMCID: PMC9953537 DOI: 10.3390/biomedicines11020427] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/25/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023] Open
Abstract
In designing porous scaffolds, permeability is essential to consider as a function of cell migration and bone tissue regeneration. Good permeability has been achieved by mimicking the complexity of natural cancellous bone. In this study, a porous scaffold was developed according to the morphological indices of cancellous bone (porosity, specific surface area, thickness, and tortuosity). The computational fluid dynamics method analyzes the fluid flow through the scaffold. The permeability values of natural cancellous bone and three types of scaffolds (cubic, octahedron pillar, and Schoen's gyroid) were compared. The results showed that the permeability of the Negative Schwarz Primitive (NSP) scaffold model was similar to that of natural cancellous bone, which was in the range of 2.0 × 10-11 m2 to 4.0 × 10-10 m2. In addition, it was observed that the tortuosity parameter significantly affected the scaffold's permeability and shear stress values. The tortuosity value of the NSP scaffold was in the range of 1.5-2.8. Therefore, tortuosity can be manipulated by changing the curvature of the surface scaffold radius to obtain a superior bone tissue engineering construction supporting cell migration and tissue regeneration. This parameter should be considered when making new scaffolds, such as our NSP. Such efforts will produce a scaffold architecturally and functionally close to the natural cancellous bone, as demonstrated in this study.
Collapse
|
44
|
Huynh QS, Holsinger RMD. Fiber and Electrical Field Alignment Increases BDNF Expression in SH-SY5Y Cells following Electrical Stimulation. Pharmaceuticals (Basel) 2023; 16:138. [PMID: 37259290 PMCID: PMC9960882 DOI: 10.3390/ph16020138] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 09/13/2024] Open
Abstract
The limited expression of neurotrophic factors that can be included in neural tissue engineering scaffolds is insufficient for sustained neural regeneration. A localized and sustained method of introducing neurotrophic factors is required. We describe our attempt at inducing neuroblastoma cells to express trophic factors following electrical stimulation. Human SH-SY5Y neuroblastoma cells, cultured on polycaprolactone electrospun nanofibers, were electrically stimulated using a 100 mV/mm electric field. Nuclear morphology and brain-derived neurotrophic factor (BDNF) expression were analyzed. Cells were classified based on the type of fiber orientation and the alignment of these fibers in relation to the electric field. Nuclear deformation was mainly influenced by fiber orientation rather than the electrical field. Similarly, fiber orientation also induced BDNF expression. Although electrical field alone had no significant effect on BDNF expression, combining fiber orientation with electrical field resulted in BDNF expression in cells that grew on electrospun fibers that were aligned perpendicular to the electrical field.
Collapse
Affiliation(s)
- Quy-Susan Huynh
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - R. M. Damian Holsinger
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
45
|
Flores-Jiménez MS, Garcia-Gonzalez A, Fuentes-Aguilar RQ. Review on Porous Scaffolds Generation Process: A Tissue Engineering Approach. ACS APPLIED BIO MATERIALS 2023; 6:1-23. [PMID: 36599046 DOI: 10.1021/acsabm.2c00740] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Porous scaffolds have been widely explored for tissue regeneration and engineering in vitro three-dimensional models. In this review, a comprehensive literature analysis is conducted to identify the steps involved in their generation. The advantages and disadvantages of the available techniques are discussed, highlighting the importance of considering pore geometrical parameters such as curvature and size, and summarizing the requirements to generate the porous scaffold according to the desired application. This paper considers the available design tools, mathematical models, materials, fabrication techniques, cell seeding methodologies, assessment methods, and the status of pore scaffolds in clinical applications. This review compiles the relevant research in the field in the past years. The trends, challenges, and future research directions are discussed in the search for the generation of a porous scaffold with improved mechanical and biological properties that can be reproducible, viable for long-term studies, and closer to being used in the clinical field.
Collapse
Affiliation(s)
- Mariana S Flores-Jiménez
- Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey Campus Guadalajara, Av. Gral. Ramon Corona No 2514, Colonia Nuevo México, 45121Zapopan, Jalisco, México
| | - Alejandro Garcia-Gonzalez
- Escuela de Medicina, Tecnologico de Monterrey Campus Guadalajara, Av. Gral. Ramon Corona No 2514, Colonia Nuevo México, 45121Zapopan, Jalisco, México
| | - Rita Q Fuentes-Aguilar
- Institute of Advanced Materials and Sustainable Manufacturing, Tecnologico de Monterrey Campus Guadalajara, Av. Gral. Ramon Corona No 2514, Colonia Nuevo México, 45121Zapopan, Jalisco, México
| |
Collapse
|
46
|
Xin L, Wen Y, Song J, Chen T, Zhai Q. Bone regeneration strategies based on organelle homeostasis of mesenchymal stem cells. Front Endocrinol (Lausanne) 2023; 14:1151691. [PMID: 37033227 PMCID: PMC10081449 DOI: 10.3389/fendo.2023.1151691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
The organelle modulation has emerged as a crucial contributor to the organismal homeostasis. The mesenchymal stem cells (MSCs), with their putative functions in maintaining the regeneration ability of adult tissues, have been identified as a major driver to underlie skeletal health. Bone is a structural and endocrine organ, in which the organelle regulation on mesenchymal stem cells (MSCs) function has most been discovered recently. Furthermore, potential treatments to control bone regeneration are developing using organelle-targeted techniques based on manipulating MSCs osteogenesis. In this review, we summarize the most current understanding of organelle regulation on MSCs in bone homeostasis, and to outline mechanistic insights as well as organelle-targeted approaches for accelerated bone regeneration.
Collapse
Affiliation(s)
- Liangjing Xin
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Yao Wen
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jinlin Song
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
- *Correspondence: Qiming Zhai, ; Tao Chen, ; Jinlin Song,
| | - Tao Chen
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
- *Correspondence: Qiming Zhai, ; Tao Chen, ; Jinlin Song,
| | - Qiming Zhai
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
- *Correspondence: Qiming Zhai, ; Tao Chen, ; Jinlin Song,
| |
Collapse
|
47
|
Li Z, Chen Z, Chen X, Zhao R. Mechanical properties of triply periodic minimal surface (TPMS) scaffolds: considering the influence of spatial angle and surface curvature. Biomech Model Mechanobiol 2022; 22:541-560. [PMID: 36550240 DOI: 10.1007/s10237-022-01661-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 11/19/2022] [Indexed: 12/24/2022]
Abstract
Triply periodic minimal surface (TPMS) has a promising application in the design of bone scaffolds due to its relevance in bone structure. Notably, the mechanical properties of TPMS scaffolds can be affected by many factors, including the spatial angle and surface curvature, which, however, remain to be discovered. This paper illustrates our study on the mechanical properties of tissue scaffolds consisting of TPMS structures (Primitive and I-WP) by considering the influence of spatial angle and surface curvature. Also, the development of a novel model representative of the mechanical properties of scaffolds based on the entropy weight fuzzy comprehensive evaluation method is also presented. For experimental investigation and validation, we employed the selective laser melting technology to manufacture scaffolds with varying structures from AlSi10Mg powder and then performed mechanical testing on the scaffolds. Our results show that for a given porosity, the Gaussian curvature of the stretched TPMS structures is more concentrated and have a higher elastic modulus and fatigue life. At the spatial angle θ = 27°, the shear modulus of the primitive unit reaches its largest value; the shear modulus of the I-WP unit is positively correlated with the spatial angle. Additionally, it is found that the comprehensive mechanical properties of TPMS structures can be significantly improved after changing the surface curvature. Taken together, the identified influence of spatial angle and surface curvature and the developed models of scaffold mechanical properties would be of significant advance and guidance for the design and development of bone scaffolds.
Collapse
Affiliation(s)
- Zhitong Li
- School of Mechatronics Engineering, Harbin Institute of Technology, Harbin, 150000, Heilongjiang, China
| | - Zhaobo Chen
- School of Mechatronics Engineering, Harbin Institute of Technology, Harbin, 150000, Heilongjiang, China.
| | - Xiongbiao Chen
- Department of Mechanical Engineering, University of Saskatchewan, Saskatoon, S7N5A9, Canada
| | - Runchao Zhao
- School of Mechatronics Engineering, Harbin Institute of Technology, Harbin, 150000, Heilongjiang, China
| |
Collapse
|
48
|
Cao D, Ding J. Recent advances in regenerative biomaterials. Regen Biomater 2022; 9:rbac098. [PMID: 36518879 PMCID: PMC9745784 DOI: 10.1093/rb/rbac098] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 07/22/2023] Open
Abstract
Nowadays, biomaterials have evolved from the inert supports or functional substitutes to the bioactive materials able to trigger or promote the regenerative potential of tissues. The interdisciplinary progress has broadened the definition of 'biomaterials', and a typical new insight is the concept of tissue induction biomaterials. The term 'regenerative biomaterials' and thus the contents of this article are relevant to yet beyond tissue induction biomaterials. This review summarizes the recent progress of medical materials including metals, ceramics, hydrogels, other polymers and bio-derived materials. As the application aspects are concerned, this article introduces regenerative biomaterials for bone and cartilage regeneration, cardiovascular repair, 3D bioprinting, wound healing and medical cosmetology. Cell-biomaterial interactions are highlighted. Since the global pandemic of coronavirus disease 2019, the review particularly mentions biomaterials for public health emergency. In the last section, perspectives are suggested: (i) creation of new materials is the source of innovation; (ii) modification of existing materials is an effective strategy for performance improvement; (iii) biomaterial degradation and tissue regeneration are required to be harmonious with each other; (iv) host responses can significantly influence the clinical outcomes; (v) the long-term outcomes should be paid more attention to; (vi) the noninvasive approaches for monitoring in vivo dynamic evolution are required to be developed; (vii) public health emergencies call for more research and development of biomaterials; and (viii) clinical translation needs to be pushed forward in a full-chain way. In the future, more new insights are expected to be shed into the brilliant field-regenerative biomaterials.
Collapse
Affiliation(s)
- Dinglingge Cao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| |
Collapse
|
49
|
Vermeulen S, Van Puyvelde B, Bengtsson del Barrio L, Almey R, van der Veer BK, Deforce D, Dhaenens M, de Boer J. Micro-Topographies Induce Epigenetic Reprogramming and Quiescence in Human Mesenchymal Stem Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 10:e2203880. [PMID: 36414384 PMCID: PMC9811462 DOI: 10.1002/advs.202203880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/19/2022] [Indexed: 06/16/2023]
Abstract
Biomaterials can control cell and nuclear morphology. Since the shape of the nucleus influences chromatin architecture, gene expression and cell identity, surface topography can control cell phenotype. This study provides fundamental insights into how surface topography influences nuclear morphology, histone modifications, and expression of histone-associated proteins through advanced histone mass spectrometry and microarray analysis. The authors find that nuclear confinement is associated with a loss of histone acetylation and nucleoli abundance, while pathway analysis reveals a substantial reduction in gene expression associated with chromosome organization. In light of previous observations where the authors found a decrease in proliferation and metabolism induced by micro-topographies, they connect these findings with a quiescent phenotype in mesenchymal stem cells, as further shown by a reduction of ribosomal proteins and the maintenance of multipotency on micro-topographies after long-term culture conditions. Also, this influence of micro-topographies on nuclear morphology and proliferation is reversible, as shown by a return of proliferation when re-cultured on a flat surface. The findings provide novel insights into how biophysical signaling influences the epigenetic landscape and subsequent cellular phenotype.
Collapse
Affiliation(s)
- Steven Vermeulen
- Department of Instructive Biomaterials EngineeringMERLN InstituteUniversity of MaastrichtMaastricht6229 ERThe Netherlands
- Department of Biomedical Engineering and Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Bart Van Puyvelde
- Laboratory of Pharmaceutical BiotechnologyDepartment of PharmaceuticsGhent UniversityGhent9000Belgium
| | - Laura Bengtsson del Barrio
- Department of Instructive Biomaterials EngineeringMERLN InstituteUniversity of MaastrichtMaastricht6229 ERThe Netherlands
| | - Ruben Almey
- Laboratory of Pharmaceutical BiotechnologyDepartment of PharmaceuticsGhent UniversityGhent9000Belgium
| | - Bernard K. van der Veer
- Laboratory for Stem Cell and Developmental EpigeneticsDepartment of Development and RegenerationKU LeuvenLeuven3000Belgium
| | - Dieter Deforce
- Laboratory of Pharmaceutical BiotechnologyDepartment of PharmaceuticsGhent UniversityGhent9000Belgium
| | - Maarten Dhaenens
- Laboratory of Pharmaceutical BiotechnologyDepartment of PharmaceuticsGhent UniversityGhent9000Belgium
| | - Jan de Boer
- Department of Biomedical Engineering and Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| |
Collapse
|
50
|
Zhang Y, Yin P, Huang J, Yang L, Liu Z, Fu D, Hu Z, Huang W, Miao Y. Scalable and high-throughput production of an injectable platelet-rich plasma (PRP)/cell-laden microcarrier/hydrogel composite system for hair follicle tissue engineering. J Nanobiotechnology 2022; 20:465. [PMID: 36329527 PMCID: PMC9632161 DOI: 10.1186/s12951-022-01671-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Tissue engineering of hair follicles (HFs) has enormous potential for hair loss treatment. However, certain challenges remain, including weakening of the dermal papilla cell (DPC) viability, proliferation, and HF inducibility, as well as the associated inefficient and tedious preparation process required to generate extracellular matrix (ECM)-mimicking substrates for biomolecules or cells. Herein, we utilized gelatin methacryloyl (GelMA) and chitosan hydrogels to prepare scalable, monodispersed, and diameter-controllable interpenetrating network GelMA/chitosan-microcarriers (IGMs) loaded with platelet-rich plasma (PRP) and seeded with DPCs, on a high-throughput microfluidic chip. RESULTS The ECM-mimicking hydrogels used for IGMs exhibited surface nano-topography and high porosity. Mass production of IGMs with distinct and precise diameters was achieved by adjusting the oil and aqueous phase flow rate ratio. Moreover, IGMs exhibited appropriate swelling and sustained growth factor release to facilitate a relatively long hair growth phase. DPCs seeded on PRP-loaded IGMs exhibited good viability (> 90%), adhesion, spreading, and proliferative properties (1.2-fold greater than control group). Importantly, PRP-loaded IGMs presented a higher hair inducibility of DPCs in vitro compared to the control and IGMs group (p < 0.05). Furthermore, DPC/PRP-laden IGMs were effectively mixed with epidermal cell (EPC)-laden GelMA to form a PRP-loaded DPC/EPC co-cultured hydrogel system (DECHS), which was subcutaneously injected into the hypodermis of nude mice. The PRP-loaded DECHS generated significantly more HFs (~ 35 per site) and novel vessels (~ 12 per site) than the other groups (p < 0.05 for each). CONCLUSION Taken together, these results illustrate that, based on high-throughput microfluidics, we obtained scalable and controllable production of ECM-mimicking IGMs and DECHS, which simulate an effective micro- and macro-environment to promote DPC bioactivity and hair regeneration, thus representing a potential new strategy for HF tissue engineering.
Collapse
Affiliation(s)
- Yufan Zhang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, 510515, Guangzhou, Guangdong Province, China
| | - Panjing Yin
- Department of Joint Surgery, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Junfei Huang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, 510515, Guangzhou, Guangdong Province, China
| | - Lunan Yang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, 510515, Guangzhou, Guangdong Province, China
| | - Zhen Liu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, 510515, Guangzhou, Guangdong Province, China
| | - Danlan Fu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, 510515, Guangzhou, Guangdong Province, China
| | - Zhiqi Hu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, 510515, Guangzhou, Guangdong Province, China.
| | - Wenhua Huang
- Department of Joint Surgery, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China.
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, 510515, Guangzhou, PR China.
| | - Yong Miao
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, 510515, Guangzhou, Guangdong Province, China.
| |
Collapse
|