1
|
Kim K, Yang J, Li C, Yang CY, Hu P, Liu Y, Huang YY, Sun X, Chi M, Huang C, Sun X, Zhao L, Wang X. Anisotropic structure of nanofiber hydrogel accelerates diabetic wound healing via triadic synergy of immune-angiogenic-neurogenic microenvironments. Bioact Mater 2025; 47:64-82. [PMID: 39877154 PMCID: PMC11772153 DOI: 10.1016/j.bioactmat.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/07/2024] [Accepted: 01/06/2025] [Indexed: 01/31/2025] Open
Abstract
Wound healing in chronic diabetic patients remains challenging due to the multiple types of cellular dysfunction and the impairment of multidimensional microenvironments. The physical signals of structural anisotropy offer significant potential for orchestrating multicellular regulation through physical contact and cellular mechanosensing pathways, irrespective of cell type. In this study, we developed a highly oriented anisotropic nanofiber hydrogel designed to provide directional guidance for cellular extension and cytoskeletal organization, thereby achieving pronounced multicellular modulation, including shape-induced polarization of macrophages, morphogenetic maturation of Schwann cells, oriented extracellular matrix (ECM) deposition by fibroblasts, and enhanced vascularization by endothelial cells. Additionally, we incorporated a VEGF-mimicking peptide to further reinforce angiogenesis, a pivotal phase that interlocks with immune regulation, neurogenesis, and tissue regeneration, ultimately contributing to optimized inter-microenvironmental crosstalk. In vivo studies validated that the anisotropic bioactive nanofiber hydrogel effectively accelerated diabetic wound healing by harnessing the triadic synergy of the immune-angiogenic-neurogenic microenvironments. Our findings highlight the promising potential of combining physical and bioactive signals for the modulation of various cell types and the refinement of the multidimensional microenvironment, offering a novel strategy for diabetic wound healing.
Collapse
Affiliation(s)
- Kunkoo Kim
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, 100084, Beijing, China
| | - Jia Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, 100084, Beijing, China
| | - Chengli Li
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, 102218, Beijing, China
| | - Chun-Yi Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, 100084, Beijing, China
- Center for Biomaterials and Regenerative Medicine, Wuzhen Laboratory, 314500, Tongxiang, China
| | - Peilun Hu
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, 102218, Beijing, China
- Beijing Friendship Hospital, Capital Medical University, 102218, Beijing, China
| | - Yaosai Liu
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, 102218, Beijing, China
| | - Yin-yuan Huang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, 100084, Beijing, China
- Department of Biomedical Engineering, Washington University in St. Louis, 63130, St. Louis, Missouri, United States
| | - Xiaohan Sun
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, 100084, Beijing, China
| | - Ming Chi
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, 100084, Beijing, China
| | - Chenyu Huang
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, 102218, Beijing, China
| | - Xiaodan Sun
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, 100084, Beijing, China
| | - Lingyun Zhao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, 100084, Beijing, China
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, 100084, Beijing, China
| |
Collapse
|
2
|
Ma S, Li Y, Yao S, Shang Y, Li R, Ling L, Fu W, Wei P, Zhao B, Zhang X, Deng J. A deformable SIS/HA composite hydrogel coaxial scaffold promotes alveolar bone regeneration after tooth extraction. Bioact Mater 2025; 46:97-117. [PMID: 39760069 PMCID: PMC11697370 DOI: 10.1016/j.bioactmat.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 01/07/2025] Open
Abstract
After tooth extraction, alveolar bone absorbs unevenly, leading to soft tissue collapse, which hinders full regeneration. Bone loss makes it harder to do dental implants and repairs. Inspired by the biological architecture of bone, a deformable SIS/HA (Small intestinal submucosa/Hydroxyapatite) composite hydrogel coaxial scaffold was designed to maintain bone volume in the socket. The SIS/HA scaffold containing GL13K as the outer layer, mimicking compact bone, while SIS hydrogel loaded with bone marrow mesenchymal stem cells-derived exosomes (BMSCs-Exos) was utilized as the inner core of the scaffolds, which are like soft tissue in the skeleton. This coaxial scaffold exhibited a modulus of elasticity of 0.82 MPa, enabling it to adaptively fill extraction sockets and maintain an osteogenic space. Concurrently, the inner layer of this composite scaffold, enriched with BMSCs-Exos, promoted the proliferation and migration of human umbilical vein endothelial cells (HUVECs) and BMSCs into the scaffold interior (≈3-fold to the control), up-regulated the expression of genes related to osteogenesis (BMP2, ALP, RUNX2, and OPN) and angiogenesis (HIF-1α and VEGF). This induced new blood vessels and bone growth within the scaffold, addressing the issue of low bone formation rates at the center of defects. GL13K was released by approximately 40.87 ± 4.37 % within the first three days, exerting a localized antibacterial effect and further promoting vascularization and new bone formation in peripheral regions. This design aims to achieve an all-around and efficient bone restoration effect in the extraction socket using coaxial scaffolds through a dual internal and external mechanism.
Collapse
Affiliation(s)
- Shiqing Ma
- Department of Stomatology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Yumeng Li
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, China
| | - Shiyu Yao
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, China
| | - Yucheng Shang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, China
| | - Rui Li
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, China
| | - Lijuan Ling
- Chinese People's Liberation Army General Hospital JingZhong MED Huangsi Out-patient department, Beijing, 100120, China
| | - Wei Fu
- Department of Stomatology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Pengfei Wei
- Beijing Biosis Healing Biological Technology Co., Ltd, Beijing, 102600, China
| | - Bo Zhao
- Beijing Biosis Healing Biological Technology Co., Ltd, Beijing, 102600, China
| | - Xuesong Zhang
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing, 100048, China
| | - Jiayin Deng
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, China
| |
Collapse
|
3
|
Ouyang X, Li R, Sun W, Gu Y, Lin J, Fan Z, Yao X, Gu H, Xie C, Li W, Yang Y, Yan Y, Wei W, Wu B, Chen X, He B, Zhang S, Hong Y, Cui Z, Wang X. 3D-Printed Dual-Lineage Inductive Approach for Efficient Osteochondral Regeneration. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40162492 DOI: 10.1021/acsami.4c14063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Osteochondral defect regeneration is challenging due to the mismatch between cartilage and subchondral bone. We developed a functionalized scaffold replicating the natural architecture, biochemical and biomechanical environment of both tissues to promote concurrent regeneration. Our bilayered, zone-specific scaffold combines tailored materials for each tissue type: gelatin methacryloyl (GelMA), modified hyaluronic acid, and umbilical cord-derived extracellular matrix (ECM) for the cartilage layer; GelMA, placenta-derived ECM, and nano amorphous calcium phosphate for the osseous layer. Using 3D digital light-processing printing, we constructed the scaffold with spatially distributed biochemical and biomechanical signaling. This approach created dual chondro-/osteogenic microenvironments facilitating bone marrow mesenchymal stem cell differentiation. In vivo studies demonstrated concurrent regeneration of cartilage and subchondral bone tissues with robust integration. This 3D-printed biomimetic scaffold, featuring dual-lineage inductive properties, shows promising potential for efficient osteochondral regeneration and addresses complex tissue engineering requirements.
Collapse
Affiliation(s)
- Xinyi Ouyang
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX12JD, U.K
| | - Rui Li
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang 314400, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang 310058, China
| | - Wei Sun
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang 314400, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang 310058, China
| | - Yuqing Gu
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang 314400, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang 310058, China
| | - Junxin Lin
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang 314400, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang 310058, China
| | - Zhang Fan
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang 314400, China
| | - Xudong Yao
- International Institutes of Medicine, The fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China
| | - Hongyi Gu
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang 314400, China
| | - Chang Xie
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang 310058, China
| | - Wenyue Li
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang 314400, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang 310058, China
| | - Yifei Yang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang 314400, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang 310058, China
| | - Yiyang Yan
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang 314400, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang 310058, China
| | - Wei Wei
- International Institutes of Medicine, The fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China
| | - Bingbing Wu
- International Institutes of Medicine, The fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China
| | - Xiuying Chen
- International Institutes of Medicine, The fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China
| | - Bin He
- Department of Orthopedic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Shufang Zhang
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang 314400, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang 310058, China
| | - Yi Hong
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang 314400, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang 310058, China
| | - Zhanfeng Cui
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX12JD, U.K
- Oxford Suzhou Centre for Advanced Research (OSCAR), University of Oxford, Suzhou Industrial Park, Jiangsu 215123, China
| | - Xiaozhao Wang
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang 314400, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang 310058, China
| |
Collapse
|
4
|
Im GB, Lee JG, Lim H, Lee JW, Park HS, Kim Y, Asad N, Kim HR, Wie JJ, Bhang SH. Soft Pneumatic Device Designed to Mimic the Periosteal Environment for Regulating the Fate of Mesenchymal Stem Cells. Adv Healthc Mater 2025:e2403229. [PMID: 40123288 DOI: 10.1002/adhm.202403229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 02/16/2025] [Indexed: 03/25/2025]
Abstract
Replicating the complex mechanical forces of muscle movement and fluid flow in in vitro cell culture systems is crucial for understanding cell differentiation and development. However, previous research focused on cell differentiation on static micro/nanotextures without a force field or flat 2-dimensional substrates under a continuous in-plane mechanical force. In this study, cell differentiation is reported using a spatial geometric platform that can periodically modulate complex mechanical forces through a custom-made soft pneumatic device (SPD) to mimic the interfaces between periosteum and interstitial fluid. To elucidate fluidic dynamics and cell fates relevant to bone physiology, the platform exhibited distinct functional responses based on mechanical force levels: low mechanotransduction induced mesenchymal stem/progenitor cells differentiation into osteoprogenitor cells (≈1.5-fold increase in osteo-differentiation), while high mechanotransduction resulted in structural disruptions resembling cell detachment without protein degradation (≈2-fold increase in effective cell detachment). Numerical simulations of SPD elucidated the principal mechanical components for programmable cell differentiation and detachment by deconvoluting the in-plane and out-of-plane mechanical forces of the SPD complex mode. This study offers comprehensive and novel insights into the correlation between mechanical forces and cell differentiation, recovery, and injury in organisms.
Collapse
Affiliation(s)
- Gwang-Bum Im
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jae Gyeong Lee
- Department of Organic and Nano Engineering, Hanyang University, Seoul, 04763, Republic of Korea
- Human-Tech Convergence Program, Hanyang University, Seoul, 04763, Republic of Korea
| | - Hosub Lim
- Division of Engineering in Medicine and Renal Division, Department of Medicine, Harvard Medical School, Brigham Women's Hospital, Boston, MA, 02114, USA
| | - Jae-Won Lee
- School of Electronic and Electrical Engineering, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Hyun Su Park
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yongju Kim
- Department of Polymer Science and Engineering, Inha University, Incheon, 22212, Republic of Korea
| | - Nauman Asad
- School of Electronic and Electrical Engineering, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Hak-Rin Kim
- School of Electronic and Electrical Engineering, Kyungpook National University, Daegu, 41566, Republic of Korea
- School of Electronics Engineering, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Jeong Jae Wie
- Department of Organic and Nano Engineering, Hanyang University, Seoul, 04763, Republic of Korea
- Human-Tech Convergence Program, Hanyang University, Seoul, 04763, Republic of Korea
- The Research Institute of Industrial Science, Hanyang University, Seoul, 04763, Republic of Korea
- Department of Chemical Engineering, Hanyang University, Seoul, 04763, Republic of Korea
- Institute of Nano Science and Technology, Hanyang University, Seoul, 04763, Republic of Korea
- Department of Chemical Engineering, State University of New York College of Environmental Science and Forestry, Syracuse, NY, 13210, USA
- The Michael M. Szwarc Polymer Research Institute, State University of New York College of Environmental Science and Forestry, Syracuse, NY, 13210, USA
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
5
|
Chen Q, Teng J, Zhu C, Du J, Wang G, Wu J. Flexible deformation and special interface structure in nanoparticle-stabilized Pickering bubbles strengthen the immunological response as adjuvant. J Mater Chem B 2025; 13:2725-2736. [PMID: 39851034 DOI: 10.1039/d4tb01763c] [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: 01/25/2025]
Abstract
Adjuvants can enhance an immunological response, which is an important part of vaccine research. Pickering bubbles have been a mega-hit for biomedical applications, including in vivo visualization and targeted drug delivery. However, there have been no studies on Pickering bubbles as an immunological adjuvant, and the special properties and structures of Pickering bubbles may play an important role in immunization. In this study, poly(lactic-co-glycolic acid) (PLGA) particles were used to construct nanoparticle-stabilized Pickering bubbles (PPBs). PPBs were evaluated as immunological adjuvants based on immune response effects and mechanisms and aiming at future applications. PPBs have a flexible gas core and a special surface structure that can increase the cell contact area to promote phagocytosis and enhance the immune response. Quartz crystal microbalance with dissipation (QCM-D) data showed the flexibility of PPBs, and confocal images captured the deformability of PPBs during cell uptake. Flow cytometry and antibody titer detection showed that PPBs significantly promoted antigen uptake and activation of bone-marrow-derived dendritic cells (BMDCs) and induced an immune response with upregulated SIINFEKL MHC I and CD127 molecules on the surface of CD8+ T cells, indicating excellent antigen cross-presentation and cellular immune triggering effects. The upregulation of CD44 and CD62L on CD4+ T cells and the IgG2a/IgG1 ratio bias further demonstrated the excellent adjuvant role of PPBs in immunity. Finally, the biosafety of PPBs as an immunological adjuvant was also demonstrated. Our study demonstrates the potential of particle-stabilized bubbles as immune adjuvants, which provides innovative ideas for vaccine development and design.
Collapse
Affiliation(s)
- Qiuting Chen
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, P. R. China.
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Jie Teng
- Yantai Research Institute, Harbin Engineering University, Yantai 264006, P. R. China.
| | - Cuixiao Zhu
- Yantai Research Institute, Harbin Engineering University, Yantai 264006, P. R. China.
| | - Jinzhi Du
- School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China.
| | - Guixiang Wang
- Yantai Research Institute, Harbin Engineering University, Yantai 264006, P. R. China.
| | - Jie Wu
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, P. R. China.
| |
Collapse
|
6
|
Cui Q, Zheng X, Bai Y, Guo Y, Liu S, Lu Y, Liu L, Song J, Liu Y, Heng BC, You F, Xu M, Deng X, Zhang X. Manipulation of Surface Potential Distribution Enhances Osteogenesis by Promoting Pro-Angiogenic Macrophage Polarization via Activation of the PI3K-Akt Signaling Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414278. [PMID: 39739591 PMCID: PMC11848552 DOI: 10.1002/advs.202414278] [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: 11/04/2024] [Revised: 12/07/2024] [Indexed: 01/02/2025]
Abstract
Regulation of the immune response is key to promoting bone regeneration by electroactive biomaterials. However, how electrical signals at the micro- and nanoscale regulate the immune response and subsequent angiogenesis during bone regeneration remains to be elucidated. Here, the distinctly different surface potential distributions on charged poly(vinylidene fluoridetrifluoroethylene) (P(VDF-TrFE)) matrix surfaces are established by altering the dimensions of ferroelectric nanofillers from 0D BaTiO3 nanoparticles (homogeneous surface potential distribution, HOPD) to 1D BaTiO3 nanofibers (heterogeneous surface potential distribution, HEPD). Compared to HOPD, HEPD is significantly better at inducing the M2 polarization of macrophages and promoting neovascularization, which results in accelerated bone regeneration in vivo. The transcriptomic analysis reveals that macrophages modulated by HEPD display high expression levels of pro-angiogenic genes, which is corroborated by tube-formation assays, RT-qPCR, and western blot analyses in vitro. Mechanistic explorations elucidate activation of the PI3K-Akt signaling pathway, which in turn induces the polarization of macrophages toward a pro-angiogenic phenotype. This study elucidates the cascade of biological processes by which heterogeneous electrical signals at the micro- and nanoscale modulate macrophage functions to promote vascularization and bone regeneration. Hence, this study provides new insights into how the micro- and nanoscale distribution characteristics of electrical signals facilitate bone regeneration.
Collapse
Affiliation(s)
- Qun Cui
- Department of Dental Materials & Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xiaona Zheng
- Department of Dental Materials & Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijing100081P. R. China
- Oral Translational Medicine Research CenterJoint Training base for Shanxi Provincial Key Laboratory in Oral and Maxillofacial RepairReconstruction and RegenerationThe First People's Hospital of JinzhongJinzhongShanxi030600P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical DevicesNMPA Key Laboratory for Dental MaterialsBeijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Yunyang Bai
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Yaru Guo
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Shuo Liu
- Department of Dental Materials & Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Yanhui Lu
- Department of Dental Materials & Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Lulu Liu
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Jia Song
- Department of Dental Materials & Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Yang Liu
- Department of Dental Materials & Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Boon Chin Heng
- Department of Dental Materials & Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Fuping You
- Institute of Systems BiomedicineSchool of Basic Medical SciencesNHC Key Laboratory of Medical ImmunologyBeijing Key Laboratory of Tumor Systems BiologyPeking University Health Science CenterBeijing100191P. R. China
| | - Mingming Xu
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xuliang Deng
- National Engineering Research Center of Oral Biomaterials and Digital Medical DevicesNMPA Key Laboratory for Dental MaterialsBeijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijing100081P. R. China
- Oral Translational Medicine Research CenterJoint Training base for Shanxi Provincial Key Laboratory in Oral and Maxillofacial RepairReconstruction and RegenerationThe First People's Hospital of JinzhongJinzhongShanxi030600P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical DevicesNMPA Key Laboratory for Dental MaterialsBeijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| |
Collapse
|
7
|
Simpson JD, Thomson L, Woodley CM, Wallace CM, Dietrich B, Loch AS, Adams DJ, Berry NG. Predicting the Mechanical Properties of Supramolecular Gels. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2415031. [PMID: 39780688 PMCID: PMC11854865 DOI: 10.1002/adma.202415031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/22/2024] [Indexed: 01/11/2025]
Abstract
The prediction of gelation is an important target, yet current models do not predict any post-gel properties. Gels can be formed through the self-assembly of many molecules, but close analogs often do not form gels. There has been success using a number of computational approaches to understand and predict gelation from molecular structures. However, these approaches focus on whether or not a gel will form, not on the properties of the resulting gels. Critically, it is the properties of the gels that are important for a specific application, not simply whether a gel will be formed. Supramolecular gels are often kinetically trapped, meaning that predicting gel properties is inherently a difficult challenge. Here, the first successful a priori prediction of gel properties for such self-assembled, supramolecular systems is reported.
Collapse
Affiliation(s)
- Jack D. Simpson
- Department of ChemistryUniversity of LiverpoolLiverpoolL69 7ZDUK
| | - Lisa Thomson
- School of ChemistryUniversity of GlasgowGlasgowG12 8QQUK
| | | | | | - Bart Dietrich
- School of ChemistryUniversity of GlasgowGlasgowG12 8QQUK
| | - Alex S. Loch
- School of ChemistryUniversity of GlasgowGlasgowG12 8QQUK
| | - Dave J. Adams
- School of ChemistryUniversity of GlasgowGlasgowG12 8QQUK
| | - Neil G. Berry
- Department of ChemistryUniversity of LiverpoolLiverpoolL69 7ZDUK
| |
Collapse
|
8
|
da Silva RLCG, Bezjak D, Corrales TP, Kappl M, Petri DFS. Chitosan/vanillin/polydimethylsiloxane scaffolds with tunable stiffness for muscle cell proliferation. Int J Biol Macromol 2025; 286:138445. [PMID: 39645124 DOI: 10.1016/j.ijbiomac.2024.138445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 11/18/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
The mechanical properties of scaffolds can significantly influence cell behavior. We propose a methodology for producing chitosan and vanillin-crosslinked chitosan films with tunable mechanical properties to be applied as scaffolds for C2C12 myoblasts. In this approach, aqueous polydimethylsiloxane (PDMS) elastomeric dispersions were prepared using polysorbate 20 as emulsifier. These dispersions were then cured and incorporated into chitosan or vanillin-crosslinked chitosan polymeric dispersions at two different volume fractions (1 % and 10 %), followed by casting into films. Atomic force microscopy in force spectroscopy mode was used to characterize the mechanical properties of the swollen systems in PBS buffer. The mechanical properties of the chitosan and vanillin-crosslinked chitosan scaffolds were modulated by the incorporation of the elastomer. The elastic modulus (E) of chitosan-based scaffolds varied from 60 to 200 kPa, while for vanillin-based scaffolds, it ranged from 200 to 600 kPa with the addition of PDMS elastomers. A general trend observed was that the softest scaffolds exhibited the highest swelling degree and the lowest gel content. After 24 h, good cell viability was observed for chitosan and chitosan-PDMS scaffolds, whereas vanillin-based scaffolds showed borderline cytotoxicity (∼70 %). C2C12 cells demonstrated good adhesion on scaffolds with E values ranging from 114 to 568 kPa.
Collapse
Affiliation(s)
| | - Dragica Bezjak
- Centro de Biotecnología, Universidad Técnica Federico Santa María, Valparaíso, Chile
| | - Tomas P Corrales
- Centro de Biotecnología, Universidad Técnica Federico Santa María, Valparaíso, Chile; Departamento de Física, Universidad Técnica Federico Santa María, Valparaíso, Chile; Millenium Nucleus in NanoBioPhysics (NNBP), Valparaíso, Chile
| | - Michael Kappl
- Max Planck Institute for Polymer Research, Mainz, Germany.
| | - Denise F S Petri
- Fundamental Chemistry Department, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil.
| |
Collapse
|
9
|
Lipari S, Sacco P, Cok M, Scognamiglio F, Romano M, Brun F, Giulianini PG, Marsich E, Aachmann FL, Donati I. Hydrogel Elastic Energy: A Stressor Triggering an Adaptive Stress-Mediated Cell Response. Adv Healthc Mater 2025; 14:e2402400. [PMID: 39535422 PMCID: PMC11730662 DOI: 10.1002/adhm.202402400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Indexed: 11/16/2024]
Abstract
The crosstalk between the cells and the extracellular matrix (ECM) is bidirectional and consists of a pushing/pulling stretch exerted by the cells and a mechanical resistance counteracted by the surrounding microenvironment. It is widely recognized that the stiffness of the ECM, its viscoelasticity, and its overall deformation are the most important traits influencing the response of the cells. Here these three parameters are combined into a concept of elastic energy, which in biological terms represents the mechanical feedback that cells perceive when the ECM is deformed. It is shown that elastic energy is a stress factor that influences the response of cells in three-dimensional (3D) cultures. Strikingly, the higher the elastic energy of the matrix and thus the mechanical feedback, the higher the stress state of the cells, which correlates with the formation of G3BP-mediated stress granules. This condition is associated with an increase in alkaline phosphatase (ALP) activity but a decrease in gene expression and is mediated by the nuclear translocation of Yes-associated protein (YAP). This work supports the importance of considering the elastic energy as mechano-controller in regulating cellular stress state in 3D cultures.
Collapse
Affiliation(s)
- Sara Lipari
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| | - Pasquale Sacco
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| | - Michela Cok
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| | | | - Maurizio Romano
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| | - Francesco Brun
- Department of Engineering and ArchitectureUniversity of TriesteVia A. Valerio 6/1Trieste34127Italy
| | | | - Eleonora Marsich
- Department of MedicineSurgery and Health SciencesUniversity of TriestePiazza dell'Ospitale 1Trieste34129Italy
| | - Finn L. Aachmann
- Department of Biotechnology and Food ScienceNorwegian Biopolymer Laboratory (NOBIPOL)NTNU Norwegian University of Science and TechnologySem Sælands vei 6/8Trondheim7491Norway
| | - Ivan Donati
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| |
Collapse
|
10
|
Chen Y, Hao M, Bousso I, Thomopoulos S, Xia Y. Reliable Fabrication of Mineral-Graded Scaffolds by Spin-Coating and Laser Machining for Use in Tendon-to-Bone Insertion Repair. Adv Healthc Mater 2024; 13:e2402531. [PMID: 39104021 PMCID: PMC11650398 DOI: 10.1002/adhm.202402531] [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: 07/09/2024] [Revised: 07/28/2024] [Indexed: 08/07/2024]
Abstract
A reliable method for fabricating biomimetic scaffolds with a controllable mineral gradient to facilitate the surgical repair of tendon-to-bone injuries and the regeneration of the enthesis is reported. The gradient in mineral content is created by sequentially spin-coating with hydroxyapatite/poly(ε-caprolactone) suspensions containing hydroxyapatite nanoparticles in decreasing concentrations. To produce pores and facilitate cell infiltration, the spin-coated film is released and patterned with an array of funnel-shaped microchannels by laser machining. The unique design provided both mechanical (i.e., substrate stiffness) and biochemical (e.g., hydroxyapatite content) cues to spatially control the graded differentiation of mesenchymal stem cells. Immunocytochemical analysis of human mesenchymal stem cell-seeded scaffolds after 14 days of culture demonstrated the formation of a spatial phenotypic cell gradient from osteoblasts to mineralized chondrocytes based on the level of mineralization in the scaffold. By successfully recreating compositional and cellular features of the native tendon enthesis, the biomimetic scaffolds offer a promising avenue for improved tendon-to-bone repair.
Collapse
Affiliation(s)
- Yidan Chen
- School of Materials Science and EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Min Hao
- The Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
| | - Ismael Bousso
- Department of Orthopedic SurgeryDepartment of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Stavros Thomopoulos
- Department of Orthopedic SurgeryDepartment of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Younan Xia
- The Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
- School of Chemistry and BiochemistryGeorgia Institute of TechnologyAtlantaGA30332USA
| |
Collapse
|
11
|
Shi G, Chang Z, Zhang P, Zou X, Zheng X, Liu X, Yan J, Xu H, Tian Z, Zhang N, Cui N, Sun L, Xu G, Yang H. Heterogeneous stiffness of the bone marrow microenvironment regulates the fate decision of haematopoietic stem and progenitor cells. Cell Prolif 2024; 57:e13715. [PMID: 38982593 PMCID: PMC11628730 DOI: 10.1111/cpr.13715] [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: 03/13/2024] [Revised: 06/04/2024] [Accepted: 06/27/2024] [Indexed: 07/11/2024] Open
Abstract
The bone marrow (BM) niches are the complex microenvironments that surround cells, providing various external stimuli to regulate a range of haematopoietic stem cell (HSC) behaviours. Recently, it has been proposed that the fate decision of HSCs is often correlated with significantly altered biophysical signals of BM niches. To thoroughly elucidate the effect of mechanical microenvironments on cell fates, we constructed 2D and 3D cell culture hydrogels using polyacrylamide to replicate the mechanical properties of heterogeneous sub-niches, including the inherent rigidity of marrow adipose tissue (2 kPa), perivascular tissue (8 kPa) and endosteum region (35 kPa) in BM. Our observations suggest that HSCs can respond to the mechanical heterogeneity of the BM microenvironment, exhibiting diversity in cell mechanics, haematopoietic pool maintenance and differentiated lineages. Hydrogels with higher stiffness promote the preservation of long-term repopulating HSCs (LT-HSCs), while those with lower stiffness support multi-potent progenitors (MPPs) viability in vitro. Furthermore, we established a comprehensive transcriptional profile of haematopoietic subpopulations to reflect the multipotency of haematopoietic stem and progenitor cells (HSPCs) that are modulated by niche-like stiffness. Our findings demonstrate that HSPCs exhibit completely distinct downstream differentiated preferences within hydrogel systems of varying stiffness. This highlights the crucial role of tissue-specific mechanical properties in HSC lineage decisions, which may provide innovative solutions to clinical challenges.
Collapse
Affiliation(s)
- Guolin Shi
- School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
- Engineering Research Center of Chinese Ministry of Education for Biological DiagnosisTreatment and Protection Technology and EquipmentXi'anChina
- Center of Special Environmental Biomechanics & Biomedical EngineeringNorthwestern Polytechnical UniversityXi'anChina
| | - Zhuo Chang
- Laboratory for Multiscale Mechanics and Medical Science, Department of Engineering Mechanics, State Key Laboratory for Strength and Vibration of Mechanical Structures, School of Aerospace EngineeringXi'an Jiaotong UniversityXi'anChina
| | - Pan Zhang
- School College of Food Science and EngineeringShaanxi University of Science and TechnologyXi'anChina
| | - Xiaohang Zou
- School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
- Engineering Research Center of Chinese Ministry of Education for Biological DiagnosisTreatment and Protection Technology and EquipmentXi'anChina
- Center of Special Environmental Biomechanics & Biomedical EngineeringNorthwestern Polytechnical UniversityXi'anChina
| | - Xinmin Zheng
- School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
- Engineering Research Center of Chinese Ministry of Education for Biological DiagnosisTreatment and Protection Technology and EquipmentXi'anChina
- Center of Special Environmental Biomechanics & Biomedical EngineeringNorthwestern Polytechnical UniversityXi'anChina
| | - Xiru Liu
- School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
- Engineering Research Center of Chinese Ministry of Education for Biological DiagnosisTreatment and Protection Technology and EquipmentXi'anChina
- Center of Special Environmental Biomechanics & Biomedical EngineeringNorthwestern Polytechnical UniversityXi'anChina
| | - Jinxiao Yan
- School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
- Engineering Research Center of Chinese Ministry of Education for Biological DiagnosisTreatment and Protection Technology and EquipmentXi'anChina
- Center of Special Environmental Biomechanics & Biomedical EngineeringNorthwestern Polytechnical UniversityXi'anChina
| | - Huiyun Xu
- School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
- Engineering Research Center of Chinese Ministry of Education for Biological DiagnosisTreatment and Protection Technology and EquipmentXi'anChina
- Center of Special Environmental Biomechanics & Biomedical EngineeringNorthwestern Polytechnical UniversityXi'anChina
| | - Zhenhao Tian
- School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
- Engineering Research Center of Chinese Ministry of Education for Biological DiagnosisTreatment and Protection Technology and EquipmentXi'anChina
- Center of Special Environmental Biomechanics & Biomedical EngineeringNorthwestern Polytechnical UniversityXi'anChina
| | - Nu Zhang
- School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
- Engineering Research Center of Chinese Ministry of Education for Biological DiagnosisTreatment and Protection Technology and EquipmentXi'anChina
- Center of Special Environmental Biomechanics & Biomedical EngineeringNorthwestern Polytechnical UniversityXi'anChina
| | - Ning Cui
- School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
- Engineering Research Center of Chinese Ministry of Education for Biological DiagnosisTreatment and Protection Technology and EquipmentXi'anChina
- Center of Special Environmental Biomechanics & Biomedical EngineeringNorthwestern Polytechnical UniversityXi'anChina
| | - Leming Sun
- School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
- Engineering Research Center of Chinese Ministry of Education for Biological DiagnosisTreatment and Protection Technology and EquipmentXi'anChina
- Center of Special Environmental Biomechanics & Biomedical EngineeringNorthwestern Polytechnical UniversityXi'anChina
| | - Guangkui Xu
- Laboratory for Multiscale Mechanics and Medical Science, Department of Engineering Mechanics, State Key Laboratory for Strength and Vibration of Mechanical Structures, School of Aerospace EngineeringXi'an Jiaotong UniversityXi'anChina
| | - Hui Yang
- School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
- Engineering Research Center of Chinese Ministry of Education for Biological DiagnosisTreatment and Protection Technology and EquipmentXi'anChina
- Center of Special Environmental Biomechanics & Biomedical EngineeringNorthwestern Polytechnical UniversityXi'anChina
| |
Collapse
|
12
|
Chauhan M, Roopmani P, Rajendran J, Narayan KP, Giri J. Injectable, in-situ forming, tunable, biocompatible gelatin hydrogels for biomedical applications. Int J Biol Macromol 2024; 285:138200. [PMID: 39617237 DOI: 10.1016/j.ijbiomac.2024.138200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/19/2024] [Accepted: 11/28/2024] [Indexed: 12/08/2024]
Abstract
Gelatin hydrogels have drawn attention for their diverse biomedical applications due to their flexible physiochemical properties. However, such gelatin hydrogels are made of toxic crosslinkers and photoinitiators, restricting their non-invasive deep tissue application. The in-situ forming chemical crosslinked without such toxic crosslinker and UV light has not been explored under physiological conditions. This study establishes a simple method to fabricate an injectable click-chemistry-based in-situ forming gelatin hydrogel in a physiological environment (without toxic UV or photoinitiator) with tunable physiochemical properties to modulate cellular response. Using Divinyl Sulfone (DVS) modification, gelatin hydrogel (GelVS) is optimized with tunable degradation properties, moduli (100 Pa -1000 Pa), gelation time, swelling, degradation, and viscoelastic behaviour. The in-vitro results using fibroblast and stem cells show that the hydrogel and its precursors were cytocompatible with diverging feedback of cells as the modulus varies. The in-vivo analysis for injectability, degradation, and biocompatibility of the GelVS hydrogel displays their biocompatible nature and lasts up to 30 days at the injecting site. Overall results indicate that DVS-modified GelVS hydrogel will be a great system with tunable physicochemical properties to modulate favorable cellular response for tissue regeneration and non-invasive deep tissue application.
Collapse
Affiliation(s)
- Meenakshi Chauhan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Purandhi Roopmani
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Jayakumar Rajendran
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Kumar Pranav Narayan
- Department of Biological Sciences, Birla Institute of Technology & Science, Pilani, Hyderabad Campus, Telangana, India
| | - Jyotsnendu Giri
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India.
| |
Collapse
|
13
|
Wang TC, Abolghasemzade S, McKee BP, Singh I, Pendyala K, Mohajeri M, Patel H, Shaji A, Kersey AL, Harsh K, Kaur S, Dollahon CR, Chukkapalli S, Lele PP, Conway DE, Gaharwar AK, Dickinson RB, Lele TP. Matrix stiffness drives drop like nuclear deformation and lamin A/C tension-dependent YAP nuclear localization. Nat Commun 2024; 15:10151. [PMID: 39578439 PMCID: PMC11584751 DOI: 10.1038/s41467-024-54577-4] [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: 03/21/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024] Open
Abstract
Extracellular matrix (ECM) stiffness influences cancer cell fate by altering gene expression. Previous studies suggest that stiffness-induced nuclear deformation may regulate gene expression through YAP nuclear localization. We investigated the role of the nuclear lamina in this process. We show that the nuclear lamina exhibits mechanical threshold behavior: once unwrinkled, the nuclear lamina is inextensible. A computational model predicts that the unwrinkled lamina is under tension, which is confirmed using a lamin tension sensor. Laminar unwrinkling is caused by nuclear flattening during cell spreading on stiff ECM. Knockdown of lamin A/C eliminates nuclear surface tension and decreases nuclear YAP localization. These findings show that nuclear deformation in cells conforms to the nuclear drop model and reveal a role for lamin A/C tension in controlling YAP localization in cancer cells.
Collapse
Affiliation(s)
- Ting-Ching Wang
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Samere Abolghasemzade
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Brendan P McKee
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Ishita Singh
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Kavya Pendyala
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Mohammad Mohajeri
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Hailee Patel
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Aakansha Shaji
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Anna L Kersey
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Kajol Harsh
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Simran Kaur
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Christina R Dollahon
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Sasanka Chukkapalli
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Pushkar P Lele
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Daniel E Conway
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | - Akhilesh K Gaharwar
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, TX, USA
| | - Richard B Dickinson
- Department of Chemical Engineering, University of Florida, Gainesville, FL, USA
| | - Tanmay P Lele
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA.
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA.
- Department of Translational Medical Sciences, Texas A&M University, Houston, TX, USA.
| |
Collapse
|
14
|
Seth P, Friedrichs J, Limasale YDP, Fertala N, Freudenberg U, Zhang Y, Lampel A, Werner C. Interpenetrating Polymer Network Hydrogels with Tunable Viscoelasticity and Proteolytic Cleavability to Direct Stem Cells In Vitro. Adv Healthc Mater 2024:e2402656. [PMID: 39506429 DOI: 10.1002/adhm.202402656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/18/2024] [Indexed: 11/08/2024]
Abstract
The dynamic nature of cellular microenvironments, regulated by the viscoelasticity and enzymatic cleavage of the extracellular matrix, remains challenging to emulate in engineered synthetic biomaterials. To address this, a novel platform of cell-instructive hydrogels is introduced, composed of two concurrently forming interpenetrating polymer networks (IPNs). These IPNs consist of the same basic building blocks - four-armed poly(ethylene glycol) and the sulfated glycosaminoglycan (sGAG) heparin - are cross-linked through either chemical or physical interactions, allowing for precise and selective tuning of the hydrogel's stiffness, viscoelasticity, and proteolytic cleavability. The studies of the individual and combined effects of these parameters on stem cell behavior revealed that human mesenchymal stem cells exhibited increased spreading and Yes-associated protein transcriptional activity in more viscoelastic and cleavable sGAG-IPN hydrogels. Furthermore, human induced pluripotent stem cell (iPSC) cysts displayed enhanced lumen formation, growth, and pluripotency maintenance when cultured in sGAG-IPN hydrogels with higher viscoelasticity. Inhibition studies emphasized the pivotal roles of actin dynamics and matrix metalloproteinase activity in iPSC cyst morphology, which varied with the viscoelastic properties of the hydrogels. Thus, the introduced sGAG-IPN hydrogel platform offers a powerful methodology for exogenous stem cell fate control.
Collapse
Affiliation(s)
- Prannoy Seth
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials Dresden, 01069, Dresden, Germany
| | - Jens Friedrichs
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials Dresden, 01069, Dresden, Germany
| | - Yanuar Dwi Putra Limasale
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials Dresden, 01069, Dresden, Germany
| | - Nicole Fertala
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials Dresden, 01069, Dresden, Germany
| | - Uwe Freudenberg
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials Dresden, 01069, Dresden, Germany
| | - Yixin Zhang
- Cluster of Excellence Physics of Life, and B CUBE - Center for Molecular Bioengineering, Dresden University of Technology, 01307, Dresden, Germany
| | - Ayala Lampel
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Center for Nanoscience and Nanotechnology Sagol Center for Regenerative Biotechnology, and Center for the Physics and Chemistry of Living Systems Tel Aviv University, Tel Aviv, 69978, Israel
| | - Carsten Werner
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials Dresden, 01069, Dresden, Germany
- Center for Regenerative Therapies Dresden, and Cluster of Excellence Physics of Life, Dresden University of Technology, 01062, Dresden, Germany
| |
Collapse
|
15
|
Kang B, Shin J, Kang D, Chang S, Rhyou C, Cho SW, Lee H. Spatial regulation of hydrogel polymerization reaction using ultrasound-driven streaming vortex. ULTRASONICS SONOCHEMISTRY 2024; 110:107053. [PMID: 39270467 DOI: 10.1016/j.ultsonch.2024.107053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/15/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024]
Abstract
Ultrasound is gaining attention as an alternative tool to regulate chemical processes due to its advantages such as high cost-effectiveness, rapid response, and contact-free operation. Previous studies have demonstrated that acoustic bubble cavitation can generate energy to synthesize functional materials. In this study, we introduce a method to control the spatial distribution of physical and chemical properties of hydrogels by using an ultrasound-mediated particle manipulation technique. We developed a surface acoustic wave device that can localize micro-hydrogel particles, which are formed during gelation, in a hydrogel solution. The hydrogel fabricated with the application of surface acoustic waves exhibited gradients in mechanical, mass transport, and structural properties. We demonstrated that the gel having the property gradients could be utilized as a cell-culture substrate dictating cellular shapes, which is beneficial for interfacial tissue engineering. The acoustic method and fabricated hydrogels with property gradients can be applied to design flexible polymeric materials for soft robotics, biomedical sensors, or bioelectronics applications.
Collapse
Affiliation(s)
- Byungjun Kang
- School of Mechanical Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Jisoo Shin
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Donyoung Kang
- School of Mechanical Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Sooho Chang
- School of Mechanical Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Chanryeol Rhyou
- School of Mechanical Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea; Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Hyungsuk Lee
- School of Mechanical Engineering, Yonsei University, Seoul 03722, Republic of Korea.
| |
Collapse
|
16
|
Dong Y, Wang C, Ding X, Ma X, Huang R, Li M, Yang Q. The characterization of cell traction force on nonflat surfaces with different curvature by elastic hydrogel microspheres. Biotechnol Bioeng 2024; 121:3537-3550. [PMID: 38978386 DOI: 10.1002/bit.28802] [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: 03/20/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024]
Abstract
It is of great importance to study the detachment/attachment behaviors of cells (cancer cell, immune cell, and epithelial cell), as they are closely related with tumor metastasis, immunoreaction, and tissue development at variety scales. To characterize the detachment/attachment during the interaction between cells and substrate, some researchers proposed using cell traction force (CTF) as the indicator. To date, various strategies have been developed to measure the CTF. However, these methods only realize the measurements of cell passive forces on flat cases. To quantify the active CTF on nonflat surfaces, which can better mimic the in vivo case, we employed elastic hydrogel microspheres as a force sensor. The microspheres were fabricated by microfluidic chips with controllable size and mechanical properties to mimic substrate. Cells were cultured on microsphere and the CTF led to the deformation of microsphere. By detecting the morphology information, the CTF exerted by attached cells can be calculated by the in-house numerical code. Using these microspheres, the CTF of various cells (including tumor cell, immunological cell, and epithelium cell) were successfully obtained on nonflat surfaces with different curvature radii. The proposed method provides a versatile platform to measure the CTF with high precision and to understand the detachment/attachment behaviors during physiology processes.
Collapse
Affiliation(s)
- Yuqing Dong
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| | - Cong Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| | - Xin Ding
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| | - Xingquan Ma
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
- School of Civil Engineering and Architecture, Xi'an University of Technology, Xi'an, P.R. China
| | - Rong Huang
- Department of Burn and Plastic Surgery, Second Affiliated Hospital, Air Force Medical University, Xi'an, China
| | - Moxiao Li
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
- Nanjing Center for Multifunctional Lightweight Materials and Structures (MLMS), Nanjing University of Aeronautics and Astronautics, Nanjing, P.R. China
| | - Qingzhen Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| |
Collapse
|
17
|
Su L, Jing L, Zeng S, Fu C, Huang D. Sorghum Prolamin Scaffolds-Based Hybrid Cultured Meat with Enriched Sensory Properties. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23355-23365. [PMID: 39380438 DOI: 10.1021/acs.jafc.4c06474] [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: 10/10/2024]
Abstract
Cultured meat (CM) has been hailed as a sustainable future meat production technology that requires scaffolds to support cell growth. Plant proteins are the most promising raw materials for edible scaffolds but remain underutilized. In this study, kafirin, an abundant, readily available, and nonallergenic prolamin extracted from red sorghum, was explored to fabricate 3D porous sponge-like scaffolds via a simple template-leaching method. The scaffolds featured fully interconnected pores with a high porosity of approximately 84% and mechanical properties of 1.0-1.9 kPa. Porcine skeletal muscle cells (PSCs) and adipose-derived stem cells (ADSCs) could adhere, proliferate, and differentiate on protein scaffolds. Thereafter, a hybrid CM was produced by culturing porcine ADSCs on kafirin scaffolds for 12 days, integrating plant protein-based and cell-based alternatives. The anthocyanins found in red sorghum contributed to the hybrid CM with meat-like color and antioxidative benefits. Moreover, the hybrid CM prototype demonstrated promising potential in providing higher protein content (22.9%) and unique mouthfeel and appearance characteristics, highlighting the viability of sorghum prolamin in promoting CM production.
Collapse
Affiliation(s)
- Lingshan Su
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou, Jiangsu 215123, China
- Department of Food Science and Technology, National University of Singapore, 2 Science Drive 2, Singapore 117542, Singapore
| | - Linzhi Jing
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou, Jiangsu 215123, China
| | - Shunjiang Zeng
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou, Jiangsu 215123, China
| | - Caili Fu
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou, Jiangsu 215123, China
| | - Dejian Huang
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou, Jiangsu 215123, China
- Department of Food Science and Technology, National University of Singapore, 2 Science Drive 2, Singapore 117542, Singapore
| |
Collapse
|
18
|
Asadikorayem M, Brunel LG, Weber P, Heilshorn SC, Zenobi-Wong M. Porosity dominates over microgel stiffness for promoting chondrogenesis in zwitterionic granular hydrogels. Biomater Sci 2024; 12:5504-5520. [PMID: 39347711 PMCID: PMC11441418 DOI: 10.1039/d4bm00233d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 09/15/2024] [Indexed: 10/01/2024]
Abstract
Granular hydrogels comprised of jammed, crosslinked microgels offer great potential as biomaterial scaffolds for cell-based therapies, including for cartilage tissue regeneration. As stiffness and porosity of hydrogels affect the phenotype of encapsulated cells and the extent of tissue regeneration, the design of tunable granular hydrogels to control and optimize these parameters is highly desirable. We hypothesized that chondrogenesis could be modulated using a granular hydrogel platform based on biocompatible, zwitterionic materials with independent intra- and inter-microgel crosslinking mechanisms. Microgels are made with mechanical fragmentation of photocrosslinked zwitterionic carboxybetaine acrylamide (CBAA) and sulfobetaine methacrylate (SBMA) hydrogels, and secondarily crosslinked in the presence of cells using horseradish peroxide (HRP) to produce cell-laden granular hydrogels. We varied the intra-microgel crosslinking density to produce microgels with varied stiffnesses (1-3 kPa) and swelling properties. These microgels, when resuspended at the same weight fraction and secondarily crosslinked, resulted in granular hydrogels with distinct porosities (5-40%) due to differing swelling properties. The greatest extent of chondrogenesis was achieved in scaffolds with the highest microgel stiffness and highest porosity. However, when scaffold porosity was kept constant and just microgel stiffness varied, cell phenotype and chondrogenesis were similar across scaffolds. These results indicate the dominant role of granular scaffold porosity on chondrogenesis, whereas microgel stiffness appears to play a relatively minor role. These observations are in contrast to cells encapsulated within conventional bulk hydrogels, where stiffness has been shown to significantly affect chondrocyte response. In summary, we introduce chemically-defined, zwitterionic biomaterials to fabricate versatile granular hydrogels allowing for tunable scaffold porosity and microgel stiffness to study and influence chondrogenesis.
Collapse
Affiliation(s)
- Maryam Asadikorayem
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland.
| | - Lucia G Brunel
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Patrick Weber
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland.
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Marcy Zenobi-Wong
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
19
|
Zhou J, Zhang Y, Fu Y, Li Q, Zhang J, Liu X, Gu Z. Visualizing and quantifying dynamic cellular forces with photonic crystal hydrogels. NANOSCALE 2024; 16:19074-19085. [PMID: 39319561 DOI: 10.1039/d4nr02834a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Cellular forces play a crucial role in numerous biological processes, including tissue development, morphogenesis, and disease progression. However, existing methods for detecting cellular forces, such as traction force microscopy and atomic force microscopy, often face limitations in terms of high throughput, real-time monitoring, and applicability to complex biological systems. In this study, we utilized a novel Photonic Crystal Cellular Force Microscopy (PCCFM) system to visualize and quantify dynamic cellular forces. This system consists of a conventional optical microscope and a photonic crystal substrate formed by the periodic arrangement of silica nanoparticles within polyacrylamide hydrogels. Taking MDCK cells and BMSCs as examples, we found that PCCFM can capture dynamic cellular forces with high spatial and temporal resolution during the cell adhesion, spread, proliferation, and osteogenic differentiation. The application of this technique revealed distinct force patterns in different cellular stages, offering insights into the interplay between cellular forces and morphological changes. By investigating the migration of cells from MDCK cyst fragments, we could gain significant insights into tumour cell migration behaviours. The real-time, high-throughput analysis of cellular biomechanics from the PCCFM system offers valuable information on the mechanisms of tumour metastasis, potentially guiding therapeutic development and improving disease treatment strategies.
Collapse
Affiliation(s)
- Jiankang Zhou
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Ying Zhang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Yifu Fu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Qiwei Li
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Jiajia Zhang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Xiaojiang Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Zhongze Gu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
20
|
Hernandez-Miranda ML, Xu D, Ben Issa AA, Johnston DA, Browne M, Cook RB, Sengers BG, Evans ND. Geometric constraint of mechanosensing by modification of hydrogel thickness prevents stiffness-induced differentiation in bone marrow stromal cells. J R Soc Interface 2024; 21:20240485. [PMID: 39353563 PMCID: PMC11444768 DOI: 10.1098/rsif.2024.0485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 10/04/2024] Open
Abstract
Extracellular matrix (ECM) stiffness is fundamental in cell division, movement and differentiation. The stiffness that cells sense is determined not only by the elastic modulus of the ECM material but also by ECM geometry and cell density. We hypothesized that these factors would influence cell traction-induced matrix deformations and cellular differentiation in bone marrow stromal cells (BMSCs). To achieve this, we cultivated BMSCs on polyacrylamide hydrogels that varied in elastic modulus and geometry and measured cell spreading, cell-imparted matrix deformations and differentiation. At low cell density BMSCs spread to a greater extent on stiff compared with soft hydrogels, or on thin compared with thick hydrogels. Cell-imparted matrix deformations were greater on soft compared with stiff hydrogels or thick compared with thin hydrogels. There were no significant differences in osteogenic differentiation relative to hydrogel elastic modulus and thickness. However, increased cell density and/or prolonged culture significantly reduced matrix deformations on soft hydrogels to levels similar to those on stiff substrates. This suggests that at high cell densities cell traction-induced matrix displacements are reduced by both neighbouring cells and the constraint imposed by an underlying stiff support. This may explain observations of the lack of difference in osteogenic differentiation as a function of stiffness.
Collapse
Affiliation(s)
- Maria L. Hernandez-Miranda
- Centre for Human Development, Stem Cells and Regenerative Medicine, Bone and Joint Research Group, Institute for Life Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Dichu Xu
- Ningbo Institute of Technology, Beihang University, Ningbo315800, People’s Republic of China
- Bioengineering Science Research Group, University of Southampton Faculty of Engineering and Physical Sciences, Southampton, UK
| | - Aya A. Ben Issa
- Centre for Human Development, Stem Cells and Regenerative Medicine, Bone and Joint Research Group, Institute for Life Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - David A. Johnston
- Biomedical Imaging Unit, University of Southampton Faculty of Medicine, Southampton, UK
| | - Martin Browne
- Bioengineering Science Research Group, University of Southampton Faculty of Engineering and Physical Sciences, Southampton, UK
| | - Richard B. Cook
- Bioengineering Science Research Group, University of Southampton Faculty of Engineering and Physical Sciences, Southampton, UK
| | - Bram G. Sengers
- Bioengineering Science Research Group, University of Southampton Faculty of Engineering and Physical Sciences, Southampton, UK
| | - Nicholas D. Evans
- Centre for Human Development, Stem Cells and Regenerative Medicine, Bone and Joint Research Group, Institute for Life Sciences, University of Southampton Faculty of Medicine, Southampton, UK
- Bioengineering Science Research Group, University of Southampton Faculty of Engineering and Physical Sciences, Southampton, UK
| |
Collapse
|
21
|
Mukundan LM, Das S, Rajasekaran R, Ganguly D, Seesala VS, Dhara S, Chattopadhyay S. Photo-annealable agarose microgels for jammed microgel printing: Transforming thermogelling hydrogel to a functional bioink. Int J Biol Macromol 2024; 278:134550. [PMID: 39116964 DOI: 10.1016/j.ijbiomac.2024.134550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/07/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Three-dimensional (3D) printing of hydrogel structures using jammed microgel inks offer distinct advantages of improved printing functionalities, as these inks are strain-yielding and self-recovering types. However, interparticle binding in granular hydrogel inks is a challenge to overcome the limited integrity and reduced macroscale modulus prevalent in the 3D printed microgel scaffolds. In this study, we prepared chemically annealable agarose microgels through a process of xerogel rehydration, applying a low-cost and high throughput method of spray drying. The crosslinked jammed microgel matrix is found to have superior mechanical properties with a Young's modulus of 2.23 MPa and extensibility up to 7.2%, surpassing those of traditional biopolymer-based and microgel-based inks. Furthermore, this study addresses the complexities encountered in the existing system of printing thermoresponsive agarose bioink using this jammed microgel printing approach. The jammed agarose microgel ink exhibited to be self-recovering, yield stress fluid and validated the temperature-independent printing. Furthermore, the 3D printed jammed microgel scaffold demonstrated good cell responsiveness as evaluated through the viability and morphological study in-vitro with mesenchymal stem cells cultured in it. This unique fabrication approach offers exciting possibilities to expand on microgel printing for varied requirements in tissue engineering.
Collapse
Affiliation(s)
- Lakshmi M Mukundan
- Rubber Technology Center, Indian Institute of Technology Kharagpur; School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Samir Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Ragavi Rajasekaran
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | | | - Venkata Sundeep Seesala
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Santanu Dhara
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | | |
Collapse
|
22
|
Gandin A, Torresan V, Panciera T, Brusatin G. A Scalable Method to Fabricate 2D Hydrogel Substrates for Mechanobiology Studies with Independent Tuning of Adhesiveness and Stiffness. Methods Protoc 2024; 7:75. [PMID: 39452789 PMCID: PMC11510107 DOI: 10.3390/mps7050075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 10/26/2024] Open
Abstract
Mechanical signals from the extracellular matrix are crucial in guiding cellular behavior. Two-dimensional hydrogel substrates for cell cultures serve as exceptional tools for mechanobiology studies because they mimic the biomechanical and adhesive characteristics of natural environments. However, the interdisciplinary knowledge required to synthetize and manipulate these biomaterials typically restricts their widespread use in biological laboratories, which may not have the material science expertise or specialized instrumentation. To address this, we propose a scalable method that requires minimal setup to produce 2D hydrogel substrates with independent modulation of the rigidity and adhesiveness within the range typical of natural tissues. In this method, norbornene-terminated 8-arm polyethylene glycol is stoichiometrically functionalized with RGD peptides and crosslinked with a di-cysteine terminated peptide via a thiol-ene click reaction. Since the synthesis process significantly influences the final properties of the hydrogels, we provide a detailed description of the chemical procedure to ensure reproducibility and high throughput results. We demonstrate examples of cell mechanosignaling by monitoring the activation state of the mechanoeffector proteins YAP/TAZ. This method effectively dissects the influence of biophysical and adhesive cues on cell behavior. We believe that our procedure will be easily adopted by other cell biology laboratories, improving its accessibility and practical application.
Collapse
Affiliation(s)
- Alessandro Gandin
- Department of Industrial Engineering, University of Padova, Via Marzolo 9, 35131 Padova, PD, Italy; (A.G.); (V.T.)
- Consorzio INSTM, Padova RU, Via Marzolo 9, 35131 Padova, PD, Italy
| | - Veronica Torresan
- Department of Industrial Engineering, University of Padova, Via Marzolo 9, 35131 Padova, PD, Italy; (A.G.); (V.T.)
- Consorzio INSTM, Padova RU, Via Marzolo 9, 35131 Padova, PD, Italy
| | - Tito Panciera
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, PD, Italy;
| | - Giovanna Brusatin
- Department of Industrial Engineering, University of Padova, Via Marzolo 9, 35131 Padova, PD, Italy; (A.G.); (V.T.)
- Consorzio INSTM, Padova RU, Via Marzolo 9, 35131 Padova, PD, Italy
| |
Collapse
|
23
|
Wang TC, Sawhney S, Morgan D, Bennett RL, Rashmi R, Estecio MR, Brock A, Singh I, Baer CF, Licht JD, Lele TP. Genetic variation drives cancer cell adaptation to ECM stiffness. Proc Natl Acad Sci U S A 2024; 121:e2403062121. [PMID: 39302966 PMCID: PMC11441511 DOI: 10.1073/pnas.2403062121] [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: 02/14/2024] [Accepted: 08/26/2024] [Indexed: 09/22/2024] Open
Abstract
The progression of many solid tumors is accompanied by temporal and spatial changes in the stiffness of the extracellular matrix (ECM). Cancer cells adapt to soft and stiff ECM through mechanisms that are not fully understood. It is well known that there is significant genetic heterogeneity from cell to cell in tumors, but how ECM stiffness as a parameter might interact with that genetic variation is not known. Here, we employed experimental evolution to study the response of genetically variable and clonal populations of tumor cells to variable ECM stiffness. Proliferation rates of genetically variable populations cultured on soft ECM increased over a period of several weeks, whereas clonal populations did not evolve. Tracking of DNA barcoded cell lineages revealed that soft ECM consistently selected for the same few variants. These data provide evidence that ECM stiffness exerts natural selection on genetically variable tumor populations. Soft-selected cells were highly migratory, with enriched oncogenic signatures and unusual behaviors such as spreading and traction force generation on ECMs with stiffness as low as 1 kPa. Rho-regulated cell spreading was found to be the directly selected trait, with yes-associated protein 1 translocation to the nucleus mediating fitness on soft ECM. Overall, these data show that genetic variation can drive cancer cell adaptation to ECM stiffness.
Collapse
Affiliation(s)
- Ting-Ching Wang
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX77843
| | - Suchitaa Sawhney
- Department of Biomedical Engineering, Texas A&M University, College Station, TX77843
| | - Daylin Morgan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX78712
| | - Richard L. Bennett
- Division of Hematology and Oncology, University of Florida Health Cancer Center, Gainesville, FL32610
| | - Richa Rashmi
- Department of Cell Biology and Genetics, Texas A&M University, Bryan, TX77807
| | - Marcos R. Estecio
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - Amy Brock
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX78712
| | - Irtisha Singh
- Department of Biomedical Engineering, Texas A&M University, College Station, TX77843
- Department of Cell Biology and Genetics, Texas A&M University, Bryan, TX77807
| | - Charles F. Baer
- Department of Biology, University of Florida, Gainesville, FL32611
| | - Jonathan D. Licht
- Division of Hematology and Oncology, University of Florida Health Cancer Center, Gainesville, FL32610
| | - Tanmay P. Lele
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX77843
- Department of Biomedical Engineering, Texas A&M University, College Station, TX77843
- Department of Translational Medical Sciences, Texas A&M University, Houston, TX77030
| |
Collapse
|
24
|
Prieto-López L, Pereiro X, Vecino E. The mechanics of the retina: Müller glia role on retinal extracellular matrix and modelling. Front Med (Lausanne) 2024; 11:1393057. [PMID: 39296899 PMCID: PMC11410058 DOI: 10.3389/fmed.2024.1393057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 08/13/2024] [Indexed: 09/21/2024] Open
Abstract
The retina is a highly heterogeneous tissue, both cell-wise but also regarding its extracellular matrix (ECM). The stiffness of the ECM is pivotal in retinal development and maturation and has also been associated with the onset and/or progression of numerous retinal pathologies, such as glaucoma, proliferative vitreoretinopathy (PVR), age-related macular degeneration (AMD), epiretinal membrane (ERM) formation or uveitis. Nonetheless, much remains unknown about the biomechanical milieu of the retina, and specifically the role that Müller glia play as principal mechanosensors and major producers of ECM constituents. So far, new approaches need to be developed to further the knowledge in the field of retinal mechanobiology for ECM-target applications to arise. In this review, we focus on the involvement of Müller glia in shaping and altering the retinal ECM under both physiological and pathological conditions and look into various biomaterial options to more accurately replicate the impact of matrix stiffness in vitro.
Collapse
Affiliation(s)
- Laura Prieto-López
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
| | - Xandra Pereiro
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
- Begiker-Ophthalmology Research Group, BioCruces Health Research Institute, Cruces Hospital, Barakaldo, Spain
| | - Elena Vecino
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
- Begiker-Ophthalmology Research Group, BioCruces Health Research Institute, Cruces Hospital, Barakaldo, Spain
| |
Collapse
|
25
|
Ahuja R, Shivhare V, Konar AD. Recent Advances in Smart Self-Assembled Bioinspired Hydrogels: A Bridging Weapon for Emerging Health Care Applications from Bench to Bedside. Macromol Rapid Commun 2024; 45:e2400255. [PMID: 38802265 DOI: 10.1002/marc.202400255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/16/2024] [Indexed: 05/29/2024]
Abstract
Stimuli-responsive low molecular weight hydrogel interventions for Biomedical challenges are a rapidly evolving paradigm in the bottom-up approach recently. Peptide-based self-assembled nano biomaterials present safer alternatives to their non-degradable counterparts as demanded for today's most urged clinical needs.Although a plethora of work has already been accomplished, programming hydrogelators with appropriate functionalities requires a better understanding as the impact of the macromolecular structure of the peptides and subsequently, their self-assembled nanostructures remain unidentified. Henceforth this review focuses on two aspects: Firstly, the underlying guidelines for building biomimetic strategies to tailor scaffolds leading to hydrogelation along with the role of non-covalent interactions that are the key components of various self-assembly processes. In the second section, it is aimed to bring together the recent achievements with designer assembly concerning their self-aggregation behaviour and applications mainly in the biomedical arena like drug delivery carrier design, antimicrobial, anti-inflammatory as well as wound healing materials. Furthermore, it is anticipated that this article will provide a conceptual demonstration of the different approaches taken towards the construction of these task-specific designer hydrogels. Finally, a collective effort among the material scientists is required to pave the path for the entrance of these intelligent materials into medicine from bench to bedside.
Collapse
Affiliation(s)
- Rishabh Ahuja
- Department of Applied Chemistry, Rajiv Gandhi Technological University, Bhopal, Madhya Pradesh, 462033, India
| | - Vaibhav Shivhare
- Department of Applied Chemistry, Rajiv Gandhi Technological University, Bhopal, Madhya Pradesh, 462033, India
| | - Anita Dutt Konar
- Department of Applied Chemistry, Rajiv Gandhi Technological University, Bhopal, Madhya Pradesh, 462033, India
- School of Pharmaceutical Sciences, Rajiv Gandhi Technological University, Bhopal, Madhya Pradesh, 462033, India
- University Grants Commission, New Delhi, 110002, India
| |
Collapse
|
26
|
Llewellyn J, Charrier A, Cuciniello R, Helfer E, Dono R. Substrate stiffness alters layer architecture and biophysics of human induced pluripotent stem cells to modulate their differentiation potential. iScience 2024; 27:110557. [PMID: 39175774 PMCID: PMC11340605 DOI: 10.1016/j.isci.2024.110557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/10/2024] [Accepted: 07/17/2024] [Indexed: 08/24/2024] Open
Abstract
Lineage-specific differentiation of human induced pluripotent stem cells (hiPSCs) relies on complex interactions between biochemical and physical cues. Here we investigated the ability of hiPSCs to undergo lineage commitment in response to inductive signals and assessed how this competence is modulated by substrate stiffness. We showed that Activin A-induced hiPSC differentiation into mesendoderm and its derivative, definitive endoderm, is enhanced on gel-based substrates softer than glass. This correlated with changes in tight junction formation and extensive cytoskeletal remodeling. Further, live imaging and biophysical studies suggested changes in cell motility and interfacial contacts underlie hiPSC layer reshaping on soft substrates. Finally, we repurposed an ultra-soft silicone gel, which may provide a suitable substrate for culturing hiPSCs at physiological stiffnesses. Our results provide mechanistic insight into how epithelial mechanics dictate the hiPSC response to chemical signals and provide a tool for their efficient differentiation in emerging stem cell therapies.
Collapse
Affiliation(s)
- Jack Llewellyn
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, NeuroMarseille, Marseille, France
- Aix Marseille University, CNRS, CINAM, Turing Centre for Living Systems, 13009 Marseille, France
| | - Anne Charrier
- Aix Marseille University, CNRS, CINAM, Turing Centre for Living Systems, 13009 Marseille, France
| | - Rossana Cuciniello
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, NeuroMarseille, Marseille, France
| | - Emmanuèle Helfer
- Aix Marseille University, CNRS, CINAM, Turing Centre for Living Systems, 13009 Marseille, France
| | - Rosanna Dono
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, NeuroMarseille, Marseille, France
| |
Collapse
|
27
|
Liu X, Zhang W, Gu J, Wang J, Wang Y, Xu Z. Single-cell SERS imaging of dual cell membrane receptors expression influenced by extracellular matrix stiffness. J Colloid Interface Sci 2024; 668:335-342. [PMID: 38678888 DOI: 10.1016/j.jcis.2024.04.170] [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: 02/09/2024] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
Membrane receptors perform a diverse range of cellular functions, accounting for more than half of all drug targets. The mechanical microenvironment regulates cell behaviors and phenotype. However, conventional analysis methods of membrane receptors often ignore the effects of the extracellular matrix stiffness, failing to reveal the heterogeneity of cell membrane receptors expression. Herein, we developed an in-situ surface-enhanced Raman scattering (SERS) imaging method to visualize single-cell membrane receptors on substrates with different stiffness. Two SERS substrates, Au@4-mercaptobenzonitrile@Ag@Sgc8c and Au@4-pethynylaniline@Ag@SYL3c, were employed to specifically target protein tyrosine kinase-7 (PTK7) and epithelial cell adhesion molecule (EpCAM), respectively. The polyacrylamide (PA) gels with tunable stiffness (2.5-25 kPa) were constructed to mimic extracellular matrix. The simultaneous SERS imaging of dual membrane receptors on single cancer cells on substrates with different stiffness was achieved. Our findings reveal decreased expression of PTK7 and EpCAM on cells cultured on stiffer substrates and higher migration ability of the cells. The results elucidate the heterogeneity of membrane receptors expression of cells cultured on the substrates with different stiffness. This single-cell analysis method offers an in-situ platform for investigating the impacts of extracellular matrix stiffness on the expression of membrane receptors, providing insights into the role of cell membrane receptors in cancer metastasis.
Collapse
Affiliation(s)
- Xiaopeng Liu
- Research Center for Analytical Sciences, Northeastern University, Shenyang 110819, PR China
| | - Wenshu Zhang
- Research Center for Analytical Sciences, Northeastern University, Shenyang 110819, PR China
| | - Jiahui Gu
- Research Center for Analytical Sciences, Northeastern University, Shenyang 110819, PR China
| | - Jie Wang
- Research Center for Analytical Sciences, Northeastern University, Shenyang 110819, PR China
| | - Yue Wang
- Research Center for Analytical Sciences, Northeastern University, Shenyang 110819, PR China
| | - Zhangrun Xu
- Research Center for Analytical Sciences, Northeastern University, Shenyang 110819, PR China.
| |
Collapse
|
28
|
Calzuola ST, Newman G, Feaugas T, Perrault CM, Blondé JB, Roy E, Porrini C, Stojanovic GM, Vidic J. Membrane-based microfluidic systems for medical and biological applications. LAB ON A CHIP 2024; 24:3579-3603. [PMID: 38954466 DOI: 10.1039/d4lc00251b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Microfluidic devices with integrated membranes that enable control of mass transport in constrained environments have shown considerable growth over the last decade. Membranes are a key component in several industrial processes such as chemical, pharmaceutical, biotechnological, food, and metallurgy separation processes as well as waste management applications, allowing for modular and compact systems. Moreover, the miniaturization of a process through microfluidic devices leads to process intensification together with reagents, waste and cost reduction, and energy and space savings. The combination of membrane technology and microfluidic devices allows therefore magnification of their respective advantages, providing more valuable solutions not only for industrial processes but also for reproducing biological processes. This review focuses on membrane-based microfluidic devices for biomedical science with an emphasis on microfluidic artificial organs and organs-on-chip. We provide the basic concepts of membrane technology and the laws governing mass transport. The role of the membrane in biomedical microfluidic devices, along with the required properties, available materials, and current challenges are summarized. We believe that the present review may be a starting point and a resource for researchers who aim to replicate a biological phenomenon on-chip by applying membrane technology, for moving forward the biomedical applications.
Collapse
Affiliation(s)
- Silvia Tea Calzuola
- UMR7646 Laboratoire d'hydrodynamique (LadHyX), Ecole Polytechnique, Palaiseau, France.
- Eden Tech, Paris, France
| | - Gwenyth Newman
- Eden Tech, Paris, France
- Department of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Thomas Feaugas
- Eden Tech, Paris, France
- Department of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Milan, Italy
| | | | | | | | | | - Goran M Stojanovic
- Faculty of Technical Sciences, University of Novi Sad, T. D. Obradovića 6, 21000 Novi Sad, Serbia
| | - Jasmina Vidic
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
29
|
Chen H, Wang S, Cao Y, Lei H. Molecular Force Sensors for Biological Application. Int J Mol Sci 2024; 25:6198. [PMID: 38892386 PMCID: PMC11173168 DOI: 10.3390/ijms25116198] [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: 04/29/2024] [Revised: 05/29/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
The mechanical forces exerted by cells on their surrounding microenvironment are known as cellular traction forces. These forces play crucial roles in various biological processes, such as tissue development, wound healing and cell functions. However, it is hard for traditional techniques to measure cellular traction forces accurately because their magnitude (from pN to nN) and the length scales over which they occur (from nm to μm) are extremely small. In order to fully understand mechanotransduction, highly sensitive tools for measuring cellular forces are needed. Current powerful techniques for measuring traction forces include traction force microscopy (TFM) and fluorescent molecular force sensors (FMFS). In this review, we elucidate the force imaging principles of TFM and FMFS. Then we highlight the application of FMFS in a variety of biological processes and offer our perspectives and insights into the potential applications of FMFS.
Collapse
Affiliation(s)
- Huiyan Chen
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing 210093, China; (H.C.); (S.W.)
| | - Shouhan Wang
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing 210093, China; (H.C.); (S.W.)
| | - Yi Cao
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing 210093, China; (H.C.); (S.W.)
| | - Hai Lei
- School of Physics, Zhejiang University, Hangzhou 310027, China
- Institute for Advanced Study in Physics, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
30
|
Ferrai C, Schulte C. Mechanotransduction in stem cells. Eur J Cell Biol 2024; 103:151417. [PMID: 38729084 DOI: 10.1016/j.ejcb.2024.151417] [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: 12/27/2023] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Nowadays, it is an established concept that the capability to reach a specialised cell identity via differentiation, as in the case of multi- and pluripotent stem cells, is not only determined by biochemical factors, but that also physical aspects of the microenvironment play a key role; interpreted by the cell through a force-based signalling pathway called mechanotransduction. However, the intricate ties between the elements involved in mechanotransduction, such as the extracellular matrix, the glycocalyx, the cell membrane, Integrin adhesion complexes, Cadherin-mediated cell/cell adhesion, the cytoskeleton, and the nucleus, are still far from being understood in detail. Here we report what is currently known about these elements in general and their specific interplay in the context of multi- and pluripotent stem cells. We furthermore merge this overview to a more comprehensive picture, that aims to cover the whole mechanotransductive pathway from the cell/microenvironment interface to the regulation of the chromatin structure in the nucleus. Ultimately, with this review we outline the current picture of the interplay between mechanotransductive cues and epigenetic regulation and how these processes might contribute to stem cell dynamics and fate.
Collapse
Affiliation(s)
- Carmelo Ferrai
- Institute of Pathology, University Medical Centre Göttingen, Germany.
| | - Carsten Schulte
- Department of Biomedical and Clinical Sciences and Department of Physics "Aldo Pontremoli", University of Milan, Italy.
| |
Collapse
|
31
|
Bozuyuk U, Wrede P, Yildiz E, Sitti M. Roadmap for Clinical Translation of Mobile Microrobotics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311462. [PMID: 38380776 DOI: 10.1002/adma.202311462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/24/2024] [Indexed: 02/22/2024]
Abstract
Medical microrobotics is an emerging field to revolutionize clinical applications in diagnostics and therapeutics of various diseases. On the other hand, the mobile microrobotics field has important obstacles to pass before clinical translation. This article focuses on these challenges and provides a roadmap of medical microrobots to enable their clinical use. From the concept of a "magic bullet" to the physicochemical interactions of microrobots in complex biological environments in medical applications, there are several translational steps to consider. Clinical translation of mobile microrobots is only possible with a close collaboration between clinical experts and microrobotics researchers to address the technical challenges in microfabrication, safety, and imaging. The clinical application potential can be materialized by designing microrobots that can solve the current main challenges, such as actuation limitations, material stability, and imaging constraints. The strengths and weaknesses of the current progress in the microrobotics field are discussed and a roadmap for their clinical applications in the near future is outlined.
Collapse
Affiliation(s)
- Ugur Bozuyuk
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
| | - Paul Wrede
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
- Institute for Biomedical Engineering, ETH Zurich, Zurich, 8093, Switzerland
| | - Erdost Yildiz
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
| | - Metin Sitti
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
- School of Medicine and College of Engineering, Koc University, Istanbul, 34450, Turkey
| |
Collapse
|
32
|
Su L, Jing L, Zeng S, Fu C, Huang D. 3D Porous Edible Scaffolds from Rye Secalin for Cell-Based Pork Fat Tissue Culturing. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:11587-11596. [PMID: 38728660 DOI: 10.1021/acs.jafc.3c09713] [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: 05/12/2024]
Abstract
Cellular agriculture holds hope for a sustainable alternative to conventional meat, yet multiple technical challenges remain. These include the large-scale production of edible scaffolds and culturing methods for fat tissues, which are key to meat texture, flavor, and nutritional values. Herein. we disclose our method in the facile fabrication of sponge-like plant protein scaffolds by applying commercial sugar cubes as highly permeable templates. The prepared secalin scaffolds feature a high porosity of 85-90%, fully interconnected pores, and high water stability. The mechanical properties of scaffolds could be tuned by varying sugar-to-protein weight ratio and post-water annealing treatment. Moreover, murine preadipocytes (3T3-L1) and porcine adipose-derived stem cells (ADSCs) readily infiltrate, adhere, proliferate, and differentiate on the secalin scaffolds to develop a fat tissue morphology. A cultured fat tissue was produced by culturing porcine ADSCs for 12 days, which remarkably resembles conventional porcine subcutaneous adipose tissue in appearance, texture, flavor, and fatty acid profiles. The research demonstrates the great potential of sponge-like secalin scaffolds for cultured fat tissue production.
Collapse
Affiliation(s)
- Lingshan Su
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou, Jiangsu 215123, China
- Department of Food Science and Technology, National University of Singapore, 2 Science Drive 2, Singapore 117542, Singapore
| | - Linzhi Jing
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou, Jiangsu 215123, China
| | - Shunjiang Zeng
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou, Jiangsu 215123, China
| | - Caili Fu
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou, Jiangsu 215123, China
| | - Dejian Huang
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou, Jiangsu 215123, China
- Department of Food Science and Technology, National University of Singapore, 2 Science Drive 2, Singapore 117542, Singapore
| |
Collapse
|
33
|
Ryoo H, Kimmel H, Rondo E, Underhill GH. Advances in high throughput cell culture technologies for therapeutic screening and biological discovery applications. Bioeng Transl Med 2024; 9:e10627. [PMID: 38818120 PMCID: PMC11135158 DOI: 10.1002/btm2.10627] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 06/01/2024] Open
Abstract
Cellular phenotypes and functional responses are modulated by the signals present in their microenvironment, including extracellular matrix (ECM) proteins, tissue mechanical properties, soluble signals and nutrients, and cell-cell interactions. To better recapitulate and analyze these complex signals within the framework of more physiologically relevant culture models, high throughput culture platforms can be transformative. High throughput methodologies enable scientists to extract increasingly robust and broad datasets from individual experiments, screen large numbers of conditions for potential hits, better qualify and predict responses for preclinical applications, and reduce reliance on animal studies. High throughput cell culture systems require uniformity, assay miniaturization, specific target identification, and process simplification. In this review, we detail the various techniques that researchers have used to face these challenges and explore cellular responses in a high throughput manner. We highlight several common approaches including two-dimensional multiwell microplates, microarrays, and microfluidic cell culture systems as well as unencapsulated and encapsulated three-dimensional high throughput cell culture systems, featuring multiwell microplates, micromolds, microwells, microarrays, granular hydrogels, and cell-encapsulated microgels. We also discuss current applications of these high throughput technologies, namely stem cell sourcing, drug discovery and predictive toxicology, and personalized medicine, along with emerging opportunities and future impact areas.
Collapse
Affiliation(s)
- Hyeon Ryoo
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Hannah Kimmel
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Evi Rondo
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Gregory H. Underhill
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| |
Collapse
|
34
|
Soliman BG, Longoni A, Major GS, Lindberg GCJ, Choi YS, Zhang YS, Woodfield TBF, Lim KS. Harnessing Macromolecular Chemistry to Design Hydrogel Micro- and Macro-Environments. Macromol Biosci 2024; 24:e2300457. [PMID: 38035637 DOI: 10.1002/mabi.202300457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/16/2023] [Indexed: 12/02/2023]
Abstract
Cell encapsulation within three-dimensional hydrogels is a promising approach to mimic tissues. However, true biomimicry of the intricate microenvironment, biophysical and biochemical gradients, and the macroscale hierarchical spatial organizations of native tissues is an unmet challenge within tissue engineering. This review provides an overview of the macromolecular chemistries that have been applied toward the design of cell-friendly hydrogels, as well as their application toward controlling biophysical and biochemical bulk and gradient properties of the microenvironment. Furthermore, biofabrication technologies provide the opportunity to simultaneously replicate macroscale features of native tissues. Biofabrication strategies are reviewed in detail with a particular focus on the compatibility of these strategies with the current macromolecular toolkit described for hydrogel design and the challenges associated with their clinical translation. This review identifies that the convergence of the ever-expanding macromolecular toolkit and technological advancements within the field of biofabrication, along with an improved biological understanding, represents a promising strategy toward the successful tissue regeneration.
Collapse
Affiliation(s)
- Bram G Soliman
- School of Materials Science and Engineering, University of New South Wales, Sydney, 2052, Australia
| | - Alessia Longoni
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, 3584CX, The Netherlands
| | - Gretel S Major
- Department of Orthopedic Surgery and Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand
| | - Gabriella C J Lindberg
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR, 97403, USA
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Perth, 6009, Australia
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02115, USA
| | - Tim B F Woodfield
- Department of Orthopedic Surgery and Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand
| | - Khoon S Lim
- Department of Orthopedic Surgery and Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand
- School of Medical Sciences, University of Sydney, Sydney, 2006, Australia
- Charles Perkins Centre, University of Sydney, Sydney, 2006, Australia
| |
Collapse
|
35
|
Karimi A, Aga M, Khan T, D'costa SD, Thaware O, White E, Kelley MJ, Gong H, Acott TS. Comparative analysis of traction forces in normal and glaucomatous trabecular meshwork cells within a 3D, active fluid-structure interaction culture environment. Acta Biomater 2024; 180:206-229. [PMID: 38641184 PMCID: PMC11095374 DOI: 10.1016/j.actbio.2024.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/26/2024] [Accepted: 04/11/2024] [Indexed: 04/21/2024]
Abstract
This study presents a 3D in vitro cell culture model, meticulously 3D printed to replicate the conventional aqueous outflow pathway anatomical structure, facilitating the study of trabecular meshwork (TM) cellular responses under glaucomatous conditions. Glaucoma affects TM cell functionality, leading to extracellular matrix (ECM) stiffening, enhanced cell-ECM adhesion, and obstructed aqueous humor outflow. Our model, reconstructed from polyacrylamide gel with elastic moduli of 1.5 and 21.7 kPa, is based on serial block-face scanning electron microscopy images of the outflow pathway. It allows for quantifying 3D, depth-dependent, dynamic traction forces exerted by both normal and glaucomatous TM cells within an active fluid-structure interaction (FSI) environment. In our experimental design, we designed two scenarios: a control group with TM cells observed over 20 hours without flow (static setting), focusing on intrinsic cellular contractile forces, and a second scenario incorporating active FSI to evaluate its impact on traction forces (dynamic setting). Our observations revealed that active FSI results in higher traction forces (normal: 1.83-fold and glaucoma: 2.24-fold) and shear strains (normal: 1.81-fold and glaucoma: 2.41-fold), with stiffer substrates amplifying this effect. Glaucomatous cells consistently exhibited larger forces than normal cells. Increasing gel stiffness led to enhanced stress fiber formation in TM cells, particularly in glaucomatous cells. Exposure to active FSI dramatically altered actin organization in both normal and glaucomatous TM cells, particularly affecting cortical actin stress fiber arrangement. This model while preliminary offers a new method in understanding TM cell biomechanics and ECM stiffening in glaucoma, highlighting the importance of FSI in these processes. STATEMENT OF SIGNIFICANCE: This pioneering project presents an advanced 3D in vitro model, meticulously replicating the human trabecular meshwork's anatomy for glaucoma research. It enables precise quantification of cellular forces in a dynamic fluid-structure interaction, a leap forward from existing 2D models. This advancement promises significant insights into trabecular meshwork cell biomechanics and the stiffening of the extracellular matrix in glaucoma, offering potential pathways for innovative treatments. This research is positioned at the forefront of ocular disease study, with implications that extend to broader biomedical applications.
Collapse
Affiliation(s)
- Alireza Karimi
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States; Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, United States.
| | - Mini Aga
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States
| | - Taaha Khan
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States
| | - Siddharth Daniel D'costa
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States
| | - Omkar Thaware
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States; Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, United States
| | - Elizabeth White
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States
| | - Mary J Kelley
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States; Department Integrative Biosciences, School of Dentistry, Oregon Health & Science University, Portland, OR, United States
| | - Haiyan Gong
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA, United States; Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Ted S Acott
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States; Department Chemical Physiology & Biochemistry, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
36
|
Li L, Griebel ME, Uroz M, Bubli SY, Gagnon KA, Trappmann B, Baker BM, Eyckmans J, Chen CS. A Protein-Adsorbent Hydrogel with Tunable Stiffness for Tissue Culture Demonstrates Matrix-Dependent Stiffness Responses. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2309567. [PMID: 38693998 PMCID: PMC11060701 DOI: 10.1002/adfm.202309567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Indexed: 05/03/2024]
Abstract
Although tissue culture plastic has been widely employed for cell culture, the rigidity of plastic is not physiologic. Softer hydrogels used to culture cells have not been widely adopted in part because coupling chemistries are required to covalently capture extracellular matrix (ECM) proteins and support cell adhesion. To create an in vitro system with tunable stiffnesses that readily adsorbs ECM proteins for cell culture, we present a novel hydrophobic hydrogel system via chemically converting hydroxyl residues on the dextran backbone to methacrylate groups, thereby transforming non-protein adhesive, hydrophilic dextran to highly protein adsorbent substrates. Increasing methacrylate functionality increases the hydrophobicity in the resulting hydrogels and enhances ECM protein adsorption without additional chemical reactions. These hydrophobic hydrogels permit facile and tunable modulation of substrate stiffness independent of hydrophobicity or ECM coatings. Using this approach, we show that substrate stiffness and ECM adsorption work together to affect cell morphology and proliferation, but the strengths of these effects vary in different cell types. Furthermore, we reveal that stiffness mediated differentiation of dermal fibroblasts into myofibroblasts is modulated by the substrate ECM. Our material system demonstrates remarkable simplicity and flexibility to tune ECM coatings and substrate stiffness and study their effects on cell function.
Collapse
Affiliation(s)
- Linqing Li
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 02215, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, 02115, United States
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, New Hampshire, 03824, United States
| | - Megan E Griebel
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 02215, United States
| | - Marina Uroz
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 02215, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, 02115, United States
| | - Saniya Yesmin Bubli
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, New Hampshire, 03824, United States
| | - Keith A Gagnon
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 02215, United States
| | - Britta Trappmann
- Bioactive Materials Laboratory, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, Münster, 48149 Germany
| | - Brendon M Baker
- Engineered Microenvironments and Mechanobiology Lab, Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109 United States
| | - Jeroen Eyckmans
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 02215, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, 02115, United States
| | - Christopher S Chen
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 02215, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, 02115, United States
| |
Collapse
|
37
|
Koch KC, Jadon N, Thesmar I, Tew GN, Minter LM. Combating bone marrow failure with polymer materials. Front Immunol 2024; 15:1396486. [PMID: 38694497 PMCID: PMC11061490 DOI: 10.3389/fimmu.2024.1396486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/03/2024] [Indexed: 05/04/2024] Open
Abstract
Bone marrow failure (BMF) has become one of the most studied autoimmune disorders, particularly due to its prevalence both as an inherited disease, but also as a result of chemotherapies. BMF is associated with severe symptoms such as bleeding episodes and susceptibility to infections, and often has underlying characteristics, such as anemia, thrombocytopenia, and neutropenia. The current treatment landscape for BMF requires stem cell transplantation or chemotherapies to induce immune suppression. However, there is limited donor cell availability or dose related toxicity associated with these treatments. Optimizing these treatments has become a necessity. Polymer-based materials have become increasingly popular, as current research efforts are focused on synthesizing novel cell matrices for stem cell expansion to solve limited donor cell availability, as well as applying polymer delivery vehicles to intracellularly deliver cargo that can aid in immunosuppression. Here, we discuss the importance and impact of polymer materials to enhance therapeutics in the context of BMF.
Collapse
Affiliation(s)
- Kayla C. Koch
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA, United States
| | - Nidhi Jadon
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| | - Iris Thesmar
- University of Massachusetts Amherst, Amherst, MA, United States
| | - Gregory N. Tew
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA, United States
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
- University of Massachusetts Amherst, Amherst, MA, United States
| | - Lisa M. Minter
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
- University of Massachusetts Amherst, Amherst, MA, United States
| |
Collapse
|
38
|
Jia K, You J, Zhu Y, Li M, Chen S, Ren S, Chen S, Zhang J, Wang H, Zhou Y. Platelet-rich fibrin as an autologous biomaterial for bone regeneration: mechanisms, applications, optimization. Front Bioeng Biotechnol 2024; 12:1286035. [PMID: 38689760 PMCID: PMC11058865 DOI: 10.3389/fbioe.2024.1286035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 03/22/2024] [Indexed: 05/02/2024] Open
Abstract
Platelet-rich fibrin, a classical autologous-derived bioactive material, consists of a fibrin scaffold and its internal loading of growth factors, platelets, and leukocytes, with the gradual degradation of the fibrin scaffold and the slow release of physiological doses of growth factors. PRF promotes vascular regeneration, promotes the proliferation and migration of osteoblast-related cells such as mesenchymal cells, osteoblasts, and osteoclasts while having certain immunomodulatory and anti-bacterial effects. PRF has excellent osteogenic potential and has been widely used in the field of bone tissue engineering and dentistry. However, there are still some limitations of PRF, and the improvement of its biological properties is one of the most important issues to be solved. Therefore, it is often combined with bone tissue engineering scaffolds to enhance its mechanical properties and delay its degradation. In this paper, we present a systematic review of the development of platelet-rich derivatives, the structure and biological properties of PRF, osteogenic mechanisms, applications, and optimization to broaden their clinical applications and provide guidance for their clinical translation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yanmin Zhou
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
| |
Collapse
|
39
|
Meli V, Rowley AT, Veerasubramanian PK, Heedy SE, Liu WF, Wang SW. Modulation of Stiffness-Dependent Macrophage Inflammatory Responses by Collagen Deposition. ACS Biomater Sci Eng 2024; 10:2212-2223. [PMID: 38467019 PMCID: PMC11005009 DOI: 10.1021/acsbiomaterials.3c01892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 03/13/2024]
Abstract
Macrophages are innate immune cells that interact with complex extracellular matrix environments, which have varied stiffness, composition, and structure, and such interactions can lead to the modulation of cellular activity. Collagen is often used in the culture of immune cells, but the effects of substrate functionalization conditions are not typically considered. Here, we show that the solvent system used to attach collagen onto a hydrogel surface affects its surface distribution and organization, and this can modulate the responses of macrophages subsequently cultured on these surfaces in terms of their inflammatory activation and expression of adhesion and mechanosensitive molecules. Collagen was solubilized in either acetic acid (Col-AA) or N-(2-hydroxyethyl)piperazine-N'-ethanesulfonic acid (HEPES) (Col-HEP) solutions and conjugated onto soft and stiff polyacrylamide (PA) hydrogel surfaces. Bone marrow-derived macrophages cultured under standard conditions (pH 7.4) on the Col-HEP-derived surfaces exhibited stiffness-dependent inflammatory activation; in contrast, the macrophages cultured on Col-AA-derived surfaces expressed high levels of inflammatory cytokines and genes, irrespective of the hydrogel stiffness. Among the collagen receptors that were examined, leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) was the most highly expressed, and knockdown of the Lair-1 gene enhanced the secretion of inflammatory cytokines. We found that the collagen distribution was more homogeneous on Col-AA surfaces but formed aggregates on Col-HEP surfaces. The macrophages cultured on Col-AA PA hydrogels were more evenly spread, expressed higher levels of vinculin, and exerted higher traction forces compared to those of cells on Col-HEP. These macrophages on Col-AA also had higher nuclear-to-cytoplasmic ratios of yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ), key molecules that control inflammation and sense substrate stiffness. Our results highlight that seemingly slight variations in substrate deposition for immunobiology studies can alter critical immune responses, and this is important to elucidate in the broader context of immunomodulatory biomaterial design.
Collapse
Affiliation(s)
- Vijaykumar
S. Meli
- Department
of Biomedical Engineering, University of
California Irvine, Irvine, California 92697, United States
- UCI
Edwards Lifesciences Foundation Cardiovascular Innovation and Research
Center, University of California Irvine, Irvine, California 92697, United States
- Department
of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California 92697, United States
| | - Andrew T. Rowley
- Department
of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California 92697, United States
| | - Praveen K. Veerasubramanian
- Department
of Biomedical Engineering, University of
California Irvine, Irvine, California 92697, United States
- UCI
Edwards Lifesciences Foundation Cardiovascular Innovation and Research
Center, University of California Irvine, Irvine, California 92697, United States
| | - Sara E. Heedy
- Department
of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California 92697, United States
| | - Wendy F. Liu
- Department
of Biomedical Engineering, University of
California Irvine, Irvine, California 92697, United States
- UCI
Edwards Lifesciences Foundation Cardiovascular Innovation and Research
Center, University of California Irvine, Irvine, California 92697, United States
- Department
of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California 92697, United States
- Department
of Molecular Biology and Biochemistry, University
of California Irvine, Irvine, California 92697, United States
- Institute
for Immunology, University of California
Irvine, Irvine, California 92697, United States
| | - Szu-Wen Wang
- Department
of Biomedical Engineering, University of
California Irvine, Irvine, California 92697, United States
- Department
of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California 92697, United States
- Institute
for Immunology, University of California
Irvine, Irvine, California 92697, United States
- Chao Family
Comprehensive Cancer Center, University
of California Irvine, Irvine, California 92697, United States
| |
Collapse
|
40
|
Dong X, Sun Q, Geng J, Liu X, Wei Q. Fiber Flexibility Reconciles Matrix Recruitment and the Fiber Modulus to Promote Cell Mechanosensing. NANO LETTERS 2024; 24:4029-4037. [PMID: 38526438 DOI: 10.1021/acs.nanolett.4c00923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
The mechanical interaction between cells and the extracellular matrix is pervasive in biological systems. On fibrous substrates, cells possess the ability to recruit neighboring fibers, thereby augmenting their own adhesion and facilitating the generation of mechanical cues. However, the matrices with high moduli impede fiber recruitment, restricting the cell mechanoresponse. Herein, by harnessing the inherent swelling properties of gelatin, the flexible gelatin methacryloyl network empowers cells to recruit fibers spanning a broad spectrum of physiological moduli during adhesion. The high flexibility concurrently facilitates the optimization of fiber distribution, deformability, and modulus, contributing to the promotion of cell mechanosensing. Consequently, the randomly distributed flexible fibers with high moduli maximize the cell adhesive forces. This study uncovers the impact of fiber recruitment on cell mechanosensing and introduces fiber flexibility as a previously unexplored property, offering an innovative perspective for the design and development of novel biomaterials.
Collapse
Affiliation(s)
- Xiangyu Dong
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China
- Department of Nephrology, Kidney Research Institute, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Qian Sun
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Jiwen Geng
- Department of Nephrology, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Xiaojing Liu
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, and Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, and Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, P. R. China
| | - Qiang Wei
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China
| |
Collapse
|
41
|
Zhang Y, Dai J, Hang R, Yao X, Bai L, Huang D, Hang R. Impact of surface biofunctionalization strategies on key effector cells response in polyacrylamide hydrogels for bone regeneration. BIOMATERIALS ADVANCES 2024; 158:213768. [PMID: 38237320 DOI: 10.1016/j.bioadv.2024.213768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/24/2023] [Accepted: 01/10/2024] [Indexed: 03/03/2024]
Abstract
Despite the clinical prevalence of various bone defect repair materials, a full understanding of their influence on bone repair and regeneration remains elusive. This study focuses on poly(acrylamide) (PAAm) hydrogels, popular 2D model substrates, which have regulable mechanical properties within physiological. However, their bio-inert nature requires surface biofunctionalization to enhance cell-material interactions and facilitate the study of bone repair mechanisms. We utilized PAAm hydrogels of varying stiffness (18, 76 and 295 kPa), employed sulfosuccinimidyl-6-(4'-azido-2'-nitropheny-lamino) hexanoate (sulfo-SANPAH) and N-(3-dimethylaminopropyl)-N-ethylcarbodiimide hydrochloride/N-hydroxysuccinimidyl acrylate (EDC/NHS) as crosslinkers, and cultured macrophages, endothelial cells, and bone mesenchymal stem cells on these hydrogels. Our findings indicated that sulfo-SANPAH's crosslinking efficiency surpassed that of EDC/NHS, irrespective of pore size and stiffness. Importantly, we observed that the stiffness and surface biofunctionalization method of hydrogels significantly impacted cell adhesion and proliferation. The collagen-modified hydrogels by EDC/NHS strategy failed to support the normal biological behavior of bone mesenchymal stem cells and hindered endothelial cell spreading. In contrast, these modified hydrogels by the sulfo-SANPAH method showed good cytocompatibility with the three types of cells. This study underscores the critical role of appropriate conjugation strategies for PAAm hydrogels, providing valuable insights for hydrogel surface modification in bone repair and regeneration research.
Collapse
Affiliation(s)
- Yi Zhang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Jinjun Dai
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Ruiyue Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Xiaohong Yao
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China.
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China.
| | - Di Huang
- Research Center for Nano-Biomaterials & Regenerative Medicine, Department of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China; Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030060, China
| | - Ruiqiang Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China.
| |
Collapse
|
42
|
Kersey AL, Cheng DY, Deo KA, Dubell CR, Wang TC, Jaiswal MK, Kim MH, Murali A, Hargett SE, Mallick S, Lele TP, Singh I, Gaharwar AK. Stiffness assisted cell-matrix remodeling trigger 3D mechanotransduction regulatory programs. Biomaterials 2024; 306:122473. [PMID: 38335719 DOI: 10.1016/j.biomaterials.2024.122473] [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: 07/11/2023] [Revised: 11/13/2023] [Accepted: 01/16/2024] [Indexed: 02/12/2024]
Abstract
Engineered matrices provide a valuable platform to understand the impact of biophysical factors on cellular behavior such as migration, proliferation, differentiation, and tissue remodeling, through mechanotransduction. While recent studies have identified some mechanisms of 3D mechanotransduction, there is still a critical knowledge gap in comprehending the interplay between 3D confinement, ECM properties, and cellular behavior. Specifically, the role of matrix stiffness in directing cellular fate in 3D microenvironment, independent of viscoelasticity, microstructure, and ligand density remains poorly understood. To address this gap, we designed a nanoparticle crosslinker to reinforce collagen-based hydrogels without altering their chemical composition, microstructure, viscoelasticity, and density of cell-adhesion ligand and utilized it to understand cellular dynamics. This crosslinking mechanism utilizes nanoparticles as crosslink epicenter, resulting in 10-fold increase in mechanical stiffness, without other changes. Human mesenchymal stem cells (hMSCs) encapsulated in 3D responded to mechanical stiffness by displaying circular morphology on soft hydrogels (5 kPa) and elongated morphology on stiff hydrogels (30 kPa). Stiff hydrogels facilitated the production and remodeling of nascent extracellular matrix (ECM) and activated mechanotransduction cascade. These changes were driven through intracellular PI3AKT signaling, regulation of epigenetic modifiers and activation of YAP/TAZ signaling. Overall, our study introduces a unique biomaterials platform to understand cell-ECM mechanotransduction in 3D for regenerative medicine as well as disease modelling.
Collapse
Affiliation(s)
- Anna L Kersey
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Daniel Y Cheng
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Kaivalya A Deo
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Christina R Dubell
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Ting-Ching Wang
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Manish K Jaiswal
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Min Hee Kim
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Aparna Murali
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Sarah E Hargett
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Sumana Mallick
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA; Department of Cell Biology and Genetics, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Tanmay P Lele
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Irtisha Singh
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA; Department of Cell Biology and Genetics, School of Medicine, Texas A&M University, Bryan, TX 77807, USA; Interdisciplinary Program in Genetics, Texas A&M University, College Station, TX 77843, USA.
| | - Akhilesh K Gaharwar
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA; Interdisciplinary Program in Genetics, Texas A&M University, College Station, TX 77843, USA; Center for Remote Health Technologies and Systems, Texas A&M University, College Station, TX 77843, USA; Department of Material Science and Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
43
|
Nguyen J, Wang L, Lei W, Hu Y, Gulati N, Chavez-Madero C, Ahn H, Ginsberg HJ, Krawetz R, Brandt M, Betz T, Gilbert PM. Culture substrate stiffness impacts human myoblast contractility-dependent proliferation and nuclear envelope wrinkling. J Cell Sci 2024; 137:jcs261666. [PMID: 38345101 PMCID: PMC11033523 DOI: 10.1242/jcs.261666] [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: 09/27/2023] [Accepted: 02/04/2024] [Indexed: 03/28/2024] Open
Abstract
Understanding how biophysical and biochemical microenvironmental cues together influence the regenerative activities of muscle stem cells and their progeny is crucial in strategizing remedies for pathological dysregulation of these cues in aging and disease. In this study, we investigated the cell-level influences of extracellular matrix (ECM) ligands and culture substrate stiffness on primary human myoblast contractility and proliferation within 16 h of plating and found that tethered fibronectin led to stronger stiffness-dependent responses compared to laminin and collagen. A proteome-wide analysis further uncovered cell metabolism, cytoskeletal and nuclear component regulation distinctions between cells cultured on soft and stiff substrates. Interestingly, we found that softer substrates increased the incidence of myoblasts with a wrinkled nucleus, and that the extent of wrinkling could predict Ki67 (also known as MKI67) expression. Nuclear wrinkling and Ki67 expression could be controlled by pharmacological manipulation of cellular contractility, offering a potential cellular mechanism. These results provide new insights into the regulation of human myoblast stiffness-dependent contractility response by ECM ligands and highlight a link between myoblast contractility and proliferation.
Collapse
Affiliation(s)
- Jo Nguyen
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3E2, Canada
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Lu Wang
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Wen Lei
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Yechen Hu
- Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada
| | - Nitya Gulati
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3E2, Canada
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Carolina Chavez-Madero
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3E2, Canada
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Henry Ahn
- Department of Surgery, University of Toronto, Toronto, ON, M5G 2C4, Canada
- Li Ka Shing Knowledge Institute, Saint Michael's Hospital, Toronto, ON, M5B 1W8, Canada
| | - Howard J. Ginsberg
- Department of Surgery, University of Toronto, Toronto, ON, M5G 2C4, Canada
- Li Ka Shing Knowledge Institute, Saint Michael's Hospital, Toronto, ON, M5B 1W8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Roman Krawetz
- McCaig Institute, University of Calgary, Calgary, AB, T2N 4Z6, Canada
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Matthias Brandt
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University Münster, 48149 Münster, Germany
| | - Timo Betz
- Third Institute of Physics – Biophysics, Georg August University Göttingen, 37077 Göttingen, Germany
| | - Penney M. Gilbert
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3E2, Canada
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada
| |
Collapse
|
44
|
Dong X, Su S, Sun Q, Wang P, Hu Q, Wei Q. Aligned Nanofibers Promote Myoblast Polarization and Myogenesis through Activating Rac-Related Signaling Pathways. ACS Biomater Sci Eng 2024; 10:1712-1721. [PMID: 38422457 DOI: 10.1021/acsbiomaterials.4c00127] [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: 03/02/2024]
Abstract
The extracellular matrix (ECM) plays a crucial role in regulating cellular behaviors and functions. However, the impact of ECM topography on muscle cell adhesion and differentiation remains poorly understood from a mechanosensing perspective. In this study, we fabricated aligned and random electrospun polycaprolactone (PCL) nanofibers to mimic the structural characteristics of ECM. Mechanism investigations revealed that the orientation of nanofibers promoted C2C12 polarization and myogenesis through Rac-related signaling pathways. Conversely, cells cultured on random fibers exhibited spreading behavior mediated by RhoA/ROCK pathways, resulting in enhanced stress fiber formation but reduced capacity for myogenic differentiation. Our findings highlight the critical role of an ECM structure in muscle regeneration and damage repair, providing novel insights into mechanosensing mechanisms underlying muscle injury diseases.
Collapse
Affiliation(s)
- Xiangyu Dong
- Department of Gynecology and Obstetrics, Frontiers Science Center for Disease-related Molecular Network, West China Second University Hospital, Sichuan University, Chengdu 610041, P. R. China
- Department of Nephrology, Kidney Research Institute, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Shan Su
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Qian Sun
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Peng Wang
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Qian Hu
- Department of Gynecology and Obstetrics, Frontiers Science Center for Disease-related Molecular Network, West China Second University Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Qiang Wei
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China
| |
Collapse
|
45
|
Nguyen J, Gilbert PM. Decoding the forces that shape muscle stem cell function. Curr Top Dev Biol 2024; 158:279-306. [PMID: 38670710 DOI: 10.1016/bs.ctdb.2024.02.009] [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: 04/28/2024]
Abstract
Skeletal muscle is a force-producing organ composed of muscle tissues, connective tissues, blood vessels, and nerves, all working in synergy to enable movement and provide support to the body. While robust biomechanical descriptions of skeletal muscle force production at the body or tissue level exist, little is known about force application on microstructures within the muscles, such as cells. Among various cell types, skeletal muscle stem cells reside in the muscle tissue environment and play a crucial role in driving the self-repair process when muscle damage occurs. Early evidence indicates that the fate and function of skeletal muscle stem cells are controlled by both biophysical and biochemical factors in their microenvironments, but much remains to accomplish in quantitatively describing the biophysical muscle stem cell microenvironment. This book chapter aims to review current knowledge on the influence of biophysical stresses and landscape properties on muscle stem cells in heath, aging, and diseases.
Collapse
Affiliation(s)
- Jo Nguyen
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Penney M Gilbert
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Donnelly Centre, University of Toronto, Toronto, ON, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
46
|
Kosmidis Papadimitriou A, Chong SW, Shen Y, Lee OS, Knowles TPJ, Grover LM, Vigolo D. Fabrication of gradient hydrogels using a thermophoretic approach in microfluidics. Biofabrication 2024; 16:025023. [PMID: 38377611 DOI: 10.1088/1758-5090/ad2b05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 02/20/2024] [Indexed: 02/22/2024]
Abstract
The extracellular matrix presents spatially varying physical cues that can influence cell behavior in many processes. Physical gradients within hydrogels that mimic the heterogenous mechanical microenvironment are useful to study the impact of these cues on cellular responses. Therefore, simple and reliable techniques to create such gradient hydrogels are highly desirable. This work demonstrates the fabrication of stiffness gradient Gellan gum (GG) hydrogels by applying a temperature gradient across a microchannel containing hydrogel precursor solution. Thermophoretic migration of components within the precursor solution generates a concentration gradient that mirrors the temperature gradient profile, which translates into mechanical gradients upon crosslinking. Using this technique, GG hydrogels with stiffness gradients ranging from 20 to 90 kPa over 600µm are created, covering the elastic moduli typical of moderately hard to hard tissues. MC3T3 osteoblast cells are then cultured on these gradient substrates, which exhibit preferential migration and enhanced osteogenic potential toward the stiffest region on the gradient. Overall, the thermophoretic approach provides a non-toxic and effective method to create hydrogels with defined mechanical gradients at the micron scale suitable forin vitrobiological studies and potentially tissue engineering applications.
Collapse
Affiliation(s)
| | - Shin Wei Chong
- The University of Sydney, School of Biomedical Engineering, Sydney, NSW 2006, Australia
- The University of Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Yi Shen
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
- The University of Sydney, School of Chemical and Biomolecular Engineering, Sydney, NSW 2006, Australia
- The University of Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Oisin Stefan Lee
- The University of Sydney, School of Biomedical Engineering, Sydney, NSW 2006, Australia
| | - Tuomas P J Knowles
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Liam M Grover
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Daniele Vigolo
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
- The University of Sydney, School of Biomedical Engineering, Sydney, NSW 2006, Australia
- The University of Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
47
|
Cheng X, Xu B, Lei B, Wang S. Opposite Mechanical Preference of Bone/Nerve Regeneration in 3D-Printed Bioelastomeric Scaffolds/Conduits Consistently Correlated with YAP-Mediated Stem Cell Osteo/Neuro-Genesis. Adv Healthc Mater 2024; 13:e2301158. [PMID: 38211963 DOI: 10.1002/adhm.202301158] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 12/31/2023] [Indexed: 01/13/2024]
Abstract
To systematically unveil how substrate stiffness, a critical factor in directing cell fate through mechanotransduction, correlates with tissue regeneration, novel biodegradable and photo-curable poly(trimethylene carbonate) fumarates (PTMCFs) for fabricating elastomeric 2D substrates and 3D bone scaffolds/nerve conduits, are presented. These substrates and structures with adjustable stiffness serve as a unique platform to evaluate how this mechanical cue affects the fate of human umbilical cord mesenchymal stem cells (hMSCs) and hard/soft tissue regeneration in rat femur bone defect and sciatic nerve transection models; whilst, decoupling from topographical and chemical cues. In addition to a positive relationship between substrate stiffness (tensile modulus: 90-990 kPa) and hMSC adhesion, spreading, and proliferation mediated through Yes-associated protein (YAP), opposite mechanical preference is revealed in the osteogenesis and neurogenesis of hMSCs as they are significantly enhanced on the stiff and compliant substrates, respectively. In vivo tissue regeneration demonstrates the same trend: bone regeneration prefers the stiffer scaffolds; while, nerve regeneration prefers the more compliant conduits. Whole-transcriptome analysis further shows that upregulation of Rho GTPase activity and the downstream genes in the compliant group promote nerve repair, providing critical insight into the design strategies of biomaterials for stem cell regulation and hard/soft tissue regeneration through mechanotransduction.
Collapse
Affiliation(s)
- Xiaopeng Cheng
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510006, China
| | - Bowen Xu
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510006, China
| | - Bingxi Lei
- Department of Neurosurgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510006, China
| | - Shanfeng Wang
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510006, China
| |
Collapse
|
48
|
Song Y, Soto J, Wong SY, Wu Y, Hoffman T, Akhtar N, Norris S, Chu J, Park H, Kelkhoff DO, Ang CE, Wernig M, Kasko A, Downing TL, Poo MM, Li S. Biphasic regulation of epigenetic state by matrix stiffness during cell reprogramming. SCIENCE ADVANCES 2024; 10:eadk0639. [PMID: 38354231 PMCID: PMC10866547 DOI: 10.1126/sciadv.adk0639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024]
Abstract
We investigate how matrix stiffness regulates chromatin reorganization and cell reprogramming and find that matrix stiffness acts as a biphasic regulator of epigenetic state and fibroblast-to-neuron conversion efficiency, maximized at an intermediate stiffness of 20 kPa. ATAC sequencing analysis shows the same trend of chromatin accessibility to neuronal genes at these stiffness levels. Concurrently, we observe peak levels of histone acetylation and histone acetyltransferase (HAT) activity in the nucleus on 20 kPa matrices, and inhibiting HAT activity abolishes matrix stiffness effects. G-actin and cofilin, the cotransporters shuttling HAT into the nucleus, rises with decreasing matrix stiffness; however, reduced importin-9 on soft matrices limits nuclear transport. These two factors result in a biphasic regulation of HAT transport into nucleus, which is directly demonstrated on matrices with dynamically tunable stiffness. Our findings unravel a mechanism of the mechano-epigenetic regulation that is valuable for cell engineering in disease modeling and regenerative medicine applications.
Collapse
Affiliation(s)
- Yang Song
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jennifer Soto
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Sze Yue Wong
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Yifan Wu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Tyler Hoffman
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Navied Akhtar
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92617, USA
| | - Sam Norris
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Julia Chu
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Hyungju Park
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Structure and Function of Neural Network, Korea Brain Research Institute (KBRI), Daegu 41068, South Korea
| | - Douglas O. Kelkhoff
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Cheen Euong Ang
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Pathology and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Marius Wernig
- Department of Pathology and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Andrea Kasko
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Timothy L. Downing
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92617, USA
| | - Mu-ming Poo
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Song Li
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
49
|
Li S, Zhang S, Dong S, Zhao M, Zhang W, Zhang C, Wu Z. Stiffness and BMP-2 Mimetic Peptide Jointly Regulate the Osteogenic Differentiation of Rat Bone Marrow Stromal Cells in a Gelatin Cryogel. Biomacromolecules 2024; 25:890-902. [PMID: 38180887 DOI: 10.1021/acs.biomac.3c01045] [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: 01/07/2024]
Abstract
Both biochemical and mechanical cues could regulate the function of stem cells, but the interaction mechanism of their signaling pathway remains unclear, especially in the three-dimensional (3D) culture mode. Higher matrix stiffness promotes osteogenic differentiation of stem cells, and bone morphogenic protein-2 (BMP-2) has been clinically applied to promote bone regeneration. Here, the crosstalk of extracellular mechanical signals on BMP-2 signaling was investigated in rat bone marrow stromal cells (rMSCs) cultured inside cryogels with interconnective pores. Stiff cryogel independently promoted osteogenic differentiation and enhanced the autocrine secretion of BMP-2, thus stimulating increased phosphorylation levels of the Smad1/5/8 complex. BMP-2 mimetic peptide (BMMP) and high cryogel stiffness jointly guided the osteogenic differentiation of rMSCs. Inhibition of rho-associated kinase (ROCK) by Y-27632 or inhibition of nonmuscle myosin II (NM II) by blebbistatin showed that osteogenesis induction by BMP-2 signaling, as well as autocrine secretion of BMP-2 and phosphorylation of the Smad complex, requires the involvement of cytoskeletal tension and ROCK pathway signaling. An interconnective microporous cryogel scaffold promoted rMSC osteogenic differentiation by combining matrix stiffness and BMMP, and it accelerated critical cranial defect repair in the rat model.
Collapse
Affiliation(s)
- Sijing Li
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
- Logistics Department, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Shixiong Zhang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Shuao Dong
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Mengen Zhao
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
- Shenzhen Institute for Drug Control, Shenzhen Testing Center of Medical Devices, Shenzhen, Guangdong 518057, China
| | - Wei Zhang
- Department of Outpatient, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Chao Zhang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Zhaoying Wu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| |
Collapse
|
50
|
Mahmoudi N, Mohamed E, Dehnavi SS, Aguilar LMC, Harvey AR, Parish CL, Williams RJ, Nisbet DR. Calming the Nerves via the Immune Instructive Physiochemical Properties of Self-Assembling Peptide Hydrogels. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303707. [PMID: 38030559 PMCID: PMC10837390 DOI: 10.1002/advs.202303707] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 10/22/2023] [Indexed: 12/01/2023]
Abstract
Current therapies for the devastating damage caused by traumatic brain injuries (TBI) are limited. This is in part due to poor drug efficacy to modulate neuroinflammation, angiogenesis and/or promoting neuroprotection and is the combined result of challenges in getting drugs across the blood brain barrier, in a targeted approach. The negative impact of the injured extracellular matrix (ECM) has been identified as a factor in restricting post-injury plasticity of residual neurons and is shown to reduce the functional integration of grafted cells. Therefore, new strategies are needed to manipulate the extracellular environment at the subacute phase to enhance brain regeneration. In this review, potential strategies are to be discussed for the treatment of TBI by using self-assembling peptide (SAP) hydrogels, fabricated via the rational design of supramolecular peptide scaffolds, as an artificial ECM which under the appropriate conditions yields a supramolecular hydrogel. Sequence selection of the peptides allows the tuning of these hydrogels' physical and biochemical properties such as charge, hydrophobicity, cell adhesiveness, stiffness, factor presentation, degradation profile and responsiveness to (external) stimuli. This review aims to facilitate the development of more intelligent biomaterials in the future to satisfy the parameters, requirements, and opportunities for the effective treatment of TBI.
Collapse
Affiliation(s)
- Negar Mahmoudi
- Laboratory of Advanced Biomaterialsthe John Curtin School of Medical ResearchAustralian National UniversityCanberraACT2601Australia
- ANU College of Engineering & Computer ScienceAustralian National UniversityCanberraACT2601Australia
- The Graeme Clark InstituteThe University of MelbourneMelbourneVIC3010Australia
- Department of Biomedical EngineeringFaculty of Engineering and Information TechnologyThe University of MelbourneMelbourneVIC3010Australia
| | - Elmira Mohamed
- Laboratory of Advanced Biomaterialsthe John Curtin School of Medical ResearchAustralian National UniversityCanberraACT2601Australia
| | - Shiva Soltani Dehnavi
- Laboratory of Advanced Biomaterialsthe John Curtin School of Medical ResearchAustralian National UniversityCanberraACT2601Australia
- ANU College of Engineering & Computer ScienceAustralian National UniversityCanberraACT2601Australia
| | - Lilith M. Caballero Aguilar
- Laboratory of Advanced Biomaterialsthe John Curtin School of Medical ResearchAustralian National UniversityCanberraACT2601Australia
- The Graeme Clark InstituteThe University of MelbourneMelbourneVIC3010Australia
- Department of Biomedical EngineeringFaculty of Engineering and Information TechnologyThe University of MelbourneMelbourneVIC3010Australia
| | - Alan R. Harvey
- School of Human SciencesThe University of Western Australiaand Perron Institute for Neurological and Translational SciencePerthWA6009Australia
| | - Clare L. Parish
- The Florey Institute of Neuroscience and Mental HealthThe University of MelbourneParkvilleMelbourneVIC3010Australia
| | | | - David R. Nisbet
- Laboratory of Advanced Biomaterialsthe John Curtin School of Medical ResearchAustralian National UniversityCanberraACT2601Australia
- The Graeme Clark InstituteThe University of MelbourneMelbourneVIC3010Australia
- Department of Biomedical EngineeringFaculty of Engineering and Information TechnologyThe University of MelbourneMelbourneVIC3010Australia
- Melbourne Medical SchoolFaculty of MedicineDentistry and Health ScienceThe University of MelbourneMelbourneVIC3010Australia
| |
Collapse
|