1
|
Cen C, Wang C, Zhang Y, Hu C, Tang L, Liu C, Cao Y, Wang T, Peng W. Osteoimmunomodulation unveiled: Enhancing bone regeneration with 3D-printed PLLA/β-TCP/CS scaffolds. Colloids Surf B Biointerfaces 2025; 252:114674. [PMID: 40186926 DOI: 10.1016/j.colsurfb.2025.114674] [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: 01/20/2025] [Revised: 03/24/2025] [Accepted: 03/29/2025] [Indexed: 04/07/2025]
Abstract
Bone tissue engineering shows great potential for repairing large segmental bone defects; however, the immune response to biological scaffolds remains a critical factor influencing bone regeneration. Despite this, there is a paucity of studies investigating the effects of biomaterial components and their degradation products on macrophage polarization and the subsequent process of bone regeneration. This study investigates the role of macrophages in osteogenesis and angiogenesis induced by PLLA(Poly-L-Lactic Acid)/β-TCP(β-Tricalcium Phosphate)/CS(Calcium Sulfate) bone scaffolds in vitro and in vivo. Various PLLA/β-TCP/CS scaffolds were fabricated via 3D printing and characterized for their physicochemical properties. The effect of P/T15/S15 (PLLA/β-TCP/CS scaffold containing 15 % β-TCP and 15 % CS) on macrophage polarization and the secretion of VEGF and BMP-2 was assessed in vitro. Additionally, the conditioned medium derived from macrophages stimulated with P/T15/S15 was evaluated for its effects on the migration and osteogenic differentiation of bone marrow-derived stem cells (BMSCs), as well as the angiogenic potential of human umbilical vein endothelial cells (HUVECs). In vivo, the relationship between macrophage polarization and osteogenesis was examined in a rabbit tibia segmental defect model. The results demonstrated that the 3D-printed P/T15/S15 scaffold exhibited favorable physicochemical properties and compatibility with BMSCs and RAW264.7 macrophages, while not disrupting BMSC apoptosis. P/T15/S15 promoted polarization of M0 macrophages towards the M2 phenotype, resulting in an increased secretion of the anti-inflammatory cytokine IL-10, as well as growth factors such as VEGF and BMP-2. However, it did not suppress TNF-α levels. Additional, the conditioned medium derived from P/T15/S15-stimulated macrophages significantly enhanced the osteogenesis of BMSCs, their migration, and the angiogenesis of HUVECs. In the rabbit model, P/T15/S15 facilitated bone regeneration by promoting macrophage polarization towards the M2 phenotype and reducing inflammation. This study highlights that P/T15/S15 regulates macrophage polarization, enhances osteogenesis and angiogenesis, and positions itself as a promising candidate for bone tissue engineering through osteoimmunomodulation.
Collapse
Affiliation(s)
- Chaode Cen
- Department of Orthopedics, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang 550014, China; School of Clinical Medicine, Guizhou Medical University, Guiyang 561113, China
| | - Chuan Wang
- Department of Emergency Surgery, The Affliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Yong Zhang
- Department of gynaecology and obstetrics, The First People'sHospital of Guiyang, Guiyang 550002, China
| | - Chaoran Hu
- School of Clinical Medicine, Guizhou Medical University, Guiyang 561113, China
| | - Lingli Tang
- School of Clinical Medicine, Guizhou Medical University, Guiyang 561113, China
| | - Chengwei Liu
- School of Clinical Medicine, Guizhou Medical University, Guiyang 561113, China
| | - Yongfei Cao
- School of Clinical Medicine, Guizhou Medical University, Guiyang 561113, China
| | - Tao Wang
- Department of Emergency Surgery, The Affliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Wuxun Peng
- School of Clinical Medicine, Guizhou Medical University, Guiyang 561113, China; Department of Emergency Surgery, The Affliated Hospital of Guizhou Medical University, Guiyang 550004, China.
| |
Collapse
|
2
|
Yao MX, Zheng JC, Wang HC, Lv HZ, Zhang YF, Zhang YQ, Shi TL, Zhu YZ, Zhang YZ, Wang XM, Chen W. Application of biphasic mineralized collagen/polycaprolactone scaffolds in the repair of large load-bearing bone defects: A study in a sheep model. J Orthop Translat 2025; 52:138-149. [PMID: 40322042 PMCID: PMC12047585 DOI: 10.1016/j.jot.2025.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 03/19/2025] [Accepted: 03/26/2025] [Indexed: 05/08/2025] Open
Abstract
Objective This study aims to evaluate the efficacy of biphasic mineralized collagen/polycaprolactone (bMC/PCL) scaffolds in repairing large load-bearing bone defects, particularly femoral defects, using a sheep model. Methods The bMC/PCL scaffolds were prepared by combining porous mineralized collagen/polycaprolactone (pMC/PCL) with compact mineralized collagen/polycaprolactone (cMC/PCL). The scaffolds were characterized using scanning electron microscopy to observe the microstructure and compression testing to assess mechanical properties. Twenty female sheep were selected to create a 20 mm femoral defect model, divided into a blank group (no material implanted) and an experimental group (bMC/PCL scaffolds implanted), with 10 sheep in each group. Bone healing and lower limb functional recovery were assessed at 1 month, 3 months, and 6 months postoperatively using Lane-Sandhu scores and visual analog scale scores for lameness. Additionally, bone repair progress was analyzed through X-ray, Micro-CT, and histological analyses. Results Compared with the blank group, the bMC/PCL scaffold group showed significant improvement in bone defect repair. At 3 and 6 months postoperatively, X-ray, Micro-CT scans, and histological staining indicated stable scaffold integration and gradual new bone formation. The Lane-Sandhu scores in the experimental group were 3.60 ± 0.548 and 4.00 ± 0.707 at 3 and 6 months, respectively, whereas the blank group experienced plate/screw breakage leading to fixation failure, with scores of 1, indicating better bone healing in the experimental group. The lameness scores in the experimental group were 2.71 ± 0.97 and 1.48 ± 0.86 at 3 and 6 months, respectively, significantly lower than those in the blank group (p < 0.0001 and p = 0.0002). Micro-CT analysis showed that bone volume to tissue volume ratio increased from 28.07 ± 9.22 % to 62.02 ± 11.82 %, bone mineral density increased from 0.392 ± 0.032 g/cm3 to 0.583 ± 0.125 g/cm3, trabecular thickness increased from 0.690 ± 0.224 mm to 1.049 ± 0.089 mm, and trabecular separation decreased from 2.766 ± 1.183 mm to 0.501 ± 0.268 mm at 3 and 6 months postoperatively. Conclusion This study evaluated the efficacy of bMC/PCL scaffolds in repairing large load-bearing bone defects. The bMC/PCL scaffolds demonstrated good bioactivity and mechanical properties, indicating promising clinical application prospects. Future studies should further verify the safety and efficacy of these scaffolds in a wider range of animal models to support their clinical application. Significance statement The bMC/PCL scaffolds offer a promising solution for large femoral bone defects, with potential for clinical use in orthopedic and trauma surgeries. The translational potential of this article The application of bMC/PCL scaffolds in clinical practice is expected to significantly advance the treatment of large bone defects, particularly weight-bearing bone defects. The study shows that bMC/PCL scaffolds have a significant impact on the repair of large weight-bearing bone defects and functional recovery, indicating their potential for application in orthopedics and trauma care. Specifically, the material's supportive role in weight-bearing bones offers new possibilities for its use in the repair of weight-bearing bone defects. Furthermore, the performance of bMC/PCL scaffolds in bone healing makes them an ideal candidate material for treating various bone defects, with broad clinical application prospects. Further clinical trials are necessary to confirm their safety and efficacy in human patients.
Collapse
Affiliation(s)
- Meng-Xuan Yao
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People's Republic of China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, 050051, People's Republic of China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Jing-Chuan Zheng
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Hai-Cheng Wang
- Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research (Preparing), Cangzhou Hospital of Integrated Traditional Chinese and Western Medicine in Hebei Province, People's Republic of China
| | - Hong-Zhi Lv
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People's Republic of China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, 050051, People's Republic of China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Yi-Fan Zhang
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People's Republic of China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, 050051, People's Republic of China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Yu-Qin Zhang
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People's Republic of China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, 050051, People's Republic of China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Tai-Long Shi
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People's Republic of China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, 050051, People's Republic of China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Yan-Ze Zhu
- Beijing Allgens Medical Science and Technology Co., Ltd., Beijing, 102629, People's Republic of China
| | - Ying-Ze Zhang
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People's Republic of China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, 050051, People's Republic of China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Xiu-Mei Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Wei Chen
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People's Republic of China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, 050051, People's Republic of China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, 050051, People's Republic of China
| |
Collapse
|
3
|
Brigi C, Aghila Rani K, Selvakumar B, Hamad M, Abou Neel EA, Samsudin A. Decoding biomaterial-associated molecular patterns (BAMPs): influential players in bone graft-related foreign body reactions. PeerJ 2025; 13:e19299. [PMID: 40292103 PMCID: PMC12024449 DOI: 10.7717/peerj.19299] [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: 10/31/2024] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
Bone grafts frequently induce immune-mediated foreign body reactions (FBR), which hinder their clinical performance and result in failure. Understanding biomaterial-associated molecular patterns (BAMPs), including physicochemical properties of biomaterial, adsorbed serum proteins, and danger signals, is crucial for improving bone graft outcomes. Recent studies have investigated the role of BAMPs in the induction and maintenance of FBR, thereby advancing the understanding of FBR kinetics, triggers, stages, and key contributors. This review outlines the stages of FBR, the components of BAMPs, and their roles in immune activation. It also discusses various bone grafting biomaterials, their physicochemical properties influencing protein adsorption and macrophage modulation, and the key mechanisms of protein adsorption on biomaterial surfaces. Recent advancements in surface modifications and immunomodulatory strategies to mitigate FBR are also discussed. Furthermore, the authors look forward to future studies that will focus on a comprehensive proteomic analysis of adsorbed serum proteins, a crucial component of BAMPs, to identify proteins that promote or limit inflammation. This understanding could facilitate the design of biomaterials that selectively adsorb beneficial proteins, thereby reducing the risk of FBR and enhancing bone regeneration.
Collapse
Affiliation(s)
- Carel Brigi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
| | - K.G. Aghila Rani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
| | - Balachandar Selvakumar
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
| | - Mawieh Hamad
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
- Department of Medical Laboratory Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Ensanya Ali Abou Neel
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
- Department of Preventive and Restorative Dentistry, College of Dental Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - A.R. Samsudin
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
- Oral and Craniofacial Health Sciences Department, College of Dental Medicine, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
4
|
Jiang H, Sun X, Liu J, Fang L, Liang Y, Zhou J, Wu Y, Lin Z. Decellularized Extracellular Matrix Scaffold Loaded with Regulatory T Cell-Conditioned Medium Induces M2 Macrophage Polarization. Biomater Res 2025; 29:0196. [PMID: 40255505 PMCID: PMC12006722 DOI: 10.34133/bmr.0196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/27/2025] [Accepted: 03/29/2025] [Indexed: 04/22/2025] Open
Abstract
Biomaterials often induce local inflammatory responses following implantation. Scaffolds that cause continuous M1 polarization typically hinder tissue healing and regeneration. Regulating the transformation of macrophages to the M2 phenotype in the inflammatory environment is crucial. We propose that regulatory T cell-conditioned medium (Treg CM) effectively promotes M2 polarization of macrophages induced by decellularized extracellular matrix (dECM) materials in inflammatory environments. In vitro results showed that in the presence of dECM, Treg CM induces the polarization of RAW264.7 macrophages to M2 and inhibits M1 macrophage polarization under inflammatory conditions (lipopolysaccharide + IFN-γ). Additionally, dECM promotes the polarization of bone marrow-derived macrophages (BMDMs) to M2, while Treg CM further promotes M2 polarization and inhibits M1 polarization in an inflammatory environment. These findings were confirmed by transcriptome sequencing. Treg CM inhibited IκB kinase/NF-κB signaling and cellular responses to oxidative stress. In vivo subcutaneous transplantation showed an increase in M2 macrophages, a decrease in M1 macrophages, and an increased M2/M1 ratio in dECM materials loaded with Treg CM. These results suggest that Treg CM can create a pro-M2 polarized microenvironment for dECM, guiding immune responses toward favorable tissue regeneration. Ultimately, this research highlights the potential of Treg CM as a therapeutic approach to modulate the immune response and improve the efficacy of regenerative biomaterials.
Collapse
Affiliation(s)
- Hongjing Jiang
- School of Medicine, South China University of Technology, 510006 Guangzhou, Guangdong, China
- Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences),
Southern Medical University, 510080 Guangzhou, Guangdong, China
| | - Xuheng Sun
- Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences),
Southern Medical University, 510080 Guangzhou, Guangdong, China
| | - Jiang Liu
- Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences),
Southern Medical University, 510080 Guangzhou, Guangdong, China
| | - Lijun Fang
- School of Medicine, South China University of Technology, 510006 Guangzhou, Guangdong, China
| | - Yuanfeng Liang
- Department of Geriatrics,
Guangdong Provincial Geriatrics Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510006 Guangzhou, Guangdong, China
| | - Jiahui Zhou
- Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences),
Southern Medical University, 510080 Guangzhou, Guangdong, China
| | - Yueheng Wu
- Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences),
Southern Medical University, 510080 Guangzhou, Guangdong, China
- Ji Hua Institute of Biomedical Engineering Technology, Ji Hua Laboratory, 528200 Foshan, Guangdong, China
| | - Zhanyi Lin
- School of Medicine, South China University of Technology, 510006 Guangzhou, Guangdong, China
- Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences),
Southern Medical University, 510080 Guangzhou, Guangdong, China
- Ji Hua Institute of Biomedical Engineering Technology, Ji Hua Laboratory, 528200 Foshan, Guangdong, China
| |
Collapse
|
5
|
Bright R, Visalakshan RM, Simon J, Rokstad AM, Ghazaryan A, Morsbach S, Hayles A, Mailänder V, Landfester K, Vasilev K. Manipulation of Serum Protein Adsorption by Nanoengineered Biomaterials Influences Subsequent Immune Responses. ACS Biomater Sci Eng 2024; 10:6230-6240. [PMID: 39213601 DOI: 10.1021/acsbiomaterials.4c01103] [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: 09/04/2024]
Abstract
The adsorption of serum proteins on biomaterial surfaces is a critical determinant for the outcome of medical procedures and therapies, which involve inserting materials and devices into the body. In this study, we aimed to understand how surface topography at the nanoscale influences the composition of the protein corona that forms on the (bio)material surface when placed in contact with serum proteins. To achieve that, we developed nanoengineered model surfaces with finely tuned topography of 16, 40, and 70 nm, overcoated with methyl oxazoline to ensure uniform outermost chemistry across all surfaces. Our findings revealed that within the studied height range, surface nanotopography had no major influence on the overall quantity of adsorbed proteins. However, significant alterations were observed in the composition of the adsorbed protein corona. For instance, clusterin adsorption decreased on all the nanotopography-modified surfaces. Conversely, there was a notable increase in the adsorption of ApoB and IgG gamma on the 70 nm nanotopography. In comparison, the adsorption of albumin was greater on surfaces that had a topography scale of 40 nm. Analysis of the gene enrichment data revealed a reduction in protein adsorption across all immune response-related biological pathways on nanotopography-modified surfaces. This reduction became more pronounced for larger surface nanoprotrusions. Macrophages were used as representative immune cells to assess the influence of the protein corona composition on inflammatory outcomes. Gene expression analysis demonstrated reduced inflammatory responses on the nanotopographically modified surface, a trend further corroborated by cytokine analysis. These findings underscore the potential of precisely engineered nanotopography-coated surfaces for augmenting biomaterial functionality.
Collapse
Affiliation(s)
- Richard Bright
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Rahul M Visalakshan
- Academic Unit of STEM, University of South Australia, Mawson Lakes, Adelaide, South Australia 5095, Australia
| | - Johanna Simon
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Anne Mari Rokstad
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, 7034 Trondheim, Norway
| | - Arthur Ghazaryan
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Svenja Morsbach
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Andrew Hayles
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Volker Mailänder
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
- Dermatology Department, University Medicine Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Katharina Landfester
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Krasimir Vasilev
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| |
Collapse
|
6
|
Hu D, Li T, Bian H, Liu H, Wang P, Wang Y, Sun J. Silk films with distinct surface topography modulate plasma membrane curvature to polarize macrophages. Mater Today Bio 2024; 28:101193. [PMID: 39221204 PMCID: PMC11364906 DOI: 10.1016/j.mtbio.2024.101193] [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: 03/26/2024] [Revised: 08/03/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
The physical properties of a biomaterial play a vital role in modulating macrophage polarization. However, discerning the specific effects of individual parameters can be intricate due to their interdependencies, limiting the mechanism underlying a specific parameter on the polarization of macrophages. Here, we engineered silk fibroin (SF) films with tunable surface roughness while maintaining similar physical properties by combining casting and salting out techniques. We demonstrate that increased surface roughness in SF films promotes M2-like macrophage polarization, characterized by enhanced secretion of anti-inflammatory cytokines. Transcriptomic analysis unveils the modulation of genes associated with extracellular matrix-cell interactions, highlighting the role of surface topography in regulating cellular processes. Mechanistically, we show that surface roughness induces macrophage membrane curvature, facilitating integrin αv endocytosis and thereby inhibiting the integrin-NF-kB signaling pathway. In vivo implantation assays corroborate that rough SF films substantially mitigate early inflammatory responses. This work establishes a direct link between surface roughness and intracellular signaling in macrophages, adding to our understanding of the biomaterial surface effect at the material-cell interface and bringing insights into material design.
Collapse
Affiliation(s)
- Doudou Hu
- Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, 999078, China
| | - Tiandong Li
- Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, 999078, China
| | - Haixu Bian
- Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Haiyu Liu
- Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Pengwei Wang
- Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Yeyuan Wang
- Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Jingchen Sun
- Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| |
Collapse
|
7
|
Wang Y, Liu M, Zhang W, Liu H, Jin F, Mao S, Han C, Wang X. Mechanical strategies to promote vascularization for tissue engineering and regenerative medicine. BURNS & TRAUMA 2024; 12:tkae039. [PMID: 39350780 PMCID: PMC11441985 DOI: 10.1093/burnst/tkae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/30/2024] [Accepted: 06/11/2024] [Indexed: 10/04/2024]
Abstract
Vascularization is a major challenge in the field of tissue engineering and regenerative medicine. Mechanical factors have been demonstrated to play a fundamental role in vasculogenesis and angiogenesis and can affect the architecture of the generated vascular network. Through the regulation of mechanical factors in engineered tissues, various mechanical strategies can be used to optimize the preformed vascular network and promote its rapid integration with host vessels. Optimization of the mechanical properties of scaffolds, including controlling scaffold stiffness, increasing surface roughness and anisotropic structure, and designing interconnected, hierarchical pore structures, is beneficial for the in vitro formation of vascular networks and the ingrowth of host blood vessels. The incorporation of hollow channels into scaffolds promotes the formation of patterned vascular networks. Dynamic stretching and perfusion can facilitate the formation and maturation of preformed vascular networks in vitro. Several indirect mechanical strategies provide sustained mechanical stimulation to engineered tissues in vivo, which further promotes the vascularization of implants within the body. Additionally, stiffness gradients, anisotropic substrates and hollow channels in scaffolds, as well as external cyclic stretch, boundary constraints and dynamic flow culture, can effectively regulate the alignment of vascular networks, thereby promoting better integration of prevascularized engineered tissues with host blood vessels. This review summarizes the influence and contribution of both scaffold-based and external stimulus-based mechanical strategies for vascularization in tissue engineering and elucidates the underlying mechanisms involved.
Collapse
Affiliation(s)
- Yiran Wang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Meixuan Liu
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Wei Zhang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Huan Liu
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Fang Jin
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Shulei Mao
- Department of Burns and Plastic Surgery, Quhua Hospital of Zhejiang, 62 Wenchang Road, Quhua, Quzhou 324004, China
| | - Chunmao Han
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Xingang Wang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| |
Collapse
|
8
|
Wang M, Hong Y, Fu X, Sun X. Advances and applications of biomimetic biomaterials for endogenous skin regeneration. Bioact Mater 2024; 39:492-520. [PMID: 38883311 PMCID: PMC11179177 DOI: 10.1016/j.bioactmat.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 06/18/2024] Open
Abstract
Endogenous regeneration is becoming an increasingly important strategy for wound healing as it facilitates skin's own regenerative potential for self-healing, thereby avoiding the risks of immune rejection and exogenous infection. However, currently applied biomaterials for inducing endogenous skin regeneration are simplistic in their structure and function, lacking the ability to accurately mimic the intricate tissue structure and regulate the disordered microenvironment. Novel biomimetic biomaterials with precise structure, chemical composition, and biophysical properties offer a promising avenue for achieving perfect endogenous skin regeneration. Here, we outline the recent advances in biomimetic materials induced endogenous skin regeneration from the aspects of structural and functional mimicry, physiological process regulation, and biophysical property design. Furthermore, novel techniques including in situ reprograming, flexible electronic skin, artificial intelligence, single-cell sequencing, and spatial transcriptomics, which have potential to contribute to the development of biomimetic biomaterials are highlighted. Finally, the prospects and challenges of further research and application of biomimetic biomaterials are discussed. This review provides reference to address the clinical problems of rapid and high-quality skin regeneration.
Collapse
Affiliation(s)
- Mengyang Wang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100089, PR China
| | - Yiyue Hong
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100089, PR China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100089, PR China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| | - Xiaoyan Sun
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100089, PR China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| |
Collapse
|
9
|
Díez-Tercero L, Bosch-Rué È, Bosch BM, Rojas-Márquez R, Caballé-Serrano J, Delgado LM, Pérez RA. Engineering a microparticle-loaded rough membrane for guided bone regeneration modulating osteoblast response without inducing inflammation. Colloids Surf B Biointerfaces 2024; 241:113994. [PMID: 38850744 DOI: 10.1016/j.colsurfb.2024.113994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/14/2024] [Accepted: 05/26/2024] [Indexed: 06/10/2024]
Abstract
Guided bone regeneration (GBR) is a widely used procedure that prevents the fast in-growth of soft tissues into bone defect. Among the different types of membranes, the use of collagen membranes is the gold standard. However, these membranes are implanted in tissue location where a severe acute inflammation will occur and can be negatively affected. The aim of this study was to develop a collagen-based membrane for GBR that incorporated alginate-hydroxyapatite microparticles. Membranes were manufactured using collagen type I and gelatin and alginate-hydroxyapatite microparticles. Membranes were assessed in terms of topography by scanning electron microscopy and confocal microscopy; stability by swelling after an overnight incubation in saline and enzymatic degradation against collagenase and mechanical properties by tensile tests. Furthermore, the biological response was assessed with SaOs-2 cells and THP-1 macrophages to determine alkaline phosphatase activity and inflammatory cytokine release. Our results showed that the incorporation of different percentages of these microparticles could induce changes in the surface topography. When the biological response was analyzed, either membranes were not cytotoxic to THP-1 macrophages or to SaOs-2 cells and they did not induce the release of pro-inflammatory cytokines. However, the different surface topographies did not induce changes in the macrophage morphology and the release of pro- and anti-inflammatory cytokines, suggesting that the effect of surface roughness on macrophage behavior could be dependent on other factors such as substrate stiffness and composition. Collagen-gelatin membranes with embedded alginate-hydroxyapatite microparticles increased ALP activity, suggesting a positive effect of them on bone regeneration, remaining unaffected the release of pro- and anti-inflammatory cytokines.
Collapse
Affiliation(s)
- Leire Díez-Tercero
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain; Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain
| | - Èlia Bosch-Rué
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain; Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain
| | - Begoña M Bosch
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain; Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain
| | - Raquel Rojas-Márquez
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain; Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain
| | - Jordi Caballé-Serrano
- Department of Oral and Maxillofacial Surgery, Universitat Internacional de Catalunya, Barcelona, Spain; Department of Periodontology, School of Dental Medicine - University of Bern, Bern, Switzerland
| | - Luis M Delgado
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain; Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain.
| | - Román A Pérez
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain; Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain.
| |
Collapse
|
10
|
Pereira Vasconcelos D, Leite Pereira C, Couto M, Neto E, Ribeiro B, Albuquerque F, Freitas A, Alves CJ, Klinkenberg G, McDonagh BH, Schmid RB, Seitz AM, de Roy L, Ignatius A, Haaparanta A, Muhonen V, Sarmento B, Lamghari M. Nanoenabled Immunomodulatory Scaffolds for Cartilage Tissue Engineering. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202400627] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Indexed: 01/06/2025]
Abstract
AbstractArticular cartilage regeneration is a challenge in tissue engineering. Although diverse materials have been developed for this purpose, cartilage regeneration remains suboptimal. The integration of nanomaterials into 3D network materials holds great potential in the improvement of key mechanical properties, particularly important for osteochondral replacement scaffolds and even to function as carriers for disease‐modifying drugs or other regulatory signals. In this study, a simple yet effective cell‐free nanoenabled Col‐PLA scaffold specially designed to enhance cartilage regeneration and modulate inflammatory response is proposed, by incorporating poly(lactic‐co‐glycolic acid) (PLGA) ibuprofen nanoparticles (NPs) into a collagen/polylactide (Col‐PLA) matrix. The developed nanoenabled scaffold successfully decreases IL‐1β release and leads to primary human chondrocytes survival, ultimately restoring extracellular matrix (ECM) production under inflammatory conditions. The nanoenabled Col‐PLA scaffolds secretome effectively decreases macrophage invasion in vitro, as well as neutrophil infiltration and inflammatory mediators’, namely the complement component C5/C5a, C‐reactive protein, IL‐1β, MMP9, CCL20, and CXCL1/KC production in vivo in a rodent air‐pouch model. Overall, the established nanoenabled scaffold has the potential to support chondrogenesis as well as modulate inflammatory response, overcoming the limitations of traditional tissue engineering strategies.
Collapse
Affiliation(s)
- Daniela Pereira Vasconcelos
- i3S ‐ Instituto de Inovação e Investigação em Saúde INEB ‐ Instituto Nacional de Engenharia Biomédica Universidade do Porto Rua Alfredo Allen, 208 Porto 4200‐135 Portugal
| | - Catarina Leite Pereira
- i3S ‐ Instituto de Inovação e Investigação em Saúde INEB ‐ Instituto Nacional de Engenharia Biomédica Universidade do Porto Rua Alfredo Allen, 208 Porto 4200‐135 Portugal
| | - Marina Couto
- i3S ‐ Instituto de Inovação e Investigação em Saúde INEB ‐ Instituto Nacional de Engenharia Biomédica Universidade do Porto Rua Alfredo Allen, 208 Porto 4200‐135 Portugal
| | - Estrela Neto
- i3S ‐ Instituto de Inovação e Investigação em Saúde INEB ‐ Instituto Nacional de Engenharia Biomédica Universidade do Porto Rua Alfredo Allen, 208 Porto 4200‐135 Portugal
| | - Beatriz Ribeiro
- i3S ‐ Instituto de Inovação e Investigação em Saúde INEB ‐ Instituto Nacional de Engenharia Biomédica Universidade do Porto Rua Alfredo Allen, 208 Porto 4200‐135 Portugal
| | - Filipe Albuquerque
- i3S ‐ Instituto de Inovação e Investigação em Saúde INEB ‐ Instituto Nacional de Engenharia Biomédica Universidade do Porto Rua Alfredo Allen, 208 Porto 4200‐135 Portugal
- School of Medicine and Biomedical Sciences (ICBAS‐UP) Faculdade de Engenharia (FEUP) Universidade do Porto Porto 4050‐313 Portugal
| | - Alexandra Freitas
- i3S ‐ Instituto de Inovação e Investigação em Saúde INEB ‐ Instituto Nacional de Engenharia Biomédica Universidade do Porto Rua Alfredo Allen, 208 Porto 4200‐135 Portugal
- School of Medicine and Biomedical Sciences (ICBAS‐UP) Faculdade de Engenharia (FEUP) Universidade do Porto Porto 4050‐313 Portugal
| | - Cecília J. Alves
- i3S ‐ Instituto de Inovação e Investigação em Saúde INEB ‐ Instituto Nacional de Engenharia Biomédica Universidade do Porto Rua Alfredo Allen, 208 Porto 4200‐135 Portugal
| | - Geir Klinkenberg
- SINTEF Industry Department of Biotechnology and Nanomedicine Trondheim 7034 Norway
| | | | | | - Andreas M. Seitz
- Institute of Orthopedic Research and Biomechanics Center for Trauma Research Ulm University Medical Center Ulm 89081 Ulm Germany
| | - Luisa de Roy
- Institute of Orthopedic Research and Biomechanics Center for Trauma Research Ulm University Medical Center Ulm 89081 Ulm Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics Center for Trauma Research Ulm University Medical Center Ulm 89081 Ulm Germany
| | | | - Virpi Muhonen
- Askel Healthcare Ltd Siltasaarenkatu 8‐10 Helsinki 00530 Finland
| | - Bruno Sarmento
- i3S ‐ Instituto de Inovação e Investigação em Saúde INEB ‐ Instituto Nacional de Engenharia Biomédica Universidade do Porto Rua Alfredo Allen, 208 Porto 4200‐135 Portugal
- Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde Gandra 4585‐116 Portugal
| | - Meriem Lamghari
- i3S ‐ Instituto de Inovação e Investigação em Saúde INEB ‐ Instituto Nacional de Engenharia Biomédica Universidade do Porto Rua Alfredo Allen, 208 Porto 4200‐135 Portugal
| |
Collapse
|
11
|
Wu Y, Chen X, Song P, Li R, Zhou Y, Wang Q, Shi J, Qiao W, Dong N. Functional Oxidized Hyaluronic Acid Cross-Linked Decellularized Heart Valves for Improved Immunomodulation, Anti-Calcification, and Recellularization. Adv Healthc Mater 2024; 13:e2303737. [PMID: 38560921 DOI: 10.1002/adhm.202303737] [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: 10/27/2023] [Revised: 03/09/2024] [Indexed: 04/04/2024]
Abstract
Tissue engineering heart valves (TEHVs) are expected to address the limitations of mechanical and bioprosthetic valves used in clinical practice. Decellularized heart valve (DHV) is an important scaffold of TEHVs due to its natural three-dimensional structure and bioactive extracellular matrix, but its mechanical properties and hemocompatibility are impaired. In this study, DHV is cross-linked with three different molecular weights of oxidized hyaluronic acid (OHA) by a Schiff base reaction and presented enhanced stability and hemocompatibility, which could be mediated by the molecular weight of OHA. Notably, DHV cross-linked with middle- and high-molecular-weight OHA could drive the macrophage polarization toward the M2 phenotype in vitro. Moreover, DHV cross-linked with middle-molecular-weight OHA scaffolds are further modified with RGD-PHSRN peptide (RPF-OHA/DHV) to block the residual aldehyde groups of the unreacted OHA. The results show that RPF-OHA/DHV not only exhibits anti-calcification properties, but also facilitates endothelial cell adhesion and proliferation in vitro. Furthermore, RPF-OHA/DHV shows excellent performance under an in vivo hemodynamic environment with favorable recellularization and immune regulation without calcification. The optimistic results demonstrate that OHA with different molecular weights has different cross-linking effects on DHV and that RPF-OHA/DHV scaffold with enhanced immune regulation, anti-calcification, and recellularization properties for clinical transformation.
Collapse
Affiliation(s)
- Yunlong Wu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Xing Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Department of Cardiovascular Surgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, 430071, China
| | - Peng Song
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Rui Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Ying Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Qin Wang
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Weihua Qiao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| |
Collapse
|
12
|
Huo Z, Yang W, Harati J, Nene A, Borghi F, Piazzoni C, Milani P, Guo S, Galluzzi M, Boraschi D. Biomechanics of Macrophages on Disordered Surface Nanotopography. ACS APPLIED MATERIALS & INTERFACES 2024; 16:27164-27176. [PMID: 38750662 DOI: 10.1021/acsami.4c04330] [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: 05/30/2024]
Abstract
Macrophages are involved in every stage of the innate/inflammatory immune responses in the body tissues, including the resolution of the reaction, and they do so in close collaboration with the extracellular matrix (ECM). Simplified substrates with nanotopographical features attempt to mimic the structural properties of the ECM to clarify the functional features of the interaction of the ECM with macrophages. We still have a limited understanding of the macrophage behavior upon interaction with disordered nanotopography, especially with features smaller than 10 nm. Here, we combine atomic force microscopy (AFM), finite element modeling (FEM), and quantitative biochemical approaches in order to understand the mechanotransduction from the nanostructured surface into cellular responses. AFM experiments show a decrease of macrophage stiffness, measured with the Young's modulus, as a biomechanical response to a nanostructured (ns-) ZrOx surface. FEM experiments suggest that ZrOx surfaces with increasing roughness represent weaker mechanical boundary conditions. The mechanical cues from the substrate are transduced into the cell through the formation of integrin-regulated focal adhesions and cytoskeletal reorganization, which, in turn, modulate cell biomechanics by downregulating cell stiffness. Surface nanotopography and consequent biomechanical response impact the overall behavior of macrophages by increasing movement and phagocytic ability without significantly influencing their inflammatory behavior. Our study suggests a strong potential of surface nanotopography for the regulation of macrophage functions, which implies a prospective application relative to coating technology for biomedical devices.
Collapse
Affiliation(s)
- Zixin Huo
- Shenzhen Key Laboratory of Smart Sensing and Intelligent Systems, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenjie Yang
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Javad Harati
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ajinkya Nene
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Francesca Borghi
- CIMaINa and Dipartimento di Fisica "Aldo Pontremoli", Università degli Studi di Milano, Via Celoria 16, 20133 Milan, Italy
| | - Claudio Piazzoni
- CIMaINa and Dipartimento di Fisica "Aldo Pontremoli", Università degli Studi di Milano, Via Celoria 16, 20133 Milan, Italy
| | - Paolo Milani
- CIMaINa and Dipartimento di Fisica "Aldo Pontremoli", Università degli Studi di Milano, Via Celoria 16, 20133 Milan, Italy
| | - Shifeng Guo
- Shenzhen Key Laboratory of Smart Sensing and Intelligent Systems, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Guangdong Provincial Key Lab of Robotics and Intelligent System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Massimiliano Galluzzi
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Diana Boraschi
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Department of Pharmacology, Shenzhen University of Advanced Technology, Shenzhen 518055, China
- China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen 518055, China
- Institute of Biomolecular Chemistry, National Research Council, 80078 Pozzuoli, Italy
- Stazione Zoologica Anton Dohrn, 80122 Napoli, Italy
| |
Collapse
|
13
|
Chen D, Kang Z, Chen H, Fu P. Molecular mechanisms of macrophage immunomodulation mediated by Areca inflorescence polysaccharides based on RNA-seq analysis. Int J Biol Macromol 2024; 263:130076. [PMID: 38354932 DOI: 10.1016/j.ijbiomac.2024.130076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/09/2023] [Accepted: 02/07/2024] [Indexed: 02/16/2024]
Abstract
The elucidation of the immunomodulatory molecular mechanisms of polysaccharides has contributed to their further development and application. In this study, the effect of Areca inflorescence polysaccharide (AFP2a) on macrophage activation was confirmed and the detailed mechanisms were investigated based on a comprehensive transcriptional study and specific inhibitors. The results showed that AFP2a induced macrophage activation (M1 polarization), promoting macrophage proliferation, reactive oxygen species production, nitric oxide and cytokine release, and costimulatory molecule expression. RNA-seq analysis identified 5919 differentially expressed genes (DEGs). For DEGs, GO, KEGG, and Reactome enrichment analyses and PPI networks were conducted, elucidating that AFP2a activated macrophages mainly by triggering the Toll-like receptor cascade and corresponding adapter proteins (TIRAP and TRIF), thereby resulting in downstream NF-κB, TNF, and JAK-STAT signaling pathway expression. The inhibition assay revealed that TLR4 and TLR2 were essential for the recognition of AFP2a. This work provides an in-depth understanding of the immunoregulatory mechanism of AFP2a while offering a molecular basis for AFP2a to serve as a potential natural immunomodulator.
Collapse
Affiliation(s)
- Di Chen
- Hainan University-HSF/LWL Collaborative Innovation Laboratory, School of Food Science and Engineering, Hainan University, Haikou, China; State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou 570228, China
| | - Zonghua Kang
- Hunan Kouweiwang Group Co., Ltd, Hunan 413499, China
| | - Haiming Chen
- Hainan University-HSF/LWL Collaborative Innovation Laboratory, School of Food Science and Engineering, Hainan University, Haikou, China; Huachuang Institute of Areca Research-Hainan, Hainan 570228, China.
| | - Pengcheng Fu
- Hainan University-HSF/LWL Collaborative Innovation Laboratory, School of Food Science and Engineering, Hainan University, Haikou, China; State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou 570228, China.
| |
Collapse
|
14
|
Wang X, Fu X, Luo D, Hou R, Li P, Chen Y, Zhang X, Meng X, Yue Y, Liu J. 3D printed high-precision porous scaffolds prepared by fused deposition modeling induce macrophage polarization to promote bone regeneration. Biomed Mater 2024; 19:035006. [PMID: 38422525 DOI: 10.1088/1748-605x/ad2ed0] [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: 10/31/2023] [Accepted: 02/29/2024] [Indexed: 03/02/2024]
Abstract
Macrophage-mediated bone immune responses significantly influence the repair of bone defects when utilizing tissue-engineered scaffolds. Notably, the scaffolds' physical structure critically impacts macrophage polarization. The optimal pore size for facilitating bone repair remains a topic of debate due to the imprecision of traditional methods in controlling scaffold pore dimensions and spatial architecture. In this investigation, we utilized fused deposition modeling (FDM) technology to fabricate high-precision porous polycaprolactone (PCL) scaffolds, aiming to elucidate the impact of pore size on macrophage polarization. We assessed the scaffolds' mechanical attributes and biocompatibility. Real-time quantitative reverse transcription polymerase chain reaction was used to detect the expression levels of macrophage-related genes, and enzyme linked immunosorbent assay for cytokine secretion levels.In vitroosteogenic capacity was determined through alkaline phosphatase and alizarin red staining. Our findings indicated that macroporous scaffolds enhanced macrophage adhesion and drove their differentiation towards the M2 phenotype. This led to the increased production of anti-inflammatory factors and a reduction in pro-inflammatory agents, highlighting the scaffolds' immunomodulatory capabilities. Moreover, conditioned media from macrophages cultured on these macroporous scaffolds bolstered the osteogenic differentiation of bone marrow mesenchymal stem cells, exhibiting superior osteogenic differentiation potential. Consequently, FDM-fabricated PCL scaffolds, with precision-controlled pore sizes, present promising prospects as superior materials for bone tissue engineering, leveraging the regulation of macrophage polarization.
Collapse
Affiliation(s)
- Xiangyu Wang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, People's Republic of China
| | - Xinyu Fu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, People's Republic of China
| | - Dongmei Luo
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, People's Republic of China
| | - Ruxia Hou
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, People's Republic of China
| | - Peiwen Li
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, People's Republic of China
| | - Yurou Chen
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, People's Republic of China
| | - Xinyao Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, People's Republic of China
| | - Xiangjie Meng
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, People's Republic of China
| | - Yingge Yue
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, People's Republic of China
| | - Junyu Liu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, People's Republic of China
| |
Collapse
|
15
|
Cao L, Zhang Z, Yuan D, Yu M, Min J. Tissue engineering applications of recombinant human collagen: a review of recent progress. Front Bioeng Biotechnol 2024; 12:1358246. [PMID: 38419725 PMCID: PMC10900516 DOI: 10.3389/fbioe.2024.1358246] [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: 12/19/2023] [Accepted: 01/31/2024] [Indexed: 03/02/2024] Open
Abstract
With the rapid development of synthetic biology, recombinant human collagen has emerged as a cutting-edge biological material globally. Its innovative applications in the fields of material science and medicine have opened new horizons in biomedical research. Recombinant human collagen stands out as a highly promising biomaterial, playing a pivotal role in crucial areas such as wound healing, stroma regeneration, and orthopedics. However, realizing its full potential by efficiently delivering it for optimal therapeutic outcomes remains a formidable challenge. This review provides a comprehensive overview of the applications of recombinant human collagen in biomedical systems, focusing on resolving this crucial issue. Additionally, it encompasses the exploration of 3D printing technologies incorporating recombinant collagen to address some urgent clinical challenges in regenerative repair in the future. The primary aim of this review also is to spotlight the advancements in the realm of biomaterials utilizing recombinant collagen, with the intention of fostering additional innovation and making significant contributions to the enhancement of regenerative biomaterials, therapeutic methodologies, and overall patient outcomes.
Collapse
Affiliation(s)
- Lili Cao
- Department of Plastic Surgery, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang, China
| | - Zhongfeng Zhang
- Department of Plastic Surgery, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang, China
| | - Dan Yuan
- Department of Plastic Surgery, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang, China
| | - Meiping Yu
- Department of Plastic Surgery, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang, China
| | - Jie Min
- General Surgery Department, Jiaxing No.1 Hospital, Jiaxing, Zhejiang, China
| |
Collapse
|
16
|
Zhu F, Wang S, Zhu X, Pang C, Cui P, Yang F, Li R, Zhan Q, Xin H. Potential effects of biomaterials on macrophage function and their signalling pathways. Biomater Sci 2023; 11:6977-7002. [PMID: 37695360 DOI: 10.1039/d3bm01213a] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
The use of biomaterials in biomedicine and healthcare has increased in recent years. Macrophages are the primary immune cells that induce inflammation and tissue repair after implantation of biomaterials. Given that macrophages exhibit high heterogeneity and plasticity, the influence of biomaterials on macrophage phenotype should be considered a crucial evaluation criterion during the development of novel biomaterials. This review provides a comprehensive summary of the physicochemical, biological, and dynamic characteristics of biomaterials that drive the regulation of immune responses in macrophages. The mechanisms involved in the interaction between macrophages and biomaterials, including endocytosis, receptors, signalling pathways, integrins, inflammasomes and long non-coding RNAs, are summarised in this review. In addition, research prospects of the interaction between macrophages and biomaterials are discussed. An in-depth understanding of mechanisms underlying the spatiotemporal changes in macrophage phenotype induced by biomaterials and their impact on macrophage polarization can facilitate the identification and development of novel biomaterials with superior performance. These biomaterials may be used for tissue repair and regeneration, vaccine or drug delivery and immunotherapy.
Collapse
Affiliation(s)
- Fujun Zhu
- Department of Burns and Plastic Surgery, the No. 924th Hospital of the Joint Logistic Support Force of the Chinese PLA, Guilin, Guangxi 541002, People's Republic of China.
| | - Shaolian Wang
- Central Sterile Supply Department, the No. 924th Hospital of the Joint Logistic Support Force of the Chinese PLA, Guilin, Guangxi 541002, People's Republic of China
| | - Xianglian Zhu
- Outpatient Department, the No. 924th Hospital of the Joint Logistic Support Force of the Chinese PLA, Guilin, Guangxi 541002, People's Republic of China
| | - Caixiang Pang
- Department of Emergency Medicine, the No. 924th Hospital of the Joint Logistic Support Force of the Chinese PLA, Guilin, Guangxi 541002, People's Republic of China
| | - Pei Cui
- Animal Laboratory, the No. 924th Hospital of the Joint Logistic Support Force of the Chinese PLA, Guilin, Guangxi 541002, People's Republic of China
| | - Fuwang Yang
- Department of Burns and Plastic Surgery, the No. 924th Hospital of the Joint Logistic Support Force of the Chinese PLA, Guilin, Guangxi 541002, People's Republic of China.
| | - Rongsheng Li
- Animal Laboratory, the No. 924th Hospital of the Joint Logistic Support Force of the Chinese PLA, Guilin, Guangxi 541002, People's Republic of China
| | - Qiu Zhan
- Animal Laboratory, the No. 924th Hospital of the Joint Logistic Support Force of the Chinese PLA, Guilin, Guangxi 541002, People's Republic of China
| | - Haiming Xin
- Department of Burns and Plastic Surgery, the No. 924th Hospital of the Joint Logistic Support Force of the Chinese PLA, Guilin, Guangxi 541002, People's Republic of China.
| |
Collapse
|
17
|
Dutta SD, Ganguly K, Patil TV, Randhawa A, Lim KT. Unraveling the potential of 3D bioprinted immunomodulatory materials for regulating macrophage polarization: State-of-the-art in bone and associated tissue regeneration. Bioact Mater 2023; 28:284-310. [PMID: 37303852 PMCID: PMC10248805 DOI: 10.1016/j.bioactmat.2023.05.014] [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: 11/17/2022] [Revised: 04/29/2023] [Accepted: 05/20/2023] [Indexed: 06/13/2023] Open
Abstract
Macrophage-assisted immunomodulation is an alternative strategy in tissue engineering, wherein the interplay between pro-inflammatory and anti-inflammatory macrophage cells and body cells determines the fate of healing or inflammation. Although several reports have demonstrated that tissue regeneration depends on spatial and temporal regulation of the biophysical or biochemical microenvironment of the biomaterial, the underlying molecular mechanism behind immunomodulation is still under consideration for developing immunomodulatory scaffolds. Currently, most fabricated immunomodulatory platforms reported in the literature show regenerative capabilities of a particular tissue, for example, endogenous tissue (e.g., bone, muscle, heart, kidney, and lungs) or exogenous tissue (e.g., skin and eye). In this review, we briefly introduced the necessity of the 3D immunomodulatory scaffolds and nanomaterials, focusing on material properties and their interaction with macrophages for general readers. This review also provides a comprehensive summary of macrophage origin and taxonomy, their diverse functions, and various signal transduction pathways during biomaterial-macrophage interaction, which is particularly helpful for material scientists and clinicians for developing next-generation immunomodulatory scaffolds. From a clinical standpoint, we briefly discussed the role of 3D biomaterial scaffolds and/or nanomaterial composites for macrophage-assisted tissue engineering with a special focus on bone and associated tissues. Finally, a summary with expert opinion is presented to address the challenges and future necessity of 3D bioprinted immunomodulatory materials for tissue engineering.
Collapse
Affiliation(s)
- Sayan Deb Dutta
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Keya Ganguly
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Tejal V. Patil
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Aayushi Randhawa
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| |
Collapse
|
18
|
Zhu X, Wang C, Bai H, Zhang J, Wang Z, Li Z, Zhao X, Wang J, Liu H. Functionalization of biomimetic mineralized collagen for bone tissue engineering. Mater Today Bio 2023; 20:100660. [PMID: 37214545 PMCID: PMC10199226 DOI: 10.1016/j.mtbio.2023.100660] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/18/2023] [Accepted: 05/04/2023] [Indexed: 05/24/2023] Open
Abstract
Mineralized collagen (MC) is the basic unit of bone structure and function and is the main component of the extracellular matrix (ECM) in bone tissue. In the biomimetic method, MC with different nanostructures of neo-bone have been constructed. Among these, extra-fibrous MC has been approved by regulatory agencies and applied in clinical practice to play an active role in bone defect repair. However, in the complex microenvironment of bone defects, such as in blood supply disorders and infections, MC is unable to effectively perform its pro-osteogenic activities and needs to be functionalized to include osteogenesis and the enhancement of angiogenesis, anti-infection, and immunomodulation. This article aimed to discuss the preparation and biological performance of MC with different nanostructures in detail, and summarize its functionalization strategy. Then we describe the recent advances in the osteo-inductive properties and multifunctional coordination of MC. Finally, the latest research progress of functionalized biomimetic MC, along with the development challenges and future trends, are discussed. This paper provides a theoretical basis and advanced design philosophy for bone tissue engineering in different bone microenvironments.
Collapse
Affiliation(s)
- Xiujie Zhu
- Department of Orthopedics, The Second Hospital of Jilin University, 4110 Yatai Street, Changchun, 130041, PR China
| | - Chenyu Wang
- Department of Plastic and Reconstruct Surgery, The First Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, PR China
| | - Haotian Bai
- Department of Orthopedics, The Second Hospital of Jilin University, 4110 Yatai Street, Changchun, 130041, PR China
| | - Jiaxin Zhang
- Department of Orthopedics, The Second Hospital of Jilin University, 4110 Yatai Street, Changchun, 130041, PR China
| | - Zhonghan Wang
- Department of Orthopedics, The Second Hospital of Jilin University, 4110 Yatai Street, Changchun, 130041, PR China
| | - Zuhao Li
- Department of Orthopedics, The Second Hospital of Jilin University, 4110 Yatai Street, Changchun, 130041, PR China
| | - Xin Zhao
- Department of Orthopedics, The Second Hospital of Jilin University, 4110 Yatai Street, Changchun, 130041, PR China
| | - Jincheng Wang
- Department of Orthopedics, The Second Hospital of Jilin University, 4110 Yatai Street, Changchun, 130041, PR China
| | - He Liu
- Department of Orthopedics, The Second Hospital of Jilin University, 4110 Yatai Street, Changchun, 130041, PR China
| |
Collapse
|
19
|
Zhou Z, Zhang J, He X, Chen X, Dong L, Lin J, Wang H, Weng W, Cheng K. Regulation of Macrophage Polarization on Chiral Potential Distribution of CFO/P(VDF-TrFE) Films. ACS Biomater Sci Eng 2023; 9:2524-2533. [PMID: 37092816 DOI: 10.1021/acsbiomaterials.3c00207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Surface potentials of biomaterials have been shown to regulate cell fate commitment. However, the effects of chirality-patterned potential distribution on macrophage polarization are still only beginning to be explored. In this work, we demonstrated that the chirality-patterned potential distribution of CoFe2O4/poly(vinylidene fluoride-trifluoroethylene) (CFO/P(VDF-TrFE)) films could significantly down-regulate the M1 polarization of bone marrow-derived macrophages (BMDMs). Specifically, the dextral-patterned surface potential distribution simultaneously up-regulated the expression of M2-related markers of BMDMs. The results were attributed to the sensitive difference of integrin subunits (α5β1 and αvβ3) to the dextral- and sinistral-patterned surface potential distribution, respectively. The interaction difference between the integrin subunits and surface potential distribution altered the cell adhesion and cytoskeletal structure and thereby the polarization behavior of BMDMs. This work, therefore, emphasizes the importance of chirality of potential distribution on cell behavior and provides a new strategy to regulate the immune response of biomaterials.
Collapse
Affiliation(s)
- Zhiyuan Zhou
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou 310027, China
| | - Jiamin Zhang
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou 310027, China
| | - Xuzhao He
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou 310027, China
| | - Xiaoyi Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Province Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Lingqing Dong
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Province Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Jun Lin
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Huiming Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Province Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Wenjian Weng
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou 310027, China
| | - Kui Cheng
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
20
|
Dal-Fabbro R, Swanson WB, Capalbo LC, Sasaki H, Bottino MC. Next-generation biomaterials for dental pulp tissue immunomodulation. Dent Mater 2023; 39:333-349. [PMID: 36894414 PMCID: PMC11034777 DOI: 10.1016/j.dental.2023.03.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/24/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023]
Abstract
OBJECTIVES The current standard for treating irreversibly damaged dental pulp is root canal therapy, which involves complete removal and debridement of the pulp space and filling with an inert biomaterial. A regenerative approach to treating diseased dental pulp may allow for complete healing of the native tooth structure and enhance the long-term outcome of once-necrotic teeth. The aim of this paper is, therefore, to highlight the current state of dental pulp tissue engineering and immunomodulatory biomaterials properties, identifying exciting opportunities for their synergy in developing next-generation biomaterials-driven technologies. METHODS An overview of the inflammatory process focusing on immune responses of the dental pulp, followed by periapical and periodontal tissue inflammation are elaborated. Then, the most recent advances in treating infection-induced inflammatory oral diseases, focusing on biocompatible materials with immunomodulatory properties are discussed. Of note, we highlight some of the most used modifications in biomaterials' surface, or content/drug incorporation focused on immunomodulation based on an extensive literature search over the last decade. RESULTS We provide the readers with a critical summary of recent advances in immunomodulation related to pulpal, periapical, and periodontal diseases while bringing light to tissue engineering strategies focusing on healing and regenerating multiple tissue types. SIGNIFICANCE Significant advances have been made in developing biomaterials that take advantage of the host's immune system to guide a specific regenerative outcome. Biomaterials that efficiently and predictably modulate cells in the dental pulp complex hold significant clinical promise for improving standards of care compared to endodontic root canal therapy.
Collapse
Affiliation(s)
- Renan Dal-Fabbro
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - W Benton Swanson
- Department of Biologic and Materials Science, Division of Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - Leticia C Capalbo
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Hajime Sasaki
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - Marco C Bottino
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
21
|
Patel DK, Ganguly K, Dutta SD, Patil TV, Lim KT. Cellulose nanocrystals vs. cellulose nanospheres: A comparative study of cytotoxicity and macrophage polarization potential. Carbohydr Polym 2023; 303:120464. [PMID: 36657847 DOI: 10.1016/j.carbpol.2022.120464] [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: 09/30/2022] [Revised: 11/24/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022]
Abstract
Nanocellulose application has been increasing owing to its appealing physicochemical properties. Monitoring of the crystallinity, surface topography, and reactivity of this high-aspect-ratio nanomaterial is crucial for efficient tissue engineering. Controlling macrophage polarization phenotype remains a challenge in regenerative medicine and tissue engineering. Herein, we monitored the effects of shape-regulated (rod and spherical) nanocellulose on the macrophage modulatory potential of RAW 246.7 cells in vitro. Spherical nanocellulose (s-NC) exhibited higher thermal stability and biocompatibility than rod nanocellulose. Macrophage polarization was profoundly affected by nanocellulose topography and incubation period. M2 polarization was observed in vitro after 1 day of treatment with s-NC, followed by M1 polarization after treatment for longer periods. Transcriptome analysis similarly revealed that M1 polarization was dominant after 1 day h of incubation with both nanocellulose types. These findings demonstrate that macrophage polarization can be controlled by selecting suitable nanocellulose shape and incubation time for desired applications.
Collapse
Affiliation(s)
- Dinesh K Patel
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Keya Ganguly
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Sayan Deb Dutta
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Tejal V Patil
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Republic of Korea; Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon 24341, Republic of Korea.
| |
Collapse
|
22
|
Modulation of Macrophage Function by Bioactive Wound Dressings with an Emphasis on Extracellular Matrix-Based Scaffolds and Nanofibrous Composites. Pharmaceutics 2023; 15:pharmaceutics15030794. [PMID: 36986655 PMCID: PMC10053223 DOI: 10.3390/pharmaceutics15030794] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/18/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023] Open
Abstract
Bioactive wound dressings that are capable of regulating the local wound microenvironment have attracted a very large interest in the field of regenerative medicine. Macrophages have many critical roles in normal wound healing, and the dysfunction of macrophages significantly contributes to impaired or non-healing skin wounds. Regulation of macrophage polarization towards an M2 phenotype provides a feasible strategy to enhance chronic wound healing, mainly by promoting the transition of chronic inflammation to the proliferation phase of wound healing, upregulating the level of anti-inflammatory cytokines around the wound area, and stimulating wound angiogenesis and re-epithelialization. Based on this, modulation of macrophage functions by the rational design of bioactive scaffolds has emerged as a promising way to accelerate delayed wound healing. This review outlines current strategies to regulate the response of macrophages using bioactive materials, with an emphasis on extracellular matrix-based scaffolds and nanofibrous composites.
Collapse
|
23
|
Cao D, Ding J. Recent advances in regenerative biomaterials. Regen Biomater 2022; 9:rbac098. [PMID: 36518879 PMCID: PMC9745784 DOI: 10.1093/rb/rbac098] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 07/22/2023] Open
Abstract
Nowadays, biomaterials have evolved from the inert supports or functional substitutes to the bioactive materials able to trigger or promote the regenerative potential of tissues. The interdisciplinary progress has broadened the definition of 'biomaterials', and a typical new insight is the concept of tissue induction biomaterials. The term 'regenerative biomaterials' and thus the contents of this article are relevant to yet beyond tissue induction biomaterials. This review summarizes the recent progress of medical materials including metals, ceramics, hydrogels, other polymers and bio-derived materials. As the application aspects are concerned, this article introduces regenerative biomaterials for bone and cartilage regeneration, cardiovascular repair, 3D bioprinting, wound healing and medical cosmetology. Cell-biomaterial interactions are highlighted. Since the global pandemic of coronavirus disease 2019, the review particularly mentions biomaterials for public health emergency. In the last section, perspectives are suggested: (i) creation of new materials is the source of innovation; (ii) modification of existing materials is an effective strategy for performance improvement; (iii) biomaterial degradation and tissue regeneration are required to be harmonious with each other; (iv) host responses can significantly influence the clinical outcomes; (v) the long-term outcomes should be paid more attention to; (vi) the noninvasive approaches for monitoring in vivo dynamic evolution are required to be developed; (vii) public health emergencies call for more research and development of biomaterials; and (viii) clinical translation needs to be pushed forward in a full-chain way. In the future, more new insights are expected to be shed into the brilliant field-regenerative biomaterials.
Collapse
Affiliation(s)
- Dinglingge Cao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| |
Collapse
|
24
|
Zhao X, Zhou X, Sun H, Shi H, Song Y, Wang Q, Zhang G, Xu D. 3D printed Ti-5Cu alloy accelerates osteogenic differentiation of MC3T3-E1 cells by stimulating the M2 phenotype polarization of macrophages. Front Immunol 2022; 13:1001526. [PMID: 36275667 PMCID: PMC9585254 DOI: 10.3389/fimmu.2022.1001526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 09/22/2022] [Indexed: 11/24/2022] Open
Abstract
Ti-5Cu alloy has been proved to have excellent mechanical properties and cell compatibility and has certain antibacterial properties due to the addition of Cu. However, there are few studies on the effects of Ti-5Cu alloy on macrophage polarization and immune-related bone formation. In this study, we prepared Ti-5Cu alloy by three-dimensional printing technology and found that Ti-5Cu alloy presented a much smoother surface compared with Ti. In addition, the CCK-8 results indicated the Ti-5Cu alloy had no cytotoxicity to RAW264.7 cells by co-culture. The results of inductively coupled plasma mass spectrometry showed that the concentration of Cu2+ was 0.133 mg/L after 7 days of co-culture, and the CCK-8 results proved that Cu2+ had no cytotoxicity to RAW264.7 at this concentration. Then, we studied the effects of Ti-5Cu alloy on macrophage polarization; it was shown that the Ti-5Cu alloy is more prone to modulate the RAW264.7 polarization towards the M2 phenotype and the conditioned medium derived from Ti-5Cu alloy significantly promoted the proliferation and osteogenic differentiation of MC3T3-E1 cells. However, when the expression of Oncostatin M (OSM) gene of RAW264.7 was knocked down, the osteogenic differentiation of MC3T3-E1 cells was decreased. This suggests that the OSM secreted by RAW264.7 co-cultured with Ti-5Cu alloy could accelerate the osteogenic differentiation of MC3T3-E1 cells by acting on OSMR/gp130 receptors.
Collapse
Affiliation(s)
- Xin Zhao
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Xing Zhou
- Shenyang National Laboratory for Materials Science, Northeastern University, Shenyang, China
- Electrobiomaterials Institute, Key Laboratory for Anisotropy and Texture of Materials (Ministry of Education), Northeastern University, Shenyang, China
| | - Hui Sun
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Huixin Shi
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Yiping Song
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Qiang Wang
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
- *Correspondence: Qiang Wang, ; Guangping Zhang,
| | - Guangping Zhang
- Department of Stomatology, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Qiang Wang, ; Guangping Zhang,
| | - Dake Xu
- Shenyang National Laboratory for Materials Science, Northeastern University, Shenyang, China
- Electrobiomaterials Institute, Key Laboratory for Anisotropy and Texture of Materials (Ministry of Education), Northeastern University, Shenyang, China
| |
Collapse
|
25
|
Monteiro NO, Casanova MR, Quinteira R, Fangueiro JF, Reis RL, Neves NM. Biomimetic surface topography as a potential modulator of macrophages inflammatory response to biomaterials. BIOMATERIALS ADVANCES 2022; 141:213128. [PMID: 36179494 DOI: 10.1016/j.bioadv.2022.213128] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 06/16/2023]
Abstract
The implantation of biomaterial devices can negatively impact the local microenvironment through several processes including the injury incurred during the implantation process and the associated host inflammatory response. Immune cell responses to implantable biomaterial devices mediate host-material interactions. Indeed, the immune system plays a central role in several biological processes required for the integration of biomaterials such as wound healing, tissue integration, inflammation, and foreign body reactions. The implant physicochemical properties such as size, shape, surface area, topography, and chemistry have been shown to provide cues to the immune system. Its induced immune-modulatory responses towards inflammatory or wound healing phenotypes can determine the success of the implant. In this work, we aim to evaluate the impact of some biomimetic surface topographies on macrophages' acute inflammatory response. For that, we selected 4 different biological surfaces to replicate through soft lithography on spin casting PCL membranes. Those topographies were: the surface of E. coli, S.eppidermidis and L929 cells cultured in polystyrene tissue culture disks, and an Eggshell membrane. We selected a model based on THP-1-derived macrophages to study the analysis of the expression of both pro-inflammatory and anti-inflammatory markers. Our results revealed that depending on the surface where these cells are seeded, they present different phenotypes. Macrophages present a M1-like phenotype when they are cultured on top of PCL membranes with the surface topography of E. coli and S. epidermidis. When cultured on membranes with L929 monolayers or Eggshell membrane surface topography, the macrophages present a M2-like phenotype. These results can be a significant advance in the development of new implantable biomaterial devices since they can help to modulate the inflammatory responses to implanted biomaterials by controlling their surface topography.
Collapse
Affiliation(s)
- N O Monteiro
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - M R Casanova
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - R Quinteira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - J F Fangueiro
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - R L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - N M Neves
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
26
|
Li J, Luo X, Lv ZY, Qiang HF, Hou CY, Liu K, Meng CX, Zhang YJ, Liu FZ, Zhang B. Microporous structures on mineralized collagen mediate osteogenesis by modulating the osteo-immune response of macrophages. Front Bioeng Biotechnol 2022; 10:917655. [PMID: 36105601 PMCID: PMC9464819 DOI: 10.3389/fbioe.2022.917655] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
It is a new hot pot in tissue engineering and regenerative medicine to study the effects of physicochemical properties of implanted biomaterials on regulating macrophage polarization to promote bone regeneration. In this study, we designed and fabricated mineralized collagen (MC) with different microporous structures via in vitro biomimetic mineralization method. The microporous structures, mechanical properties, shore hardness and water contact angle measurements were tested. Live/dead cell staining, CCK-8 assay, phalloidine staining, staining of focal adhesions were used to detect cell behavior. ELISA, qRT-PCR, ALP, and alizarin red staining (ARS) were performed to appraise osteogenic differentiation and investigated macrophage response and their subsequent effects on the osteogenic differentiation. The results showed that RAW264.7 and MC3T3-E1 cells were able to survive on the MC. MC with the microporous structure of approximately 84 μm and 70%–80% porosity could promote M2 macrophage polarization and increase the expression level of TGF-β and VEGF. Moreover, the gene expression of the osteogenic markers ALP, COL-1, and OCN increased. Therefore, MC with different microporous structures mediated osteoimmunomodulation in bone regeneration. These data will provide a new idea of biomaterials inducing bone repair and direct the optimal design of novel immune biomaterials, development, and rational usage.
Collapse
Affiliation(s)
- Jun Li
- Depertment of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Shandong University & Shandong Provincial Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China
- Liaocheng People’s Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, China
| | - Xin Luo
- Liaocheng People’s Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, China
| | - Zhao-Yong Lv
- Liaocheng People’s Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, China
| | - Hui-Fen Qiang
- Department of Materials Science and Engineering, Liaocheng University, Liaocheng, China
| | - Cai-Yao Hou
- Department of Materials Science and Engineering, Liaocheng University, Liaocheng, China
| | - Kun Liu
- Depertment of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Shandong University & Shandong Provincial Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China
- Liaocheng People’s Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, China
| | - Chun-Xiu Meng
- Liaocheng People’s Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, China
| | - Yu-Jue Zhang
- Liaocheng People’s Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, China
- *Correspondence: Yu-Jue Zhang, ; Feng-Zhen Liu, ; Bin Zhang,
| | - Feng-Zhen Liu
- Liaocheng People’s Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, China
- Department of Materials Science and Engineering, Liaocheng University, Liaocheng, China
- *Correspondence: Yu-Jue Zhang, ; Feng-Zhen Liu, ; Bin Zhang,
| | - Bin Zhang
- Depertment of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Shandong University & Shandong Provincial Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China
- Liaocheng People’s Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, China
- *Correspondence: Yu-Jue Zhang, ; Feng-Zhen Liu, ; Bin Zhang,
| |
Collapse
|
27
|
Wang G, Gao C, Xiao B, Zhang J, Jiang X, Wang Q, Guo J, Zhang D, Liu J, Xie Y, Shu C, Ding J. Research and clinical translation of trilayer stent-graft of expanded polytetrafluoroethylene for interventional treatment of aortic dissection. Regen Biomater 2022; 9:rbac049. [PMID: 35958517 PMCID: PMC9362767 DOI: 10.1093/rb/rbac049] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/03/2022] [Accepted: 07/10/2022] [Indexed: 11/14/2022] Open
Abstract
Abstract
The aortic dissection (AD) is a life-threatening disease. The transcatheter endovascular aortic repair (EVAR) affords a minimally invasive technique to save lives of these critical patients, and an appropriate stent-graft gets to be the key medical device during an EVAR procedure. Herein, we report a trilayer stent-graft and corresponding delivery system used for the treatment of the AD disease. The stent-graft is made of nitinol stents with an asymmetric Z-wave design and two expanded polytetrafluoroethylene (ePTFE) membranes. Each of inner and outer surfaces of the stent-graft was covered by an ePTFE membrane, and the two membranes were then sintered together. The biological studies of the sintered ePTFE membranes indicated that the stent-graft had excellent cytocompatibility and hemocompatibility in vitro. Both the stent-graft and the delivery system exhibited satisfactory mechanical properties and operability. The safety and efficacy of this stent-graft and the corresponding delivery system were demonstrated in vivo. In 9 canine experiments, the blood vessels of the animals implanted with the stent-grafts were of good patency, and there were no thrombus and obvious stenosis by angiography after implantation for 6 months. Furthermore, all of the 9 clinical cases experienced successful implantation using the stent-graft and its post-release delivery system, and the one-year follow-ups indicated the preliminary safety and efficacy of the trilayer stent-graft with an asymmetric Z-wave design for interventional treatment.
Collapse
Affiliation(s)
- Gang Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University , Shanghai, 200438, China
- R&D Center, Lifetech Scientific (Shenzhen) Co., Ltd. , Shenzhen, 518057, China
| | - Caiyun Gao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University , Shanghai, 200438, China
| | - Benhao Xiao
- R&D Center, Lifetech Scientific (Shenzhen) Co., Ltd. , Shenzhen, 518057, China
| | - Jie Zhang
- R&D Center, Lifetech Scientific (Shenzhen) Co., Ltd. , Shenzhen, 518057, China
| | - Xunyuan Jiang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University , Shanghai, 200438, China
| | - Qunsong Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University , Shanghai, 200438, China
| | - Jingzhen Guo
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University , Shanghai, 200438, China
| | - Deyuan Zhang
- R&D Center, Lifetech Scientific (Shenzhen) Co., Ltd. , Shenzhen, 518057, China
| | - Jianxiong Liu
- R&D Center, Lifetech Scientific (Shenzhen) Co., Ltd. , Shenzhen, 518057, China
| | - Yuehui Xie
- R&D Center, Lifetech Scientific (Shenzhen) Co., Ltd. , Shenzhen, 518057, China
| | - Chang Shu
- Department of Vascular Surgery, the Second Xiangya Hospital of Central South University , Changsha, 410011, China
- State Key Laboratory of Cardiovascular Diseases, Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College , Beijing, 100037, China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University , Shanghai, 200438, China
| |
Collapse
|
28
|
Shao J, Weng L, Li J, Lin H, Wang H, Lin J. Regulation of Macrophage Polarization by Mineralized Collagen Coating to Accelerate the Osteogenic Differentiation of Mesenchymal Stem Cells. ACS Biomater Sci Eng 2022; 8:610-619. [PMID: 34991308 DOI: 10.1021/acsbiomaterials.1c00834] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Osteogenesis on the interface between the implant and host bone is a synergistic processing of multiple systems involved in immune response, angiogenesis, osteogenesis, etc. However, regulation of the osteoimmune microenvironment on the implant surface to accelerate the osteogenesis through manipulating the polarization of macrophage phenotype is still beginning to be explored. We here demonstrate that macrophage phenotype is able to be regulated by decoration of mineralized collagen (MC) coating on the titanium implant surface via triggering the integrin-related cascade pathway of macrophages. Furthermore, regulation of the macrophage polarization and construction of the osteoimmune microenvironment by MC coating would subsequently accelerate the osteogenic differentiation of the mesenchymal stem cells. This work therefore emphasizes the importance of the osteoimmune microenvironment on osteogenesis and provides a promising strategy to improve the osteointegration of implants.
Collapse
Affiliation(s)
- Jiaqi Shao
- Department of Stomatology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Luxi Weng
- Department of Stomatology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Juan Li
- Department of Stomatology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Huiping Lin
- Department of Stomatology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Huiming Wang
- Department of Stomatology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.,Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Jun Lin
- Department of Stomatology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
29
|
Xuan Y, Li L, Ma M, Cao J, Zhang Z. Hierarchical Intrafibrillarly Mineralized Collagen Membrane Promotes Guided Bone Regeneration and Regulates M2 Macrophage Polarization. Front Bioeng Biotechnol 2022; 9:781268. [PMID: 35155400 PMCID: PMC8826568 DOI: 10.3389/fbioe.2021.781268] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/23/2021] [Indexed: 12/02/2022] Open
Abstract
Mineralized collagen has been introduced as a promising barrier membrane material for guided bone regeneration (GBR) due to its biomimetic nanostructure. Immune interaction between materials and host significantly influences the outcome of GBR. However, current barrier membranes are insufficient for clinical application due to limited mechanical or osteoimmunomodulatory properties. In this study, we fabricated hierarchical intrafibrillarly mineralized collagen (HIMC) membrane, comparing with collagen (COL) and extrafibrillarly mineralized collagen (EMC) membranes, HIMC membrane exhibited preferable physicochemical properties by mimicking the nanostructure of natural bone. Bone marrow mesenchymal stem cells (BMSCs) seeded on HIMC membrane showed superior proliferation, adhesion, and osteogenic differentiation capacity. HIMC membrane induced CD206+Arg-1+ M2 macrophage polarization, which in turn promoted more BMSCs migration. In rat skull defects, HIMC membrane promoted the regeneration of new bone with more bone mass and more mature bone architecture. The expression levels of Runx2 and osterix and CD68 + CD206 + M2 macrophage polarization were significantly enhanced. HIMC membrane provides an appropriate osteoimmune microenvironment to promote GBR and represents a promising material for further clinical application.
Collapse
Affiliation(s)
- Yaowei Xuan
- Medical School of Chinese PLA, Beijing, China
- Department of Stomatology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Lin Li
- Medical School of Chinese PLA, Beijing, China
- Department of Stomatology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Muzhi Ma
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Junkai Cao
- Department of Stomatology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Zhen Zhang, ; Junkai Cao,
| | - Zhen Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
- *Correspondence: Zhen Zhang, ; Junkai Cao,
| |
Collapse
|
30
|
Liu K, Luo X, Lv ZY, Zhang YJ, Meng Z, Li J, Meng CX, Qiang HF, Hou CY, Hou L, Liu FZ, Zhang B. Macrophage-Derived Exosomes Promote Bone Mesenchymal Stem Cells Towards Osteoblastic Fate Through microRNA-21a-5p. Front Bioeng Biotechnol 2022; 9:801432. [PMID: 35071209 PMCID: PMC8766741 DOI: 10.3389/fbioe.2021.801432] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/08/2021] [Indexed: 12/31/2022] Open
Abstract
The effective healing of a bone defect is dependent on the careful coordination of inflammatory and bone-forming cells. In the current work, pro-inflammatory M1 and anti-inflammatory M2 macrophages were co-cultured with primary murine bone mesenchymal stem cells (BMSCs), in vitro, to establish the cross-talk among polarized macrophages and BMSCs, and as well as their effects on osteogenesis. Meanwhile, macrophages influence the osteogenesis of BMSCs through paracrine forms such as exosomes. We focused on whether exosomes of macrophages promote osteogenic differentiation. The results indicated that M1 and M2 polarized macrophage exosomes all can promote osteogenesis of BMSCs. Especially, M1 macrophage-derived exosomes promote osteogenesis of BMSCs through microRNA-21a-5p at the early stage of inflammation. This research helps to develop an understanding of the intricate interactions among BMSCs and macrophages, which can help to improve the process of bone healing as well as additional regenerative processes by local sustained release of exosomes.
Collapse
Affiliation(s)
- Kun Liu
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Shandong University and Shandong Provincial Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China.,Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, China
| | - Xin Luo
- Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, China
| | - Zhao-Yong Lv
- Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, China
| | - Yu-Jue Zhang
- Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, China
| | - Zhen Meng
- Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, China
| | - Jun Li
- Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, China
| | - Chun-Xiu Meng
- Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, China
| | - Hui-Fen Qiang
- Department of Materials Science and Engineering, Liaocheng University, Liaocheng, China
| | - Cai-Yao Hou
- Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, China
| | - Lei Hou
- Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, China
| | - Feng-Zhen Liu
- Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, China.,Department of Materials Science and Engineering, Liaocheng University, Liaocheng, China
| | - Bin Zhang
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Shandong University and Shandong Provincial Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China.,Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, China
| |
Collapse
|
31
|
Cui Y, Li H, Li Y, Mao L. Novel insights into nanomaterials for immunomodulatory bone regeneration. NANOSCALE ADVANCES 2022; 4:334-352. [PMID: 36132687 PMCID: PMC9418834 DOI: 10.1039/d1na00741f] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/13/2021] [Indexed: 05/02/2023]
Abstract
Bone defect repair caused by trauma, congenital malformation, tumors, infection or systemic diseases remains the focus of attention in regeneration medicine. Recent advances in osteoimmunology indicate that immune cells and correlative cytokines modulate the delicate balance between osteoblasts and osteoclasts and induce a favorable microenvironment for bone regeneration. With superior attributes that imitate the three-dimensional architecture of natural bone, excellent fabricability, mechanical and biological properties, nanomaterials (NMs) are becoming attractive in the field of bone tissue engineering. Particularly, it could be an effective strategy for immunomodulatory bone regeneration by engineering NMs involved in composition nature, nanoarchitectural morphology, surface chemistry, topography and biological molecules, whose mechanisms potentially refer to regulating the phenotype of high-plastic immune cells and inducing cytokine secretion to accelerate osteogenesis. Despite these prominent achievements, the employment of NMs is poorly translated into clinical trials due to the lack of knowledge about the interaction between NMs and the immune system. For this reason, we sketch out the hierarchical structure of bone and its natural healing process, followed by discussion about the effects of immune cells on bone regeneration. Novel horizons focusing on recent progressions in the architectural and physicochemical performances of NMs and their impacts on the body defence mechanism are also emphasized, hoping to provide novel insights for the fabrication of bone graft materials in tissue engineering.
Collapse
Affiliation(s)
- Ya Cui
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology Shanghai China
| | - Hairui Li
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology Shanghai China
| | - Yaxin Li
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology Shanghai China
| | - Lixia Mao
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology Shanghai China
| |
Collapse
|
32
|
Luo Q, Li X, Zhong W, Cao W, Zhu M, Wu A, Chen W, Ye Z, Han Q, Natarajan D, Pathak JL, Zhang Q. Dicalcium silicate-induced mitochondrial dysfunction and autophagy-mediated macrophagic inflammation promotes osteogenic differentiation of BMSCs. Regen Biomater 2021; 9:rbab075. [PMID: 35480858 PMCID: PMC9039510 DOI: 10.1093/rb/rbab075] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/17/2021] [Accepted: 11/30/2021] [Indexed: 11/14/2022] Open
Abstract
Dicalcium silicate (Ca2SiO4, C2S) has osteogenic potential but induces macrophagic inflammation. Mitochondrial function plays a vital role in macrophage polarization and macrophagic inflammation. The mitochondrial function of C2S-treated macrophages is still unclear. This study hypothesized: (i) the C2S modulates mitochondrial function and autophagy in macrophages to regulate macrophagic inflammation, and (ii) C2S-induced macrophagic inflammation regulates osteogenesis. We used RAW264.7 cells as a model of macrophage. The C2S (75-150 μg/ml) extract was used to analyze the macrophagic mitochondrial function and macrophage-mediated effect on osteogenic differentiation of mouse bone marrow-derived mesenchymal stem cells (BMSCs). The results showed that C2S extract (150 μg/ml) induced TNF-α, IL-1β and IL-6 production in macrophages. C2S extract (150 μg/ml) enhanced reactive oxygen species level and intracellular calcium level but reduced mitochondrial membrane potential and ATP production. TEM images showed reduced mitochondrial abundance and altered the mitochondrial morphology in C2S (150 μg/ml)-treated macrophages. Protein level expression of PINK1, Parkin, Beclin1 and LC3 was upregulated but TOMM20 was downregulated. mRNA sequencing and KEGG analysis showed that C2S-induced differentially expressed mRNAs in macrophages were mainly distributed in the essential signaling pathways involved in mitochondrial function and autophagy. The conditioned medium from C2S-treated macrophage robustly promoted osteogenic differentiation in BMSCs. In conclusion, our results indicate mitochondrial dysfunction and autophagy as the possible mechanism of C2S-induced macrophagic inflammation. The promotion of osteogenic differentiation of BMSCs by the C2S-induced macrophagic inflammation suggests the potential application of C2S in developing immunomodulatory bone grafts.
Collapse
Affiliation(s)
- Qianting Luo
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
- Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen 529030, China
| | - Xingyang Li
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Wenchao Zhong
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Wei Cao
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
- Department of Oral Cell Biology, Academic Centre of Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam and University of Amsterdam, Amsterdam 1081LA, The Netherlands
| | - Mingjing Zhu
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Antong Wu
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Wanyi Chen
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Zhitong Ye
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Qiao Han
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Duraipandy Natarajan
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Janak L Pathak
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Qingbin Zhang
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| |
Collapse
|
33
|
Ping J, Zhou C, Dong Y, Wu X, Huang X, Sun B, Zeng B, Xu F, Liang W. Modulating immune microenvironment during bone repair using biomaterials: Focusing on the role of macrophages. Mol Immunol 2021; 138:110-120. [PMID: 34392109 DOI: 10.1016/j.molimm.2021.08.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/09/2021] [Accepted: 08/03/2021] [Indexed: 12/16/2022]
Abstract
Bone is a self-regenerative tissue that can repair small defects and fractures. In large defects, bone tissue is unable to provide nutrients and oxygen for repair, and autologous grafting is used as the gold standard. As an alternative method, the bone tissue regeneration approach uses osteoconductive biomaterials to overcome bone graft disadvantages. However, biomaterials are considered as foreign components that can stimulate host immune responses. Although traditional principles have been aimed to minimize immune reactions, the design of biomaterials has steadily shifted toward creating an immunomodulatory microenvironment to harness immune cells and responses to repair damaged tissue. Among immune cells, macrophages secrete various immunomodulatory mediators and crosstalk with bone-forming cells and play key roles in bone tissue engineering. Macrophage polarization toward M1 and M2 subtypes mediate pro-inflammatory and anti-inflammatory responses, respectively, which are crucial for bone repairing at different stages. This review provides an overview of the crosstalk between various immune cells and biomaterials, macrophage polarization, and the effect of physicochemical properties of biomaterials on the immune responses, especially macrophages, in bone tissue engineering.
Collapse
Affiliation(s)
- Jianfeng Ping
- Department of Orthopaedics, Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing 312000, Zhejiang Province, PR China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua Hospital, Zhoushan 316000, Zhejiang Province, PR China
| | - Yongqiang Dong
- Department of Orthopaedics, Xinchang People's Hospital, Shaoxing 312500, Zhejiang Province, PR China
| | - Xudong Wu
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000, Zhejiang Province, PR China
| | - Xiaogang Huang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000, Zhejiang Province, PR China
| | - Bin Sun
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000, Zhejiang Province, PR China
| | - Bin Zeng
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000, Zhejiang Province, PR China
| | - Fangming Xu
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000, Zhejiang Province, PR China.
| | - Wenqing Liang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000, Zhejiang Province, PR China.
| |
Collapse
|
34
|
Shen Z, Wang S, Shen Z, Tang Y, Xu J, Lin C, Chen X, Huang Q. Deciphering controversial results of cell proliferation on TiO 2 nanotubes using machine learning. Regen Biomater 2021; 8:rbab025. [PMID: 34168893 PMCID: PMC8218935 DOI: 10.1093/rb/rbab025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/19/2021] [Accepted: 05/09/2021] [Indexed: 12/27/2022] Open
Abstract
With the rapid development of biomedical sciences, contradictory results on the relationships between biological responses and material properties emerge continuously, adding to the challenge of interpreting the incomprehensible interfacial process. In the present paper, we use cell proliferation on titanium dioxide nanotubes (TNTs) as a case study and apply machine learning methodologies to decipher contradictory results in the literature. The gradient boosting decision tree model demonstrates that cell density has a higher impact on cell proliferation than other obtainable experimental features in most publications. Together with the variation of other essential features, the controversy of cell proliferation trends on various TNTs is understandable. By traversing all combinational experimental features and the corresponding forecast using an exhausted grid search strategy, we find that adjusting cell density and sterilization methods can simultaneously induce opposite cell proliferation trends on various TNTs diameter, which is further validated by experiments. This case study reveals that machine learning is a burgeoning tool in deciphering controversial results in biomedical researches, opening up an avenue to explore the structure-property relationships of biomaterials.
Collapse
Affiliation(s)
- Ziao Shen
- Department of Physics, Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Laboratory for Soft Functional Materials Research, Xiamen University, Zengcuoan West Road, Siming District, Xiamen 361005, China
| | - Si Wang
- Department of Physics, Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Laboratory for Soft Functional Materials Research, Xiamen University, Zengcuoan West Road, Siming District, Xiamen 361005, China
| | - Zhenyu Shen
- Department of Physics, Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Laboratory for Soft Functional Materials Research, Xiamen University, Zengcuoan West Road, Siming District, Xiamen 361005, China
| | - Yufei Tang
- Department of Physics, Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Laboratory for Soft Functional Materials Research, Xiamen University, Zengcuoan West Road, Siming District, Xiamen 361005, China
| | - Junbin Xu
- Department of Physics, Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Laboratory for Soft Functional Materials Research, Xiamen University, Zengcuoan West Road, Siming District, Xiamen 361005, China
| | - Changjian Lin
- State Key Laboratory for Physical Chemistry of Solid Surfaces, and Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, 422 Siming South Road, Siming District, Xiamen 361005, China
| | - Xun Chen
- Wenzhou Institute, University of Chinese Academy of Sciences, No.16 Xinsan Road, Hi-tech Industrial Park, Wenzhou, Zhejiang, 325000, China
| | - Qiaoling Huang
- Department of Physics, Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Laboratory for Soft Functional Materials Research, Xiamen University, Zengcuoan West Road, Siming District, Xiamen 361005, China
| |
Collapse
|
35
|
Yao X, Wang X, Ding J. Exploration of possible cell chirality using material techniques of surface patterning. Acta Biomater 2021; 126:92-108. [PMID: 33684535 DOI: 10.1016/j.actbio.2021.02.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/10/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023]
Abstract
Consistent left-right (LR) asymmetry or chirality is critical for embryonic development and function maintenance. While chirality on either molecular or organism level has been well established, that on the cellular level has remained an open question for a long time. Although it remains unclear whether chirality exists universally on the cellular level, valuable efforts have recently been made to explore this fundamental topic pertinent to both cell biology and biomaterial science. The development of material fabrication techniques, surface patterning, in particular, has afforded a unique platform to study cell-material interactions. By using patterning techniques, chirality on the cellular level has been examined for cell clusters and single cells in vitro in well-designed experiments. In this review, we first introduce typical fabrication techniques of surface patterning suitable for cell studies and then summarize the main aspects of preliminary evidence of cell chirality on patterned surfaces to date. We finally indicate the limitations of the studies conducted thus far and describe the perspectives of future research in this challenging field. STATEMENT OF SIGNIFICANCE: While both biomacromolecules and organisms can exhibit chirality, it is not yet conclusive whether a cell has left-right (LR) asymmetry. It is important yet challenging to study and reveal the possible existence of cell chirality. By using the technique of surface patterning, the recent decade has witnessed progress in the exploration of possible cell chirality within cell clusters and single cells. Herein, some important preliminary evidence of cell chirality is collected and analyzed. The open questions and perspectives are also described to promote further investigations of cell chirality in biomaterials.
Collapse
|
36
|
Liu Y, Rui Z, Cheng W, Song L, Xu Y, Li R, Zhang X. Characterization and evaluation of a femtosecond laser-induced osseointegration and an anti-inflammatory structure generated on a titanium alloy. Regen Biomater 2021; 8:rbab006. [PMID: 33738120 PMCID: PMC7955712 DOI: 10.1093/rb/rbab006] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 01/05/2021] [Accepted: 01/14/2021] [Indexed: 02/07/2023] Open
Abstract
Cell–material interactions during early osseointegration of the bone–implant interface are critical and involve crosstalk between osteoblasts and osteoclasts. The surface properties of titanium implants also play a critical role in cell–material interactions. In this study, femtosecond laser treatment and sandblasting were used to alter the surface morphology, roughness and wettability of a titanium alloy. Osteoblasts and osteoclasts were then cultured on the resulting titanium alloy disks. Four disk groups were tested: a polished titanium alloy (pTi) control; a hydrophilic micro-dislocation titanium alloy (sandblasted Ti (STi)); a hydrophobic nano-mastoid Ti alloy (femtosecond laser-treated Ti (FTi)); and a hydrophilic hierarchical hybrid micro-/nanostructured Ti alloy [femtosecond laser-treated and sandblasted Ti (FSTi)]. The titanium surface treated by the femtosecond laser and sandblasting showed higher biomineralization activity and lower cytotoxicity in simulated body fluid and lactate dehydrogenase assays. Compared to the control surface, the multifunctional titanium surface induced a better cellular response in terms of proliferation, differentiation, mineralization and collagen secretion. Further investigation of macrophage polarization revealed that increased anti-inflammatory factor secretion and decreased proinflammatory factor secretion occurred in the early response of macrophages. Based on the above results, the synergistic effect of the surface properties produced an excellent cellular response at the bone–implant interface, which was mainly reflected by the promotion of early ossteointegration and macrophage polarization.
Collapse
Affiliation(s)
- Yang Liu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, China.,Institute of Medical Service Support, Institute of Systems Engineering, Academy of Military Sciences, Tianjin, China
| | - Zhongying Rui
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Wei Cheng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, China.,Institute of Medical Service Support, Institute of Systems Engineering, Academy of Military Sciences, Tianjin, China
| | - Licheng Song
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, China.,Institute of Medical Service Support, Institute of Systems Engineering, Academy of Military Sciences, Tianjin, China
| | - Yunqiang Xu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ruixin Li
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin 300041, China
| | - Xizheng Zhang
- Institute of Medical Service Support, Institute of Systems Engineering, Academy of Military Sciences, Tianjin, China
| |
Collapse
|
37
|
Li J, Jiang X, Li H, Gelinsky M, Gu Z. Tailoring Materials for Modulation of Macrophage Fate. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2004172. [PMID: 33565154 PMCID: PMC9245340 DOI: 10.1002/adma.202004172] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/31/2020] [Indexed: 05/03/2023]
Abstract
Human immune system acts as a pivotal role in the tissue homeostasis and disease progression. Immunomodulatory biomaterials that can manipulate innate immunity and adaptive immunity hold great promise for a broad range of prophylactic and therapeutic purposes. This review is focused on the design strategies and principles of immunomodulatory biomaterials from the standpoint of materials science to regulate macrophage fate, such as activation, polarization, adhesion, migration, proliferation, and secretion. It offers a comprehensive survey and discussion on the tunability of material designs regarding physical, chemical, biological, and dynamic cues for modulating macrophage immune response. The range of such tailorable cues encompasses surface properties, surface topography, materials mechanics, materials composition, and materials dynamics. The representative immunoengineering applications selected herein demonstrate how macrophage-immunomodulating biomaterials are being exploited for cancer immunotherapy, infection immunotherapy, tissue regeneration, inflammation resolution, and vaccination. A perspective on the future research directions of immunoregulatory biomaterials is also provided.
Collapse
Affiliation(s)
- Jinhua Li
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Xinquan Jiang
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Hongjun Li
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| |
Collapse
|
38
|
Meng C, Liu K, Lv Z, Zhang Y, Li J, Luo X, Qiang H, Li K, Liu F, Zhang B, Cui F. Inflammation and immunity gene expression profiling of macrophages on mineralized collagen. J Biomed Mater Res A 2020; 109:1328-1336. [PMID: 33089616 DOI: 10.1002/jbm.a.37124] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/11/2020] [Accepted: 10/16/2020] [Indexed: 12/18/2022]
Abstract
Mineralized collagen (MC) is a biomaterial that is commonly used in the treatment of bone defects. However, the inflammatory response after biomaterial implantation is a recurrent problem that requires urgent attention. Our previous studies on MC-macrophage interactions were descriptive but we did not perform an in-depth analysis on a genetic level to investigate the underlying mechanisms. In this study, we cultured RAW264.7 cells on MC or collagen and examined the proliferation of the macrophages by Cell Counting Kit-8 assay. We sequenced the RNA of the cultured cells to discover differential gene expression patterns and found that a total of 1183 genes were differentially expressed between the MC- and collagen-cultured groups, of which 396 genes were upregulated and 787 were downregulated. Gene ontology analysis revealed that biological processes in MC-cultured cells, such as inflammation and innate immunity, were downregulated; whereas nucleosome assembly, megakaryocyte differentiation, and chromatin assembly were upregulated. We identified several pathways associated with immunity that were significantly enriched using the Kyoto Encyclopedia of Genes and Genomes. Furthermore, we validated the differentially expressed genes from RNA sequencing by quantitative real-time polymerase chain reaction. This study provides insight into the macrophage phenotype based on the microenvironment, which is the foundation for the clinical application of MC-based interventions.
Collapse
Affiliation(s)
- Chunxiu Meng
- Depertment of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Shandong University and Shandong Provincial Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China.,Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, China
| | - Kun Liu
- Depertment of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Shandong University and Shandong Provincial Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China.,Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, China
| | - Zhaoyong Lv
- Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, China
| | - Yujue Zhang
- Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, China
| | - Jun Li
- Depertment of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Shandong University and Shandong Provincial Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China.,Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, China
| | - Xin Luo
- Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, China
| | - Huifen Qiang
- College of Materials Science and Engineering of Liaocheng University, Liaocheng, China
| | - Keyi Li
- Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, China
| | - Fengzhen Liu
- Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, China.,College of Materials Science and Engineering of Liaocheng University, Liaocheng, China
| | - Bin Zhang
- Depertment of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Shandong University and Shandong Provincial Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China.,Liaocheng People's Hospital, Medical College of Liaocheng University, Liaocheng, China
| | - Fuzhai Cui
- Department of Materials Science and Engineering, Tsinghua University, Beijing, China
| |
Collapse
|