1
|
Asadi Tokmedash M, Kim C, Chavda AP, Li A, Robins J, Min J. Engineering multifunctional surface topography to regulate multiple biological responses. Biomaterials 2025; 319:123136. [PMID: 39978049 PMCID: PMC11893264 DOI: 10.1016/j.biomaterials.2025.123136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/04/2025] [Accepted: 01/23/2025] [Indexed: 02/22/2025]
Abstract
Surface topography or curvature plays a crucial role in regulating cell behavior, influencing processes such as adhesion, proliferation, and gene expression. Recent advancements in nano- and micro-fabrication techniques have enabled the development of biomimetic systems that mimic native extracellular matrix (ECM) structures, providing new insights into cell-adhesion mechanisms, mechanotransduction, and cell-environment interactions. This review examines the diverse applications of engineered topographies across multiple domains, including antibacterial surfaces, immunomodulatory devices, tissue engineering scaffolds, and cancer therapies. It highlights how nanoscale features like nanopillars and nanospikes exhibit bactericidal properties, while many microscale patterns can direct stem cell differentiation and modulate immune cell responses. Furthermore, we discuss the interdisciplinary use of topography for combined applications, such as the simultaneous regulation of immune and tissue cells in 2D and 3D environments. Despite significant advances, key knowledge gaps remain, particularly regarding the effects of topographical cues on multicellular interactions and dynamic 3D contexts. This review summarizes current fabrication methods, explores specific and interdisciplinary applications, and proposes future research directions to enhance the design and utility of topographically patterned biomaterials in clinical and experimental settings.
Collapse
Affiliation(s)
| | - Changheon Kim
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ajay P Chavda
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Adrian Li
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jacob Robins
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jouha Min
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA; Weil Institute for Critical Care Research and Innovation, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
2
|
Ziegelmeyer T, Martins de Sousa K, Liao TY, Lartizien R, Delay A, Vollaire J, Josserand V, Linklater D, Le PH, Coll JL, Bettega G, Ivanova EP, Martel-Frachet V. Multifunctional micro/nano-textured titanium with bactericidal, osteogenic, angiogenic and anti-inflammatory properties: Insights from in vitro and in vivo studies. Mater Today Bio 2025; 32:101710. [PMID: 40230651 PMCID: PMC11994386 DOI: 10.1016/j.mtbio.2025.101710] [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: 12/05/2024] [Revised: 03/06/2025] [Accepted: 03/25/2025] [Indexed: 04/16/2025] Open
Abstract
Titanium (Ti) is widely used as an implantable material for bone repair in orthopedics and dentistry. However, Ti implants are vulnerable to bacterial infections, which can compromise patient recovery and lead to implant failure. While a controlled inflammatory response promotes bone regeneration, chronic inflammation caused by infections can lead to implant failure. Bone repair is a complex process in which inflammation, angiogenesis and osteogenesis are tightly interconnected, requiring cooperation between mesenchymal stem cells (MSC), macrophages and endothelial cells. Here, we fabricated bio-inspired Ti implants with either microstructured (Micro Ti) or nanostructured (Nano Ti) surface textures that exhibit robust mechano-bactericidal properties. In vitro, both textured surfaces improved blood coagulation and osteogenic marker expression compared to smooth Ti surfaces. Additionally, Nano Ti promoted macrophage polarization towards the M2 phenotype and enhanced the paracrine effects of MSCs on angiogenesis, key processes in tissue regeneration. In vivo kinetic analysis of bone reconstruction in a rat calvarial model showed that Nano Ti improved osseointegration, as evidenced by increased bone volume, mineral density, and bone-implant contact. Notably, the Micro Ti surface showed no significant differences from the control implants. These findings highlight the potential of mechano-bactericidal surface nanopatterns to simultaneously prevent infections and enhance osseointegration by modulating protein adsorption, inflammation, angiogenesis and osteogenesis. This study provides new insights into the development of bifunctional Ti implants, offering new perspectives for the next generation of implantable bone-related biomaterials.
Collapse
Affiliation(s)
- Théo Ziegelmeyer
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Institute for Advanced Biosciences, F38000, Grenoble, France
| | | | - Tzu-Ying Liao
- School of Science, RMIT University, Melbourne, VIC, 3000, Australia
| | - Rodolphe Lartizien
- Service de Chirurgie Maxillo-Faciale, Centre Hospitalier Annecy Genevois, 1 Avenue de L'hôpital, Epagny Metz-Tessy, F-74370, France
| | - Alexandra Delay
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Institute for Advanced Biosciences, F38000, Grenoble, France
| | - Julien Vollaire
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Institute for Advanced Biosciences, F38000, Grenoble, France
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Optimal Platform, Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Véronique Josserand
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Institute for Advanced Biosciences, F38000, Grenoble, France
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Optimal Platform, Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Denver Linklater
- Department of Biomedical Engineering, The Graeme Clark Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Phuc H. Le
- School of Science, RMIT University, Melbourne, VIC, 3000, Australia
| | - Jean-Luc Coll
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Institute for Advanced Biosciences, F38000, Grenoble, France
| | - Georges Bettega
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Institute for Advanced Biosciences, F38000, Grenoble, France
- Service de Chirurgie Maxillo-Faciale, Centre Hospitalier Annecy Genevois, 1 Avenue de L'hôpital, Epagny Metz-Tessy, F-74370, France
| | - Elena P. Ivanova
- School of Science, RMIT University, Melbourne, VIC, 3000, Australia
| | - Véronique Martel-Frachet
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Institute for Advanced Biosciences, F38000, Grenoble, France
- University PSL Research, EPHE, F75014, Paris, France
| |
Collapse
|
3
|
Sanati M, Pieterman I, Levy N, Akbari T, Tavakoli M, Hassani Najafabadi A, Amin Yavari S. Osteoimmunomodulation by bone implant materials: harnessing physicochemical properties and chemical composition. Biomater Sci 2025; 13:2836-2870. [PMID: 40289736 DOI: 10.1039/d5bm00357a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Chronic inflammation at bone defect sites can impede regenerative processes, but local immune responses can be adjusted to promote healing. Regulating the osteoimmune microenvironment, particularly through macrophage polarization, has become a key focus in bone regeneration research. While bone implants are crucial for addressing significant bone defects, they are often recognized by the immune system as foreign, triggering inflammation that leads to bone resorption and implant issues like fibrous encapsulation and aseptic loosening. Developing osteoimmunomodulatory implants offers a promising approach to transforming destructive inflammation into healing processes, enhancing implant integration and bone regeneration. This review explores strategies based on tuning the physicochemical attributes and chemical composition of materials in engineering osteoimmunomodulatory and pro-regenerative bone implants.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands.
| | - Ines Pieterman
- Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Natacha Levy
- Metabolic Diseases Pediatrics Division, University Medical Centre Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Tayebeh Akbari
- Department of Microbiology, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Mohamadreza Tavakoli
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Saber Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands.
- Regenerative Medicine Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
4
|
Thilakan AT, Nandakumar N, Menon RS, Nair SV, Shenoy V, Sathy BN. The impact of size scales and orientations of polymeric scaffold architectural cues on human macrophage polarisation. Biomed Mater 2025; 20:035026. [PMID: 40273929 DOI: 10.1088/1748-605x/add06e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 04/24/2025] [Indexed: 04/26/2025]
Abstract
Macrophage polarisation is crucial for initiating inflammation in response to biomaterial scaffolds, significantly influencing tissue integration and regenerationin vivo. Modulating macrophage polarisation towards a tissue-regeneration-favouring phenotype through the physical properties of scaffolds offers a promising strategy to enhance tissue regeneration while minimising unfavourable immune responses. However, the critical impact of scaffold physical properties, such as size-scale dimensions, orientation of architectural cues, and local-stiffness of these cues on macrophage polarisation, remains largely unexplored and inadequately understood. This study investigates the combinatorial effects of the physical properties of 3D scaffolds made from poly (ϵ-caprolactone) on human macrophage polarisation. Our findings indicate that the size-scale dimensions and orientation of the architectural cues of the scaffold play crucial roles in determining cell shape, attachment, and the modulation of key gene expression (iNOS, IL-1β, MRC1, ARG), as well as cytokines (TNF-α, IL-10) release associated with the polarisation of human macrophages. Specifically, in scaffolds with architectural cues at larger scales (⩾300 µm diameter), macrophage polarisation is primarily determined by the size-scale of the architectural cues and scaffold stiffness, rather than orientation. Conversely, at smaller scales (⩽15 µm), the orientation of the scaffold's architectural cues plays a more critical role. These insights underscore the pivotal role of scaffold design in modulating immune responses for enhanced tissue regeneration, offering valuable guidance for the rational development of biomaterial scaffolds in regenerative medicine.
Collapse
Affiliation(s)
- Akhil T Thilakan
- Amrita School of Nanosciences and Molecular medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Niji Nandakumar
- Amrita School of Nanosciences and Molecular medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Revathy S Menon
- Amrita School of Nanosciences and Molecular medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Shantikumar V Nair
- Amrita School of Nanosciences and Molecular medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Veena Shenoy
- Department of Transfusion Medicine, Amrita Institute of Medical Sciences and Research Centre, Kochi, Kerala, India
| | - Binulal Nelson Sathy
- Amrita School of Nanosciences and Molecular medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| |
Collapse
|
5
|
Bai FJ, Wang H, Hu YQ, Shao YF, Zhu YR, Jiang YL, Hu JC, Zhao HJ, Zhang KQ. Effectively Guiding Cell Elongation and Alignment by Constructing Micro/Nano Hierarchical Patterned Titania on Titanium Substrate. Biotechnol Bioeng 2025; 122:1272-1283. [PMID: 39887678 DOI: 10.1002/bit.28934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 02/01/2025]
Abstract
Based on the innate sensitivity of cell to substrate topographical cues, modulating cell-directed growth behavior is crucial for promoting tissue repair and reconstruction. Although photolithography technology has been extensively employed to fabricate a variety of anisotropic patterned structures to guide cell growth, it remains a great challenge to design high-resolution micro/nano hierarchical structures directly onto medical titanium (Ti)-based implants. Herein, we present a rapid, reliable and reproducible approach combining photolithography and hydrothermal technology to construct a micro/nano hierarchical structure including anisotropic micro-strips and a porous structure composed of TiO2 nanotubes features. In vitro biological and physicochemical analyses revealed that the micro/nano hierarchical structures not only efficiently facilitate the localization and adsorption of BSA molecules, but also enhances the control of cell growth behavior. The synergistic effect between the physical limitation for organizing cellular cytoskeleton at micropattern and the control of focal adhesion sits at the nanoscale can effectively guide cells to maintain stable elongation and alignment, even at large micro-stripe width of 100 μm. This study presents a promising strategy to precisely construct micro/nano multi-level patterned structure on Ti substrate using biomaterials with excellent biocompatibility. These functional micro/nano hybrid micropatterns offer a powerful platform for regulating bioreagent localization and cell behaviors in various applications including tissue engineering, regenerative medicine, drug screening, and biosensors.
Collapse
Affiliation(s)
- Feng-Jiao Bai
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, PR China
| | - Hui Wang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, PR China
| | - Yu-Qing Hu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, PR China
| | - Yun-Fei Shao
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, PR China
| | - Yi-Ran Zhu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, PR China
| | - Yu-Lin Jiang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, PR China
| | - Jian-Chen Hu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, PR China
| | - Hui-Jing Zhao
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, PR China
| | - Ke-Qin Zhang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, PR China
| |
Collapse
|
6
|
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
|
7
|
Cai X, Zhang S, Xiao C, Dang Z, Huang W, Xu W, Wu G. Orchestrating Macrophage and Bone Mesenchymal Stem Cells to Promote Bone Regeneration via Modulation of the Internal Surface Morphology inside 3D Printed Scaffolds. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:8853-8865. [PMID: 40146177 DOI: 10.1021/acs.langmuir.5c00200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Surface morphology has been widely used to orchestrate multicellular function. However, most studies are mainly based on two-dimensional (2D) surface morphology. Therefore, a new scaffold that could be used to design and obtain controllable internal surface morphology was fabricated to explore the effect of a micropatterned scaffold on bone repair. In this study, through the combination of three-dimensional (2D) printing and soft lithography, a controllable micropatterned poly(ε-caprolactone) scaffold was obtained, which realized the transformation from 2D micropattern research to 3D research. Pit micropatterns with morphology sizes of 0, 25, and 45 μm (Flat, P25, and P45) were constructed. In vitro, the results showed that the P25 micropattern had a better effect on the promotion of M2 polarization, inhibition of the M1 polarization of RAW264.7 cells, and promotion of the osteogenic differentiation of bone marrow stromal stem cells (BMSCs). Direct and indirect coculture models of macrophages and BMSCs were constructed to study the bone immunomodulation of the pit micropatterns. Compared with the Flat and P45 groups, the P25 group could promote the secretion of M2 markers, inhibit the secretion of M1 markers, and immunomodulate the promotion of osteogenic differentiation of BMSCs. In vivo, the results also showed that the P25 group had a lower proinflammatory effect and better performance than scaffolds without micropatterned surfaces and a bigger morphology size (the P45 group), which could regulate the immune function of macrophages, reduce the inflammatory response, and accelerate bone regeneration and repair. This work provides a new strategy for the preparation of scaffolds for bone defect regeneration.
Collapse
Affiliation(s)
- Xiayu Cai
- National Engineering Research Center for Human Tissue Restoration and Function Reconstruction, South China University of Technology, Wushan Road 381, Guangzhou, Guangdong 510006, China
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong 510316, China
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangdong Institute of Medical Instruments, No. 1307, Guangzhou Avenue Central, Tianhe District, Guangzhou, Guangdong 510500, China
| | - Shaohui Zhang
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong 510316, China
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangdong Institute of Medical Instruments, No. 1307, Guangzhou Avenue Central, Tianhe District, Guangzhou, Guangdong 510500, China
| | - Chujie Xiao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong 510316, China
- School of Biology and Biological Engineering, South China University of Technology, No. 381 Wushan Road, Tianhe District, Guangzhou, Guangdong 510641, China
| | - Zhaohui Dang
- National Engineering Research Center for Human Tissue Restoration and Function Reconstruction, South China University of Technology, Wushan Road 381, Guangzhou, Guangdong 510006, China
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong 510316, China
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangdong Institute of Medical Instruments, No. 1307, Guangzhou Avenue Central, Tianhe District, Guangzhou, Guangdong 510500, China
| | - Weihua Huang
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong 510316, China
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangdong Institute of Medical Instruments, No. 1307, Guangzhou Avenue Central, Tianhe District, Guangzhou, Guangdong 510500, China
- The Second Affiliated Hospital of Guangzhou Medical University, The Second Clinical Medicine School of Guangzhou Medical University, No. 250, Changgang East Road, Haizhu District, Guangzhou, Guangdong 510260, China
- Department of Orthopedic Surgery, Affiliated Qingyuan Hospital, Qingyuan People's Hospital, Guangzhou Medical University, No. 35, Yinquan North Road, Qingcheng District, Qingyuan, Guangdong 511518, China
| | - Weikang Xu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong 510316, China
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangdong Institute of Medical Instruments, No. 1307, Guangzhou Avenue Central, Tianhe District, Guangzhou, Guangdong 510500, China
- Guangdong Chinese Medicine Intelligent Diagnosis and Treatment Engineering Technology Research Center, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong 510316, China
| | - Gang Wu
- National Engineering Research Center for Human Tissue Restoration and Function Reconstruction, South China University of Technology, Wushan Road 381, Guangzhou, Guangdong 510006, China
| |
Collapse
|
8
|
Liu Y, Gao M, Yan Y, Wang X, Dong Z, Cheng L, Xu Y. Immunology in Osseointegration After Implantation. J Biomed Mater Res B Appl Biomater 2025; 113:e35566. [PMID: 40130467 DOI: 10.1002/jbm.b.35566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/26/2025]
Abstract
Bone tissue is renowned for its regenerative capabilities, yet handling extensive defects and complex fractures presents considerable medical challenges. Osteoimmunology, studying the complex mechanism of the mutual influence within the range of immunity and skeletal systems, has highlighted the indispensable function of immune reactions in the process of bone integration. This procedure, primarily immune-driven, fosters new bone formation around implants instead of relying solely on osteogenic mechanisms. Traditionally, implant research has emphasized bone bonding and osteoinduction, often overlooking the significant influence of immune interactions. Implants pose risks including donor site morbidity, decreased bioactivity, and pathogen transmission risks. To mitigate these, implant surfaces are modified through altering local factors such as electrical fields and applying physical treatments to change roughness, hydrophilicity, and nanotopography. These modifications aim to regulate immune reactions at the surface of the bone implant, improving osseointegration and the repair of bone. This review examines the types of immune cells in osseointegration, especially the pivotal function that macrophages serve in the regeneration of bone tissue, and investigates key implant features-morphology, wettability, cytokine interaction, and metal ion and bioactive molecule adsorption-that impact immune responses. These insights underscore the immune system's importance in bone repair and advance osteoimmunology as essential for developing strategies to control bone immune responses, enhancing implant integration and bone regeneration.
Collapse
Affiliation(s)
- Yuyang Liu
- School of Basic Medical Sciences, Chengdu University, Chengdu, China
| | - Min Gao
- School of Basic Medical Sciences, Chengdu University, Chengdu, China
| | - Yikun Yan
- School of Basic Medical Sciences, Chengdu University, Chengdu, China
| | - Xue Wang
- School of Basic Medical Sciences, Chengdu University, Chengdu, China
| | - Zhihong Dong
- School of Basic Medical Sciences, Chengdu University, Chengdu, China
| | - Lijia Cheng
- School of Basic Medical Sciences, Chengdu University, Chengdu, China
| | - Yaji Xu
- School of Basic Medical Sciences, Chengdu University, Chengdu, China
| |
Collapse
|
9
|
Negrescu AM, Nistorescu S, Bonciu A, Rusen L, Dumitrescu N, Urzica I, Moldovan A, Hoffmann P, Pircalaboiu GG, Cimpean A, Dinca V. PDMS biointerfaces featuring honeycomb-like well microtextures designed for a pro-healing environment. RSC Adv 2025; 15:9952-9967. [PMID: 40171285 PMCID: PMC11959266 DOI: 10.1039/d5ra00063g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/21/2025] [Indexed: 04/03/2025] Open
Abstract
Even today, the reduction of complications following breast implant surgery together with the enhancement of implant integration and performance through the modulation of the foreign body response (FBR), remains a fundamental challenge in the field of plastic surgery. Therefore, tailoring the material's physical characteristics to modulate FBR can represent an effective approach in implantology. While polydimethylsiloxane (PDMS) patterning on 2D substrates is a relatively established and available procedure, micropatterning multiscaled biointerfaces on a controlled large area has been more challenging. Therefore, in the present work, a specific designed honeycomb-like well biointerface was designed and obtained by replication in PDMS at large scale and its effectiveness towards creating a pro-healing environment was investigated. The grayscale masks assisted laser-based 3D texturing method was used for creating the required moulds in Polycarbonate for large area replication. By comparison to the smooth substrate, the honeycomb topography altered the fibroblasts' behaviour in terms of adhesion and morphology and reduced the macrophages' inflammatory response. Additionally, the microstructured surface effectively inhibited macrophage fusion, significantly limiting the colonization of both Gram-positive and Gram-negative microbial strains on the tested surfaces. Overall, this study introduces an innovative approach to mitigate the in vitro FBR to silicone, achieved through the creation of a honeycomb-inspired topography for prosthetic interfaces.
Collapse
Affiliation(s)
- Andreea Mariana Negrescu
- Faculty of Biology, University of Bucharest Splaiul Independenţei 91-95 050095 Bucharest Romania
- Research Institute of the University of Bucharest (ICUB), University of Bucharest 050657 Bucharest Romania
| | - Simona Nistorescu
- Faculty of Biology, University of Bucharest Splaiul Independenţei 91-95 050095 Bucharest Romania
- National Institute for Lasers, Plasma, and Radiation Physics 409 Atomistilor Street 077125 Romania
| | - Anca Bonciu
- National Institute for Lasers, Plasma, and Radiation Physics 409 Atomistilor Street 077125 Romania
| | - Laurentiu Rusen
- National Institute for Lasers, Plasma, and Radiation Physics 409 Atomistilor Street 077125 Romania
| | - Nicoleta Dumitrescu
- National Institute for Lasers, Plasma, and Radiation Physics 409 Atomistilor Street 077125 Romania
| | - Iuliana Urzica
- National Institute for Lasers, Plasma, and Radiation Physics 409 Atomistilor Street 077125 Romania
| | - Antoniu Moldovan
- National Institute for Lasers, Plasma, and Radiation Physics 409 Atomistilor Street 077125 Romania
| | - Patrick Hoffmann
- Laboratory for Advanced Materials Processing, EMPA, Swiss Federal Institute for Materials Science and Technology Feuerstrasse 39 CH-3602 Thun Swizerland
| | - Gratiela Gradisteanu Pircalaboiu
- Faculty of Biology, University of Bucharest Splaiul Independenţei 91-95 050095 Bucharest Romania
- Polytechnic University of Bucharest Splaiul Independenţei 313 Bucharest 060042 Romania
| | - Anisoara Cimpean
- Faculty of Biology, University of Bucharest Splaiul Independenţei 91-95 050095 Bucharest Romania
| | - Valentina Dinca
- National Institute for Lasers, Plasma, and Radiation Physics 409 Atomistilor Street 077125 Romania
| |
Collapse
|
10
|
Zhao Y, An Y, Wu F, Liu L, Tay FR, Jiao Y, Wang J. Regulation of immune microenvironments by polyetheretherketone surface topography for improving osseointegration. J Nanobiotechnology 2025; 23:199. [PMID: 40069791 PMCID: PMC11895393 DOI: 10.1186/s12951-025-03272-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 02/24/2025] [Indexed: 03/15/2025] Open
Abstract
Optimizing the immune microenvironment is essential for successful implant osseointegration. In this study, four different nano/microstructures were fabricated on polyetheretherketone (PEEK) substrates by varying the agitation speed during sulfonation to influence osteoimmunomodulation and implant integration. The results indicate that nano/microstructures with minimal dimensions (SP450) inhibit actin polymerization by reducing calcium influx through PIEZO1, activating the anti-inflammatory M2 macrophage phenotype. Among the tested specimens, SP450 exhibited the lowest expression levels of tumor necrosis factor-α and interleukin-1β while releasing the highest levels of anti-inflammatory factors, including interleukin-4 and interleukin-10. This optimized immune environment promotes the osteogenesis of MC3T3-E1 pre-osteoblasts and enhances the osseointegration of PEEK implants. Transcriptomic analysis and validation experiment further revealed that SP450 inhibits osteoclastic differentiation by down-regulating transforming growth factor-β2 and suppressing the NF-κB signaling pathway. These findings suggest that manipulating the surface topography of PEEK implants is an effective strategy for enhancing osseointegration with promising clinical applications.
Collapse
Affiliation(s)
- Yuqing Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, PR China
| | - Yanxin An
- Department of General Surgery, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, PR China
| | - Fan Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, PR China
| | - Lipeng Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, PR China
| | - Franklin R Tay
- The Graduate School, Augusta University, Augusta, GA, 30912, USA
| | - Yang Jiao
- Department of Stomatology, The Seventh Medical Center of PLA General Hospital, Beijing, 100700, PR China.
| | - Jing Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, PR China.
| |
Collapse
|
11
|
Liu H, Cui H, Liu G. The Intersection between Immune System and Idiopathic Pulmonary Fibrosis-A Concise Review. FIBROSIS (HONG KONG, CHINA) 2025; 3:10004. [PMID: 40124525 PMCID: PMC11928166 DOI: 10.70322/fibrosis.2025.10004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is marked by progressive alveolar destruction, impaired tissue regeneration, and relentless fibrogenesis, culminating in respiratory failure and death. A diverse array of resident and non-resident cells within the lung contribute to disease pathogenesis. Notably, immune cells, both resident and recruited, respond to cues from sites of lung injury by undergoing phenotypic transitions and producing a wide range of mediators that influence, initiate, or dictate the function, or dysfunction, of key effector cells in IPF pathology, such as alveolar epithelial cells, lung fibroblasts, and capillary endothelial cells. The role of the immune system in IPF has undergone an interesting evolution, oscillating from initial enthusiasm to skepticism, and now to a renewed focus. This shift reflects both the past failures of immune-targeting therapies for IPF and the unprecedented insights into immune cell heterogeneity provided by emerging technologies. In this article, we review the historical evolution of perspectives on the immune system's role in IPF pathogenesis and examine the lessons learned from previous therapeutic failures targeting immune responses. We discuss the major immune cell types implicated in IPF progression, highlighting their phenotypic transitions and mechanisms of action. Finally, we identify key knowledge gaps and propose future directions for research on the immune system in IPF.
Collapse
Affiliation(s)
- Hongli Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Huachun Cui
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gang Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
12
|
Mishchenko O, Volchykhina K, Maksymov D, Manukhina O, Pogorielov M, Pavlenko M, Iatsunskyi I. Advanced Strategies for Enhancing the Biocompatibility and Antibacterial Properties of Implantable Structures. MATERIALS (BASEL, SWITZERLAND) 2025; 18:822. [PMID: 40004345 PMCID: PMC11857362 DOI: 10.3390/ma18040822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025]
Abstract
This review explores the latest advancements in enhancing the biocompatibility and antibacterial properties of implantable structures, with a focus on titanium (Ti) and its alloys. Titanium implants, widely used in dental and orthopedic applications, demonstrate excellent mechanical strength and biocompatibility, yet face challenges such as peri-implantitis, a bacterial infection that can lead to implant failure. To address these issues, both passive and active surface modification strategies have been developed. Passive modifications, such as altering surface texture and chemistry, aim to prevent bacterial adhesion, while active approaches incorporate antimicrobial agents for sustained infection control. Nanotechnology has emerged as a transformative tool, enabling the creation of nanoscale materials and coatings like TiO2 and ZnO that promote osseointegration and inhibit biofilm formation. Techniques such as plasma spraying, ion implantation, and plasma electrolytic oxidation (PEO) show promising results in improving implant integration and durability. Despite significant progress, further research is needed to refine these technologies, optimize surface properties, and address the clinical challenges associated with implant longevity and safety. This review highlights the intersection of surface engineering, nanotechnology, and biomedical innovation, paving the way for the next generation of implantable devices.
Collapse
Affiliation(s)
- Oleg Mishchenko
- Department of Dentistry of Postgraduate Education, Zaporizhzhia State Medical and Pharmaceutical University, 26 Marii Prymachenko Blvd., 69035 Zaporizhzhia, Ukraine; (K.V.); (D.M.); (O.M.)
| | - Kristina Volchykhina
- Department of Dentistry of Postgraduate Education, Zaporizhzhia State Medical and Pharmaceutical University, 26 Marii Prymachenko Blvd., 69035 Zaporizhzhia, Ukraine; (K.V.); (D.M.); (O.M.)
| | - Denis Maksymov
- Department of Dentistry of Postgraduate Education, Zaporizhzhia State Medical and Pharmaceutical University, 26 Marii Prymachenko Blvd., 69035 Zaporizhzhia, Ukraine; (K.V.); (D.M.); (O.M.)
| | - Olesia Manukhina
- Department of Dentistry of Postgraduate Education, Zaporizhzhia State Medical and Pharmaceutical University, 26 Marii Prymachenko Blvd., 69035 Zaporizhzhia, Ukraine; (K.V.); (D.M.); (O.M.)
| | - Maksym Pogorielov
- Insitute of Atomic Physics and Spectroscopy, University of Latvia, 3 Jelgavas Str., LV-1004 Riga, Latvia;
| | - Mykola Pavlenko
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej Str. 3, 61-614 Poznan, Poland;
| | - Igor Iatsunskyi
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej Str. 3, 61-614 Poznan, Poland;
| |
Collapse
|
13
|
Zhai Z, Yang C, Yin W, Liu Y, Li S, Ye Z, Xie M, Song X. Engineered Strategies to Interfere with Macrophage Fate in Myocardial Infarction. ACS Biomater Sci Eng 2025; 11:784-805. [PMID: 39884780 DOI: 10.1021/acsbiomaterials.4c02061] [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: 02/01/2025]
Abstract
Myocardial infarction (MI), a severe cardiovascular condition, is typically triggered by coronary artery disease, resulting in ischemic damage and the subsequent necrosis of the myocardium. Macrophages, known for their remarkable plasticity, are capable of exhibiting a range of phenotypes and functions as they react to diverse stimuli within their local microenvironment. In recent years, there has been an increasing number of studies on the regulation of macrophage behavior based on tissue engineering strategies, and its regulatory mechanisms deserve further investigation. This review first summarizes the effects of key regulatory factors of engineered biomaterials (including bioactive molecules, conductivity, and some microenvironmental factors) on macrophage behavior, then explores specific methods for inducing macrophage behavior through tissue engineering materials to promote myocardial repair, and summarizes the role of macrophage-host cell crosstalk in regulating inflammation, vascularization, and tissue remodeling. Finally, we propose some future challenges in regulating macrophage-material interactions and tailoring personalized biomaterials to guide macrophage phenotypes.
Collapse
Affiliation(s)
- Zitong Zhai
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Chang Yang
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Wenming Yin
- Department of Neurology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Yali Liu
- Department of Neurology, Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong 528000, China
| | - Shimin Li
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Ziyi Ye
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Mingxiang Xie
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Xiaoping Song
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
- Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
14
|
Nazari M, Taremi S, Elahi R, Mostanadi P, Esmeilzadeh A. Therapeutic Properties of M2 Macrophages in Chronic Wounds: An Innovative Area of Biomaterial-Assisted M2 Macrophage Targeted Therapy. Stem Cell Rev Rep 2025; 21:390-422. [PMID: 39556244 DOI: 10.1007/s12015-024-10806-3] [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] [Accepted: 10/16/2024] [Indexed: 11/19/2024]
Abstract
Wound healing is a dynamic, multi-stage process essential for restoring skin integrity. Dysregulated wound healing is often linked to impaired macrophage function, particularly in individuals with chronic underlying conditions. Macrophages, as key regulators of wound healing, exhibit significant phenotypic diversity, ranging from the pro-healing M2 phenotype to the pro-inflammatory M1 phenotype. Imbalances in the M1/M2 ratio or hyperactivation of the M1 phenotype can delay the normal healing. Consequently, strategies aimed at suppressing the M1 phenotype or promoting the shift of local skin macrophages toward the M2 phenotype can potentially treat chronic non-healing wounds. This manuscript provides an overview of macrophages' role in normal and pathological wound-healing processes. It examines various therapeutic approaches targeting M2 macrophages, such as ex vivo-activated macrophage therapy, immunopharmacological strategies, and biomaterial-directed macrophage polarization. However, it also highlights that M2 macrophage therapies and immunopharmacological interventions may have drawbacks, including rapid phenotypic changes, adverse effects on other skin cells, biotoxicity, and concerns related to biocompatibility, stability, and drug degradation. Therefore, there is a need for more targeted macrophage-based therapies that ensure optimal biosafety, allowing for effective reprogramming of dysregulated macrophages and improved therapeutic outcomes. Recent advances in nano-biomaterials have demonstrated promising regenerative potential compared to traditional treatments. This review discusses the progress of biomaterial-assisted macrophage targeting in chronic wound repair and addresses the challenges faced in its clinical application. Additionally, it explores novel design concepts for combinational therapies, such as incorporating regenerative particles like exosomes into dressing materials or encapsulating them in microneedling systems to enhance wound healing rates.
Collapse
Affiliation(s)
- Mahdis Nazari
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Siavash Taremi
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Reza Elahi
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Parsa Mostanadi
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Abdolreza Esmeilzadeh
- Department of Immunology, Zanjan University of Medical Sciences, Zanjan, Iran.
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
15
|
Han Y, Geng Q, Dong A, Jiang M, Ma J, Song W, Fan P, Li Y, Gao J, Zhang F, Leng J, Yuan H. Anti-Scar Effects of Micropatterned Hydrogel after Glaucoma Drainage Device Implantation. RESEARCH (WASHINGTON, D.C.) 2025; 8:0561. [PMID: 39845708 PMCID: PMC11751202 DOI: 10.34133/research.0561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/02/2024] [Accepted: 12/08/2024] [Indexed: 01/24/2025]
Abstract
Excessive fibrosis is the primary factor for the failure of glaucoma drainage device (GDD) implantation. Thus, strategies to suppress scar formation in GDD implantation are crucial. Although it is known that in implanted medical devices, microscale modification of the implant surface can modulate cell behavior and reduce the incidence of fibrosis, in the field of ophthalmic implants, especially the modification and effects of hydrogel micropatterns have rarely been reported. Here, we designed the patterned gelatin/acrylamide double network hydrogel and developed an innovative GDD with micropattern to suppress inflammatory and fibroblast activation after GDD implantation. Pattern topography suppressed F-actin expression and mitigated actin-dependent nuclear migration of myocardin-related transcription factor A (MRTF-A) during the proliferative phase after GDD implantation. Ultimately, the expression of α-smooth muscle actin (α-SMA), a key fibrosis-related gene product, was suppressed. Moreover, the modified GDD effectively controlled intraocular pressure (IOP), mitigated fibrous formation, and remodeled extracellular matrix (ECM) collagen distribution in vivo. Therefore, the novel GDD with surface patterning interventions provides a promising strategy to inhibit scar formation after GDD implantation and raise the efficacy of GDD implantation.
Collapse
Affiliation(s)
- Yiling Han
- Department of Ophthalmology, The Future Medicine Laboratory,
The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People’s Republic of China
| | - Qiangwang Geng
- Centre for Composite Materials and Structures,
Harbin Institute of Technology (HIT), Harbin 150080, People’s Republic of China
| | - Aimeng Dong
- Department of Ophthalmology, The Future Medicine Laboratory,
The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People’s Republic of China
| | - Menglu Jiang
- Department of Ophthalmology, The Future Medicine Laboratory,
The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People’s Republic of China
| | - Jingyi Ma
- Department of Ophthalmology, The Future Medicine Laboratory,
The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People’s Republic of China
| | - Wulian Song
- Department of Ophthalmology, The Future Medicine Laboratory,
The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People’s Republic of China
| | - Pan Fan
- Department of Ophthalmology, The Future Medicine Laboratory,
The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People’s Republic of China
| | - Yuanyuan Li
- Department of Ophthalmology, The Future Medicine Laboratory,
The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People’s Republic of China
| | - Jiawen Gao
- Department of Ophthalmology, The Future Medicine Laboratory,
The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People’s Republic of China
| | - Fenghua Zhang
- Centre for Composite Materials and Structures,
Harbin Institute of Technology (HIT), Harbin 150080, People’s Republic of China
| | - Jinsong Leng
- Centre for Composite Materials and Structures,
Harbin Institute of Technology (HIT), Harbin 150080, People’s Republic of China
| | - Huiping Yuan
- Department of Ophthalmology, The Future Medicine Laboratory,
The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People’s Republic of China
| |
Collapse
|
16
|
Li H, Li D, Wang X, Zeng Z, Pahlavan S, Zhang W, Wang X, Wang K. Progress in Biomaterials-Enhanced Vascularization by Modulating Physical Properties. ACS Biomater Sci Eng 2025; 11:33-54. [PMID: 39615049 DOI: 10.1021/acsbiomaterials.4c01106] [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: 01/06/2025]
Abstract
Sufficient vascular system and adequate blood perfusion is crucial for ensuring nutrient and oxygen supply within biomaterials. Actively exploring the optimal physical properties of biomaterials in various application scenarios has provided clues for enhancing vascularization within materials, leading to improved outcomes in tissue engineering and clinical translation. Here we focus on reviewing the physical properties of biomaterials, including pore structure, surface topography, and stiffness, and their effects on promoting vascularization. This angiogenic capability has the potential to provide better standardized research models and personalized treatment strategies for bone regeneration, wound healing, islet transplantation and cardiac repair.
Collapse
Affiliation(s)
- Hao Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Dayan Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Xue Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
| | - Ziyuan Zeng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
| | - Wei Zhang
- TianXinFu (Beijing) Medical Appliance Co., Ltd., Beijing 102200, China
| | - Xi Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
| | - Kai Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| |
Collapse
|
17
|
Song J, Huang S, Linghu X, Wu H, Wen Z, Li X, Huang Q, Xu W, Wa Q. 3D printing of different fibres towards HA/PCL scaffolding induces macrophage polarization and promotes osteogenic differentiation of BMSCs. PLoS One 2025; 20:e0314150. [PMID: 39804888 PMCID: PMC11729943 DOI: 10.1371/journal.pone.0314150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/05/2024] [Indexed: 01/16/2025] Open
Abstract
With the rise of bone tissue engineering (BET), 3D-printed HA/PCL scaffolds for bone defect repair have been extensively studied. However, little research has been conducted on the differences in osteogenic induction and regulation of macrophage (MPs) polarisation properties of HA/PCL scaffolds with different fibre orientations. Here, we applied 3D printing technology to prepare three sets of HA/PCL scaffolds with different fibre orientations (0-90, 0-90-135, and 0-90-45) to study the differences in physicochemical properties and to investigate the response effects of MPs and bone marrow mesenchymal stem cells (BMSCs) on scaffolds with different fibre orientations. The results showed that multi-angle staggered fibres affected the overall porosity and compressive strength of the scaffolds. Compared with the other two groups, the 0-90-45 scaffold induced osteogenic differentiation of BMSCs more significantly, while promoting the polarisation of MPs towards the M2 phenotype to form an osteogenic-friendly immune microenvironment. Unexpectedly, the 0-90-45 scaffold significantly upregulated the expression of angiogenic genes (PDGF, VEGF). Therefore, we conclude that the multi-angle interlaced fibres better mimic the physiological structure of cancellous bone, and that the excellent biomimetic properties reflect the best in vitro osteogenic, immunomodulatory and angiogenic effects. In conclusion, this study is a step forward in the exploration of BET scaffolds and provides a very promising bone filling material.
Collapse
Affiliation(s)
- Jiaxiang Song
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Honghuagang District, Guizhou, China
| | - Shuai Huang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Haizhu District, Guangdong, China
| | - Xitao Linghu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Honghuagang District, Guizhou, China
| | - Hengpeng Wu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Honghuagang District, Guizhou, China
- Institute of biological and Medical Engineering, Guangdong Academy of Sciences, Haizhu District, Guangzhou, Guangdong, China
| | - Zhenyu Wen
- Zunyi Medical University, Xinpu New District, Zunyi City, Guizhou, China
| | - Xiang Li
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Honghuagang District, Guizhou, China
| | - Qiping Huang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Honghuagang District, Guizhou, China
| | - Weikang Xu
- Institute of biological and Medical Engineering, Guangdong Academy of Sciences, Haizhu District, Guangzhou, Guangdong, China
- Guangdong Institute of Medical Instruments, National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Tianhe District, Guangzhou, Guangdong, China
| | - Qingde Wa
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Honghuagang District, Guizhou, China
| |
Collapse
|
18
|
Sovar A, Patrick MD, Annamalai RT. Substrate curvature influences cytoskeletal rearrangement and modulates macrophage phenotype. Front Immunol 2025; 15:1478464. [PMID: 39835126 PMCID: PMC11743265 DOI: 10.3389/fimmu.2024.1478464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction Inflammation is a vital immune response, tightly orchestrated through both biochemical and biophysical cues. Dysregulated inflammation contributes to chronic diseases, highlighting the need for novel therapies that modulate immune responses with minimal side effects. While several biochemical pathways of inflammation are well understood, the influence of physical properties such as substrate curvature on immune cell behavior remains underexplored. This study investigates how substrate curvature impacts macrophage cytoskeletal dynamics, gene expression, and immunophenotype through mechanosensitive pathways. Methods Gelatin-based microgels with tunable surface curvatures were fabricated via water-in-oil emulsification and crosslinked with genipin. Microgels were sorted into three size ranges, yielding high (40-50 µm), intermediate (150-250 µm), and low (350-400 µm) curvature profiles. Macrophages were seeded onto these microgels, and cytoskeletal dynamics were examined using confocal microscopy, SEM, and actin-specific staining. Gene expression of pro- and anti-inflammatory markers was quantified using qPCR. The role of actin polymerization was assessed using Latrunculin-A (Lat-A) treatment. Results Macrophages adhered effectively to both high- and low-curvature microgels, displaying curvature-dependent morphological changes. Confocal imaging revealed that macrophages on low-curvature microgels exhibited significantly higher F-actin density than those on high-curvature microgels. Correspondingly, qPCR analysis showed upregulation of pro-inflammatory markers (e.g., Tnf, Nos2) in high-curvature conditions, while anti-inflammatory markers (e.g., Arg1) were elevated in low-curvature conditions. Lat-A treatment reduced F-actin density and modulated gene expression patterns, confirming the cytoskeletal regulation of macrophage phenotype. Discussion These findings demonstrate that substrate curvature influences macrophage behavior by modulating cytoskeletal dynamics and associated immunophenotypic markers through actin-mediated transcriptional pathways. By controlling curvature, therapeutic biomaterials may direct immune responses, offering a new avenue for treating inflammatory diseases. This mechanobiological approach presents a promising strategy for precision immunomodulation in regenerative medicine.
Collapse
Affiliation(s)
- Austin Sovar
- Department of Biomedical Engineering, University of Kentucky, Lexington, KY, United States
| | - Matthew D. Patrick
- Department of Biomedical Engineering, University at Buffalo, Buffalo, NY, United States
| | - Ramkumar T. Annamalai
- Department of Biomedical Engineering, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
19
|
Zhang Y, Rao Y, Lu J, Wang J, Ker DFE, Zhou J, Wang DM. The influence of biophysical niche on tumor-associated macrophages in liver cancer. Hepatol Commun 2024; 8:e0569. [PMID: 39470328 PMCID: PMC11524744 DOI: 10.1097/hc9.0000000000000569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 08/30/2024] [Indexed: 10/30/2024] Open
Abstract
HCC, the most common type of primary liver cancer, is a leading cause of cancer-related mortality worldwide. Although the advancement of immunotherapies by immune checkpoint inhibitors (ICIs) that target programmed cell death 1 or programmed cell death 1-ligand 1 has revolutionized the treatment for HCC, the majority is still not beneficial. Accumulating evidence has pointed out that the potent immunosuppressive tumor microenvironment in HCC poses a great challenge to ICI therapeutic efficacy. As a key component in tumor microenvironment, tumor-associated macrophages (TAMs) play vital roles in HCC development, progression, and ICI low responsiveness. Mechanistically, TAM can promote cancer invasion and metastasis, angiogenesis, epithelial-mesenchymal transition, maintenance of stemness, and most importantly, immunosuppression. Targeting TAMs, therefore, represents an opportunity to enhance the ICI therapeutic efficacy in patients with HCC. While previous research has primarily focused on biochemical cues influencing macrophages, emerging evidence highlights the critical role of biophysical signals, such as substrate stiffness, topography, and external forces. In this review, we summarize the influence of biophysical characteristics within the tumor microenvironment that regulate the phenotype and function of TAMs in HCC pathogenesis and progression. We also explore the possible mechanisms and discuss the potential of manipulating biophysical cues in regulating TAM for HCC therapy. By gaining a deeper understanding of how macrophages sense and respond to mechanical forces, we may potentially usher in a path toward a curative approach for combinatory cancer immunotherapies.
Collapse
Affiliation(s)
- Ying Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Ying Rao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Jiahuan Lu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Jiyu Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Dai Fei Elmer Ker
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Sha Tin, Hong Kong, SAR, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, SAR, China
| | - Jingying Zhou
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Dan Michelle Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Sha Tin, Hong Kong, SAR, China
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| |
Collapse
|
20
|
Amani H, Alipour M, Shahriari E, Taboas JM. Immunomodulatory Biomaterials: Tailoring Surface Properties to Mitigate Foreign Body Reaction and Enhance Tissue Regeneration. Adv Healthc Mater 2024:e2401253. [PMID: 39370571 DOI: 10.1002/adhm.202401253] [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: 04/04/2024] [Revised: 08/28/2024] [Indexed: 10/08/2024]
Abstract
The immune cells have demonstrated the ability to promote tissue repair by removing debris, breaking down the extracellular matrix, and regulating cytokine secretion profile. If the behavior of immune cells is not well directed, chronic inflammation and foreign body reaction (FBR) will lead to scar formation and loss of biomaterial functionality. The immunologic response toward tissue repair or chronic inflammation after injury and implantation can be modulated by manipulating the surface properties of biomaterials. Tailoring surface properties of biomaterials enables the regulation of immune cell fate such as adhesion, proliferation, recruitment, polarization, and cytokine secretion profile. This review begins with an overview of the role of immune cells in tissue healing and their interactions with biomaterials. It then discusses how the surface properties of biomaterials influence immune cell behavior. The core focus is reviewing surface modification methods to create innovative materials that reduce foreign body reactions and enhance tissue repair and regeneration by modulating immune cell activities. The review concludes with insights into future advancements in surface modification techniques and the associated challenges.
Collapse
Affiliation(s)
- Hamed Amani
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Science, Tehran, Iran
| | - Mahdieh Alipour
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Elahe Shahriari
- Department of Physiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Juan M Taboas
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| |
Collapse
|
21
|
Alemifar A, Burnette K, Jandres B, Hurt S, Tse HM, Robinson JL. Electrospun Fiber Surface Roughness Modulates Human Monocyte-Derived Macrophage Phenotype. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.30.610568. [PMID: 39282362 PMCID: PMC11398424 DOI: 10.1101/2024.08.30.610568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Injuries to fibrous connective tissues have very little capacity for self-renewal and exhibit poor healing after injury. Phenotypic shifts in macrophages play a vital role in mediating the healing response, creating an opportunity to design immunomodulatory biomaterials which control macrophage polarization and promote regeneration. In this study, electrospun poly(-caprolactone) fibers with increasing surface roughness (SR) were produced by increasing relative humidity and inducing vapor-induced phase separation during the electrospinning process. The impact of surface roughness on macrophage phenotype was assessed using human monocyte-derived macrophages in vitro and in vivo using B6.Cg-Tg(Csf1r-EGFP)1Hume/J (MacGreen) mice. In vitro experiments showed that macrophages cultured on mesh with increasing SR exhibited decreased release of both pro- and anti-inflammatory cytokines potentially driven by increased protein adsorption and biophysical impacts on the cells. Further, increasing SR led to an increase in the expression of the pro-regenerative cell surface marker CD206 relative to the pro-inflammatory marker CD80. Mesh with increasing SR were implanted subcutaneously in MacGreen mice, again showing an increase in the ratio of cells expressing CD206 to those expressing CD80 visualized by immunofluorescence. SR on implanted biomaterials is sufficient to drive macrophage polarization, demonstrating a simple feature to include in biomaterial design to control innate immunity.
Collapse
Affiliation(s)
- Aidan Alemifar
- Department of Orthopaedic Surgery and Sports Medicine, University of Washington
- Bioengineering Graduate Program, University of Kansas
| | - KaLia Burnette
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center
| | - Bryan Jandres
- Department of Biochemistry, University of Washington
| | - Samuel Hurt
- Department of Chemical and Petroleum Engineering, University of Kansas
| | - Hubert M Tse
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center
| | - Jennifer L Robinson
- Department of Orthopaedic Surgery and Sports Medicine, University of Washington
- Department of Mechanical Engineering, University of Washington
| |
Collapse
|
22
|
Sovar A, Patrick M, Annamalai RT. Substrate Curvature Influences Cytoskeletal Rearrangement and Modulates Macrophage Phenotype. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.12.607593. [PMID: 39211272 PMCID: PMC11361075 DOI: 10.1101/2024.08.12.607593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Inflammation serves as a critical defense mechanism against pathogens and tissue damage but can lead to chronic diseases, such as cardiovascular disease and diabetes, when dysregulated. Macrophages play a pivotal role in orchestrating inflammatory responses, transitioning from pro-inflammatory M1 to anti-inflammatory M2 phenotypes to resolve inflammation and promote tissue repair. Current approaches to modulate macrophage phenotype predominantly rely on biochemical cues, which may induce systemic side effects. Given the mechanosensitivity of macrophages, this study investigates biophysical cues, specifically substrate curvature, as a localized strategy to regulate macrophage phenotype and minimize systemic repercussions. We hypothesized that substrate curvature influences macrophage immunophenotype by modulating F-actin polymerization. To test this hypothesis, we fabricated spherical microgels with tunable curvatures and characterized their biophysical properties. Our findings indicate that macrophages adhere to microgel surfaces irrespective of curvature, but the curvature significantly alters F-actin dynamics. Furthermore, manipulating cytoskeletal dynamics via selective actin inhibition partially reversed curvature-induced changes in macrophage phenotype. These results underscore the pivotal role of substrate curvature in modulating macrophage behavior and immunophenotype. Overall, our study demonstrates that substrate curvature significantly influences macrophage cytoskeletal dynamics and resulting immunophenotype. This simple approach can be utilized as a localized immunomodulatory treatment for inflammatory diseases.
Collapse
|
23
|
del Carmen Morán M, Cirisano F, Ferrari M. Superhydrophobicity Effects on Spheroid Formation and Polarization of Macrophages. Pharmaceuticals (Basel) 2024; 17:1042. [PMID: 39204146 PMCID: PMC11357281 DOI: 10.3390/ph17081042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/27/2024] [Accepted: 08/01/2024] [Indexed: 09/03/2024] Open
Abstract
The interaction of biomaterials with the immune system is ruled by the action of macrophages. The surface features of these biomaterials, like wettability, which is an expression of chemical composition, texture, and geometry, can affect macrophages response. Such surface parameters can be then efficiently exploited to improve biocompatibility by lowering undesired immunological reactions and at the same time creating the substrate for positive interactions. In this work, the preparation and physicochemical characterization of highly water-repellent surfaces to develop and characterize 3D spheroids derived from monocyte-macrophages (RAW 264.7 cell line) has been carried out. As a measure of cell viability over time, the obtained aggregates have been transferred under standard 2D cell culture conditions. Significant changes on the morphology-associated polarization of the derived cellular entities have been evaluated at the nanoscale through 3D profilometry. The results suggested that the spheroid formation using highly repellent substrates induced the activation of M2-type cells. This simple and cost-effective approach can be used for preparing M2-based macrophages for regenerative purposes.
Collapse
Affiliation(s)
- María del Carmen Morán
- Departament de Bioquímica i Fisiologia, Secció de Fisiologia—Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, Avda. Joan XXIII, 27-31, 08028 Barcelona, Spain
- Institut de Nanociència i Nanotecnologia—IN2UB, Universitat de Barcelona, Avda. Diagonal, 645, 08028 Barcelona, Spain
| | - Francesca Cirisano
- CNR-ICMATE Istituto di Chimica della Materia Condensata e di Tecnologie per l’Energia, Via De Marini, 6, 16149 Genova, Italy;
| | - Michele Ferrari
- Institut de Nanociència i Nanotecnologia—IN2UB, Universitat de Barcelona, Avda. Diagonal, 645, 08028 Barcelona, Spain
- CNR-ICMATE Istituto di Chimica della Materia Condensata e di Tecnologie per l’Energia, Via De Marini, 6, 16149 Genova, Italy;
| |
Collapse
|
24
|
Yang Y, He X, Zhao Z, Yi J. Macrophage-Centric Biomaterials for Bone Regeneration in Diabetes Mellitus: Contemporary Advancements, Challenges, and Future Trajectories. Cureus 2024; 16:e66621. [PMID: 39258053 PMCID: PMC11386247 DOI: 10.7759/cureus.66621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2024] [Indexed: 09/12/2024] Open
Abstract
Increased susceptibility to bone fragility and the diminution of bone regenerative capacity are recognized as significant and frequent sequelae of diabetes mellitus. Research has elucidated the pivotal role of macrophages in the pathogenesis and repair of diabetic bone defects. Notwithstanding this, the therapeutic efficacy of traditional interventions remains predominantly inadequate. Concomitant with substantial advancements in tissue engineering in recent epochs, there has been an escalation in the development of biomaterials designed to modulate macrophage activity, thereby augmenting osseous tissue regeneration in the context of hyperglycemia. This review amalgamates insights from extant research and delineates recent progressions in the domain of biomaterials that target macrophages for the regeneration of diabetic bone, whilst also addressing the clinical challenges and envisaging future directions within this field.
Collapse
Affiliation(s)
- Yiyan Yang
- Department of Orthodontics, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, CHN
| | - Xiaoli He
- Department of Orthodontics, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, CHN
| | - Zhihe Zhao
- Department of Orthodontics, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, CHN
| | - Jianru Yi
- Department of Orthodontics, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, CHN
| |
Collapse
|
25
|
Ao J, Zhang X, You Y, Chen Y, Liu Z, Gao J, Qin C, Hao L, Zhao J, Jiang R. Bioinspired Hybrid Nanostructured PEEK Implant with Enhanced Antibacterial and Anti-inflammatory Synergy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:38989-39004. [PMID: 39034661 DOI: 10.1021/acsami.4c06322] [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: 07/23/2024]
Abstract
Implant-associated infections and excessive immune responses are two major postsurgical issues for successful implantation. However, conventional strategies including antibiotic treatment and inflammatory regulation are always compromised due to the comodification of various biochemical agents and instances of functional interference. It is imperative to provide implant surfaces with satisfactory antibacterial and anti-inflammatory properties. Here, a dual-effect nanostructured polyetheretherketone (PEEK) surface (NP@PDA/Zn) with bionic mechano-bactericidal nanopillars and immobilized immunomodulatory Zn2+ is designed. The constructed hybrid nanopillars display remarkable antibacterial performance against Gram-negative and Gram-positive strains through the synergy of physical and chemical bactericidal effects imposed by nanopillars and Zn2+. Meanwhile, the immunoregulatory property is evaluated through the investigation of macrophage polarization both in vitro and in vivo, and the results reveal that NP@PDA/Zn could downregulate the expression of M1-related cytokines and decrease the M1 macrophage recruitment to lower the inflammatory response. Notably, the surface exhibited exceptional biocompatibility with discerning biocidal activity between bacterial and mammalian cells and antioxidant performance that effectively scavenges ROS, minimizing potential cytotoxicity. Taken together, NP@PDA/Zn presents a convenient and promising strategy of combining synergistic bactericidal activity and inflammatory regulation without any mutual interference, which can support the development of multifunctional implant-associated materials.
Collapse
Affiliation(s)
- Ji Ao
- Key Laboratory of Bionic Engineering, Ministry of Education, Jilin University, Changchun 130022, China
| | - Xin Zhang
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
- College of Chemistry, Jilin University, Changchun 130022, China
| | - Yunhao You
- Department of Spine Surgery, Qilu Hospital of Shandong University, Jinan 250012 China
| | - Yuxiang Chen
- Key Laboratory of Bionic Engineering, Ministry of Education, Jilin University, Changchun 130022, China
- Department of Mechanical Engineering, The University of Hong Kong, SAR, Hong Kong 999077, China
| | - Zequan Liu
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Jie Gao
- Key Laboratory of Bionic Engineering, Ministry of Education, Jilin University, Changchun 130022, China
| | - Chenyang Qin
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Lingwan Hao
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Jie Zhao
- Key Laboratory of Bionic Engineering, Ministry of Education, Jilin University, Changchun 130022, China
| | - Rujian Jiang
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| |
Collapse
|
26
|
Cao X, Luo B, Mu Y, Wang C, Lu R, Yao Y, Chen S. The regulatory effect of TiO 2 nanotubes loaded with graphene oxide on macrophage polarization in an inflammatory environment. BMC Oral Health 2024; 24:824. [PMID: 39033148 PMCID: PMC11265100 DOI: 10.1186/s12903-024-04608-9] [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: 01/31/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Excessive inflammation is a major cause of implant failure. The surface morphology, hydrophilicity, and loading of biomaterials are major properties modulating anti-inflammatory macrophage activation. This paper investigates the regulatory effects of modifying the surface of Titanium dioxide nanotubes (TNTs) with graphene oxide (GO) on the polarization of mouse monocyte macrophages (RAW264.7). METHODS TNT was produced by the anodic oxidation of titanium. GO was subsequently electrodeposited on the TNT to obtain a TNT-GO composite. The samples were characterised through scanning electron microscopy (SEM), Raman spectroscopy, and X-ray diffraction. RAW264.7 cells were separately seeded onto the surface of three groups of samples: pure Ti, TNT, and TNT-GO. Under the condition of lipopolysaccharide stimulation, the influence of the sample surfaces on the gene expression profiles was investigated through RNA sequence analysis. In addition, cell spreading was observed through SEM, cell adhesion and proliferation were analysed using the CCK8 assay, and the expression of inflammation-related factors was investigated by ELISA and cellular immunofluorescence staining. The production of reactive oxygen species (ROS) in the RAW264.7 cells on the surface of the three groups was detected via immunofluorescence staining. RESULTS The CCK8 results indicated that the adhesion and proliferation of the RAW264.7 cells were reduced on the TNT and TNT-GO surfaces. ELISA results revealed significant differences in the pro-inflammatory factors tumour necrosis factor-α and interleukin-6 secretion among the three groups at 24 h (p < 0.05). The secretion of pro-inflammatory factors significantly reduced and the expression of anti-inflammatory factor IL-10 increased on the TNT and TNT-GO surfaces. The RNA sequencing, ELISA, and cell immunofluorescence staining test results suggested that the inflammatory response of M1 polarization was reduced and the M2 polarization of macrophages was induced on the TNT-GO surface, which may be attributed to the reduction in ROS production. CONCLUSIONS Under lipopolysaccharide stimulation, the inflammatory response of the RAW264.7 cells was reduced and the M2 polarization of macrophages was promoted on the TNT-GO surface, which may be caused by the reduced ROS production. Consequently, the designed TNT-GO material is promising for implants owing to its excellent inflammation regulation ability.
Collapse
Affiliation(s)
- Xu Cao
- Laboratory of Biomaterials and Biomechanics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Bin Luo
- Laboratory of Biomaterials and Biomechanics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Yanting Mu
- Laboratory of Biomaterials and Biomechanics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Caiyun Wang
- Laboratory of Biomaterials and Biomechanics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Ran Lu
- Laboratory of Biomaterials and Biomechanics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Yao Yao
- Laboratory of Biomaterials and Biomechanics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Su Chen
- Laboratory of Biomaterials and Biomechanics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
27
|
Du Q, Dickinson A, Nakuleswaran P, Maghami S, Alagoda S, Hook AL, Ghaemmaghami AM. Targeting Macrophage Polarization for Reinstating Homeostasis following Tissue Damage. Int J Mol Sci 2024; 25:7278. [PMID: 39000385 PMCID: PMC11242417 DOI: 10.3390/ijms25137278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Tissue regeneration and remodeling involve many complex stages. Macrophages are critical in maintaining micro-environmental homeostasis by regulating inflammation and orchestrating wound healing. They display high plasticity in response to various stimuli, showing a spectrum of functional phenotypes that vary from M1 (pro-inflammatory) to M2 (anti-inflammatory) macrophages. While transient inflammation is an essential trigger for tissue healing following an injury, sustained inflammation (e.g., in foreign body response to implants, diabetes or inflammatory diseases) can hinder tissue healing and cause tissue damage. Modulating macrophage polarization has emerged as an effective strategy for enhancing immune-mediated tissue regeneration and promoting better integration of implantable materials in the host. This article provides an overview of macrophages' functional properties followed by discussing different strategies for modulating macrophage polarization. Advances in the use of synthetic and natural biomaterials to fabricate immune-modulatory materials are highlighted. This reveals that the development and clinical application of more effective immunomodulatory systems targeting macrophage polarization under pathological conditions will be driven by a detailed understanding of the factors that regulate macrophage polarization and biological function in order to optimize existing methods and generate novel strategies to control cell phenotype.
Collapse
Affiliation(s)
- Qiran Du
- Immuno-Bioengineering Group, School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Anna Dickinson
- Medical School, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (A.D.); (P.N.); (S.A.)
| | - Pruthvi Nakuleswaran
- Medical School, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (A.D.); (P.N.); (S.A.)
| | - Susan Maghami
- Hull York Medical School, University of York, York YO10 5DD, UK;
| | - Savindu Alagoda
- Medical School, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (A.D.); (P.N.); (S.A.)
| | - Andrew L. Hook
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Amir M. Ghaemmaghami
- Immuno-Bioengineering Group, School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK;
| |
Collapse
|
28
|
ten Brink T, Damanik F, Rotmans JI, Moroni L. Unraveling and Harnessing the Immune Response at the Cell-Biomaterial Interface for Tissue Engineering Purposes. Adv Healthc Mater 2024; 13:e2301939. [PMID: 38217464 PMCID: PMC11468937 DOI: 10.1002/adhm.202301939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 12/14/2023] [Indexed: 01/15/2024]
Abstract
Biomaterials are defined as "engineered materials" and include a range of natural and synthetic products, designed for their introduction into and interaction with living tissues. Biomaterials are considered prominent tools in regenerative medicine that support the restoration of tissue defects and retain physiologic functionality. Although commonly used in the medical field, these constructs are inherently foreign toward the host and induce an immune response at the material-tissue interface, defined as the foreign body response (FBR). A strong connection between the foreign body response and tissue regeneration is suggested, in which an appropriate amount of immune response and macrophage polarization is necessary to trigger autologous tissue formation. Recent developments in this field have led to the characterization of immunomodulatory traits that optimizes bioactivity, the integration of biomaterials and determines the fate of tissue regeneration. This review addresses a variety of aspects that are involved in steering the inflammatory response, including immune cell interactions, physical characteristics, biochemical cues, and metabolomics. Harnessing the advancing knowledge of the FBR allows for the optimization of biomaterial-based implants, aiming to prevent damage of the implant, improve natural regeneration, and provide the tools for an efficient and successful in vivo implantation.
Collapse
Affiliation(s)
- Tim ten Brink
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| | - Febriyani Damanik
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| | - Joris I. Rotmans
- Department of Internal MedicineLeiden University Medical CenterAlbinusdreef 2Leiden2333ZAThe Netherlands
| | - Lorenzo Moroni
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| |
Collapse
|
29
|
He Z, Liu Y, Zheng ZL, Lv JC, Liu SB, Zhang J, Liu HH, Xu JZ, Li ZM, Luo E. Periodic Lamellae-Based Nanofibers for Precise Immunomodulation to Treat Inflammatory Bone Loss in Periodontitis. Adv Healthc Mater 2024; 13:e2303549. [PMID: 38333940 DOI: 10.1002/adhm.202303549] [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: 10/16/2023] [Revised: 01/30/2024] [Indexed: 02/10/2024]
Abstract
Periodontitis is a common oral disease accompanied by inflammatory bone loss. The pathological characteristics of periodontitis usually accompany an imbalance in the periodontal immune microenvironment, leading to difficulty in bone regeneration. Therefore, effective treatment strategies are needed to modulate the immune environment in order to treat periodontitis. Here, highly-oriented periodic lamellae poly(ε-caprolactone) electrospun nanofibers (PLN) are developed by surface-directed epitaxial crystallization. The in vitro result shows that the PLN can precisely modulate macrophage polarization toward the M2 phenotype. Macrophages polarized by PLN significantly enhance the migration and osteogenic differentiation of Bone marrow stromal cells. Notably, results suggest that the topographical cues presented by PLN can modulate macrophage polarization by activating YAP, which reciprocally inhibits the NF-κB signaling pathway. The in vivo results indicate that PLN can inhibit inflammatory bone loss and facilitate bone regeneration in periodontitis. The authors' findings suggest that topographical nanofibers with periodic lamellae is a promising strategy for modulating immune environment to treat inflammatory bone loss in periodontitis.
Collapse
Affiliation(s)
- Ze He
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yao Liu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Zi-Li Zheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, P. R. China
| | - Jia-Cheng Lv
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, P. R. China
| | - Shi-Bo Liu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Ju Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Hang-Hang Liu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jia-Zhuang Xu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, P. R. China
| | - Zhong-Ming Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, P. R. China
| | - En Luo
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
30
|
Yan Y, Zhang Y, Li K, Li Y, Qian W, Zhang W, Wang Y, Ma W, Li L. Synergistic effects of graphene microgrooves and electrical stimulation on M2 macrophage polarization. Biochem Biophys Res Commun 2024; 711:149911. [PMID: 38603832 DOI: 10.1016/j.bbrc.2024.149911] [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: 01/30/2024] [Revised: 03/24/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024]
Abstract
Macrophages play a crucial role in host response and wound healing, with M2 polarization contributing to the reduction of foreign-body reactions induced by the implantation of biomaterials and promoting tissue regeneration. Electrical stimulation (ES) and micropatterned substrates have a significant impact on the macrophage polarization. However, there is currently a lack of well-established cell culture platforms for studying the synergistic effects of these two factors. In this study, we prepared a graphene free-standing substrate with 20 μm microgrooves using capillary forces induced by water evaporation. Subsequently, we established an ES cell culture platform for macrophage cultivation by integrating a self-designed multi-well chamber cell culture device. We observed that graphene microgrooves, in combination with ES, significantly reduce cell spreading area and circularity. Results from immunofluorescence, ELISA, and flow cytometry demonstrate that the synergistic effect of graphene microgrooves and ES effectively promotes macrophage M2 phenotypic polarization. Finally, RNA sequencing results reveal that the synergistic effects of ES and graphene microgrooves inhibit the macrophage actin polymerization and the downstream PI3K signaling pathway, thereby influencing the phenotypic transition. Our results demonstrate the potential of graphene-based microgrooves and ES to synergistically modulate macrophage polarization, offering promising applications in regenerative medicine.
Collapse
Affiliation(s)
- Yan Yan
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, 300041, PR China
| | - Yilin Zhang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
| | - Kexin Li
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
| | - Yudong Li
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
| | - Wei Qian
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
| | - Wenbo Zhang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
| | - Yue Wang
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, 300041, PR China
| | - Wensheng Ma
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, 300041, PR China.
| | - Linhao Li
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China.
| |
Collapse
|
31
|
Nordberg RC, Bielajew BJ, Takahashi T, Dai S, Hu JC, Athanasiou KA. Recent advancements in cartilage tissue engineering innovation and translation. Nat Rev Rheumatol 2024; 20:323-346. [PMID: 38740860 PMCID: PMC11524031 DOI: 10.1038/s41584-024-01118-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2024] [Indexed: 05/16/2024]
Abstract
Articular cartilage was expected to be one of the first successfully engineered tissues, but today, cartilage repair products are few and they exhibit considerable limitations. For example, of the cell-based products that are available globally, only one is marketed for non-knee indications, none are indicated for severe osteoarthritis or rheumatoid arthritis, and only one is approved for marketing in the USA. However, advances in cartilage tissue engineering might now finally lead to the development of new cartilage repair products. To understand the potential in this field, it helps to consider the current landscape of tissue-engineered products for articular cartilage repair and particularly cell-based therapies. Advances relating to cell sources, bioactive stimuli and scaffold or scaffold-free approaches should now contribute to progress in therapeutic development. Engineering for an inflammatory environment is required because of the need for implants to withstand immune challenge within joints affected by osteoarthritis or rheumatoid arthritis. Bringing additional cartilage repair products to the market will require an understanding of the translational vector for their commercialization. Advances thus far can facilitate the future translation of engineered cartilage products to benefit the millions of patients who suffer from cartilage injuries and arthritides.
Collapse
Affiliation(s)
- Rachel C Nordberg
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Benjamin J Bielajew
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Takumi Takahashi
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Shuyan Dai
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Jerry C Hu
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Kyriacos A Athanasiou
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
32
|
Asadi Tokmedash M, Min J. Designer Micro-/Nanocrumpled MXene Multilayer Coatings Accelerate Osteogenesis and Regulate Macrophage Polarization. ACS APPLIED MATERIALS & INTERFACES 2024; 16:21415-21426. [PMID: 38445580 DOI: 10.1021/acsami.3c18158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
Effective tissue regeneration and immune responses are essential for the success of biomaterial implantation. Although the interaction between synthetic materials and biological systems is well-recognized, the role of surface topographical cues in regulating the local osteoimmune microenvironment─specifically, their impact on host tissue and immune cells, and their dynamic interactions─remains underexplored. This study addresses this gap by investigating the impact of surface topography on osteogenesis and immunomodulation. We fabricated MXene/hydroxyapatite (HAP)-coated surfaces with controlled 2.5D nano-, submicro-, and microscale topographical patterns using our custom bottom-up patterning method. These engineered surfaces were employed to assess the behavior of osteoblast precursor cells and macrophage polarization. Our results demonstrate that MXene/HAP-coated surfaces with microscale crumpled topography significantly influence osteogenic activity and macrophage polarization: these surfaces notably enhanced osteoblast precursor cell spreading, proliferation, and differentiation and facilitated a shift in macrophages toward an anti-inflammatory, prohealing M2 phenotype. The observed cell responses indicate that the physical cues from the crumpled topographies, combined with the chemical cues from the MXene/HAP coatings, synergistically create a favorable osteoimmune microenvironment. This study presents the first evidence of employing MXene/HAP-multilayer coated surfaces with finely crumpled topography to concurrently facilitate osteogenesis and immunomodulation for improved implant-to-tissue integration. The tunable topographic patterns of these coatings coupled with a facile and scalable fabrication process make them widely applicable for various biomedical purposes. Our results highlight the potential of these multilayer coatings with controlled topography to improve the in vivo performance and fate of implants by modulating the host response at the material interface.
Collapse
Affiliation(s)
- Mohammad Asadi Tokmedash
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jouha Min
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Macromolecular Science and Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
- Weil Institute for Critical Care Research and Innovation, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
33
|
Wang J, Zhang L, Wang L, Tang J, Wang W, Xu Y, Li Z, Ding Z, Jiang X, Xi K, Chen L, Gu Y. Ligand-Selective Targeting of Macrophage Hydrogel Elicits Bone Immune-Stem Cell Endogenous Self-Healing Program to Promote Bone Regeneration. Adv Healthc Mater 2024; 13:e2303851. [PMID: 38226706 PMCID: PMC11468030 DOI: 10.1002/adhm.202303851] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 01/05/2024] [Indexed: 01/17/2024]
Abstract
Targeting macrophages can facilitate the site-specific repair of critical bone defects. Herein, a composite hydrogel, gelatin-Bletilla striata polysaccharide-mesoporous bioactive glass hydrogel (GBMgel), is constructed via the self-assembly of mesoporous bioactive glass on polysaccharide structures, through the Schiff base reaction. GBMgel can efficiently capture macrophages and drive the recruitment of seed stem cells and vascular budding required for regeneration in the early stages of bone injury, and the observed sustained release of inorganic silicon ions further enhances bone matrix deposition, mineralization, and vascular maturation. Moreover, the use of macrophage-depleted rat calvarial defect models further confirms that GBMgel, with ligand-selective macrophage targeting, increases the bone regeneration area and the proportion of mature bone. Mechanistic studies reveal that GBMgel upregulates the TLR4/NF-κB and MAPK macrophage pathways in the early stages and the JAK/STAT3 pathway in the later stages; thus initiating macrophage polarization at different time points. In conclusion, this study is based on the endogenous self-healing properties of bone macrophages, which enhances stem cell homing, and provides a research and theoretical basis upon which bone tissue can be reshaped and regenerated using the body's immune power, providing a new strategy for the treatment of critical bone defects.
Collapse
Affiliation(s)
- Jiahao Wang
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| | - Liang Zhang
- Department of OrthopedicsBeijing Friendship HospitalCapital Medical UniversityNo. 95, Yong An Road, XiCheng DistrictBeijing100050P. R. China
| | - Lingjun Wang
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| | - Jincheng Tang
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| | - Wei Wang
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| | - Yichang Xu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| | - Ziang Li
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| | - Zhouye Ding
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| | - Xinzhao Jiang
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| | - Kun Xi
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| | - Liang Chen
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| | - Yong Gu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| |
Collapse
|
34
|
Xu Y, Ying L, Lang JK, Hinz B, Zhao R. Modeling mechanical activation of macrophages during pulmonary fibrogenesis for targeted anti-fibrosis therapy. SCIENCE ADVANCES 2024; 10:eadj9559. [PMID: 38552026 PMCID: PMC10980276 DOI: 10.1126/sciadv.adj9559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 02/23/2024] [Indexed: 04/01/2024]
Abstract
Pulmonary fibrosis is an often fatal lung disease. Immune cells such as macrophages were shown to accumulate in the fibrotic lung, but their contribution to the fibrosis development is unclear. To recapitulate the involvement of macrophages in the development of pulmonary fibrosis, we developed a fibrotic microtissue model with cocultured human macrophages and fibroblasts. We show that profibrotic macrophages seeded on topographically controlled stromal tissues became mechanically activated. The resulting co-alignment of macrophages, collagen fibers, and fibroblasts promoted widespread fibrogenesis in micro-engineered lung tissues. Anti-fibrosis treatment using pirfenidone disrupts the polarization and mechanical activation of profibrotic macrophages, leading to fibrosis inhibition. Pirfenidone inhibits the mechanical activation of macrophages by suppressing integrin αMβ2 and Rho-associated kinase 2. These results demonstrate a potential pulmonary fibrogenesis mechanism at the tissue level contributed by macrophages. The cocultured microtissue model is a powerful tool to study the immune-stromal cell interactions and the anti-fibrosis drug mechanism.
Collapse
Affiliation(s)
- Ying Xu
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Linxuan Ying
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Jennifer K. Lang
- Division of Cardiovascular Medicine and the Clinical and Translational Research Center, University at Buffalo, State University of New York; Veterans Affairs Western New York Health Care System, University at Buffalo, State University of New York; Department of Biomedical Engineering, University at Buffalo, State University of New York; Department of Medicine, University at Buffalo, State University of New York; Department of Pharmacology and Toxicology, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Ruogang Zhao
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| |
Collapse
|
35
|
Huang W, Cai X, Xiao C, Song W, Yin H, Xu W. Surface micropatterning of 3D printed PCL scaffolds promotes osteogenic differentiation of BMSCs and regulates macrophage M2 polarization. Heliyon 2024; 10:e26621. [PMID: 38434344 PMCID: PMC10907665 DOI: 10.1016/j.heliyon.2024.e26621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 03/05/2024] Open
Abstract
Micropatterned structures on the surface of materials possessing biomimetic properties to mimic the extracellular matrix and induce cellular behaviors have been widely studied. However, it is still a major challenge to obtain internally stable and controllable micropatterned 3D scaffolds for bone repair and regeneration. In this study, 3D scaffolds with regular grating arrays using polycaprolactone (PCL) as a matrix material were prepared by combining 3D printing and soft lithography, and the effects of grating micropatterning on osteogenic differentiation of BMSCs and M1/M2 polarization of macrophages were investigated. The results showed that compared with the planar group and the 30um grating spacing group, PCL with a grating spacing of 20um significantly promoted the osteogenic differentiation of BMSCs, induced the polarization of RAW264.7 cells toward M2 type, and suppressed the expression of M1-type pro-inflammatory genes and markers. In conclusion, we successfully constructed PCL-based three-dimensional scaffolds with stable and controllable micrographs (grating arrays) inside, which possess excellent osteogenic properties and promote the formation of an immune microenvironment conducive to osteogenesis. This study is a step forward to the exploration of bone-filling materials affecting cell behavior, and makes a new contribution to the provision of high-quality materials.
Collapse
Affiliation(s)
- Weihua Huang
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong, 510316, China
- Department of Orthopaedic Surgery, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, No.35, Yinquan North Road, Qingcheng District, Qingyuan, Guangdong, 511518, China
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangdong Institute of Medical Instruments, No. 1307 Guangzhou Avenue Central, Tianhe District, Guangzhou, Guangdong, 510500, China
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Guangzhou Medical University, The Second Clinical Medicine School of Guangzhou Medical University, No. 250, Changgang East Road, Haizhu District, Guangzhou, Guangdong, 510260, China
| | - Xiayu Cai
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong, 510316, China
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangdong Institute of Medical Instruments, No. 1307 Guangzhou Avenue Central, Tianhe District, Guangzhou, Guangdong, 510500, China
| | - Chujie Xiao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong, 510316, China
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangdong Institute of Medical Instruments, No. 1307 Guangzhou Avenue Central, Tianhe District, Guangzhou, Guangdong, 510500, China
- National Engineering Research Center for Human Tissue Restoration and Function Reconstruction, South China University of Technology, Wushan Road 381, Guangzhou, Guangdong, 510275, China
| | - Wenlu Song
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong, 510316, China
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangdong Institute of Medical Instruments, No. 1307 Guangzhou Avenue Central, Tianhe District, Guangzhou, Guangdong, 510500, China
- Sun Yat-Sen University, Xingang West Road 135, Guangzhou, Guangdong, 510006, China
| | - Huinan Yin
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Guangzhou Medical University, The Second Clinical Medicine School of Guangzhou Medical University, No. 250, Changgang East Road, Haizhu District, Guangzhou, Guangdong, 510260, China
| | - Weikang Xu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong, 510316, China
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangdong Institute of Medical Instruments, No. 1307 Guangzhou Avenue Central, Tianhe District, Guangzhou, Guangdong, 510500, China
- Guangdong Chinese Medicine Intelligent Diagnosis and Treatment Engineering Technology Research center, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong 510316, China
| |
Collapse
|
36
|
Wang Y, Lu X, Lu J, Hernigou P, Jin F. The role of macrophage polarization in tendon healing and therapeutic strategies: Insights from animal models. Front Bioeng Biotechnol 2024; 12:1366398. [PMID: 38486869 PMCID: PMC10937537 DOI: 10.3389/fbioe.2024.1366398] [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: 01/06/2024] [Accepted: 02/19/2024] [Indexed: 03/17/2024] Open
Abstract
Tendon injuries, a common musculoskeletal issue, usually result in adhesions to the surrounding tissue, that will impact functional recovery. Macrophages, particularly through their M1 and M2 polarizations, play a pivotal role in the inflammatory and healing phases of tendon repair. In this review, we explore the role of macrophage polarization in tendon healing, focusing on insights from animal models. The review delves into the complex interplay of macrophages in tendon pathology, detailing how various macrophage phenotypes contribute to both healing and adhesion formation. It also explores the potential of modulating macrophage activity to enhance tendon repair and minimize adhesions. With advancements in understanding macrophage behavior and the development of innovative biomaterials, this review highlights promising therapeutic strategies for tendon injuries.
Collapse
Affiliation(s)
- Yicheng Wang
- Department of Pediatric Orthopedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Lu
- Shanghai Bio-lu Biomaterials Co., Ltd., Shanghai, China
- Shanghai Technology Innovation Center of Orthopedic Biomaterials, Shanghai, China
| | - Jianxi Lu
- Shanghai Bio-lu Biomaterials Co., Ltd., Shanghai, China
- Shanghai Technology Innovation Center of Orthopedic Biomaterials, Shanghai, China
| | - Philippe Hernigou
- University Paris East, Orthopedic Hospital Geoffroy Saint Hilaire, Paris, France
| | - Fangchun Jin
- Department of Pediatric Orthopedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
37
|
Zhao X, Deng H, Feng Y, Wang Y, Yao X, Ma Y, Zhang L, Jie J, Yang P, Yang Y. Immune-cell-mediated tissue engineering strategies for peripheral nerve injury and regeneration. J Mater Chem B 2024; 12:2217-2235. [PMID: 38345580 DOI: 10.1039/d3tb02557h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
During the process of peripheral nerve repair, there are many complex pathological and physiological changes, including multi-cellular responses and various signaling molecules, and all these events establish a dynamic microenvironment for axon repair, regeneration, and target tissue/organ reinnervation. The immune system plays an indispensable role in the process of nerve repair and function recovery. An effective immune response not only involves innate-immune and adaptive-immune cells but also consists of chemokines and cytokines released by these immune cells. The elucidation of the orchestrated interplay of immune cells with nerve regeneration and functional restoration is meaningful for the exploration of therapeutic strategies. This review mainly enumerates the general immune cell response to peripheral nerve injury and focuses on their contributions to functional recovery. The tissue engineering-mediated strategies to regulate macrophages and T cells through physical and biochemical factors combined with scaffolds are discussed. The dynamic immune responses during peripheral nerve repair and immune-cell-mediated tissue engineering methods are presented, which provide a new insight and inspiration for immunomodulatory therapies in peripheral nerve regeneration.
Collapse
Affiliation(s)
- Xueying Zhao
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Hui Deng
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Yuan Feng
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Yuehan Wang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Xiaomin Yao
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Yuyang Ma
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Luzhong Zhang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Jing Jie
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nantong University, 226001, Nantong, P. R. China.
| | - Pengxiang Yang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| |
Collapse
|
38
|
Carrara SC, Davila-Lezama A, Cabriel C, Berenschot EJ, Krol S, Gardeniers J, Izeddin I, Kolmar H, Susarrey-Arce A. 3D topographies promote macrophage M2d-Subset differentiation. Mater Today Bio 2024; 24:100897. [PMID: 38169974 PMCID: PMC10758855 DOI: 10.1016/j.mtbio.2023.100897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 11/11/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024] Open
Abstract
In vitro cellular models denote a crucial part of drug discovery programs as they aid in identifying successful drug candidates based on their initial efficacy and potency. While tremendous headway has been achieved in improving 2D and 3D culture techniques, there is still a need for physiologically relevant systems that can mimic or alter cellular responses without the addition of external biochemical stimuli. A way forward to alter cellular responses is using physical cues, like 3D topographical inorganic substrates, to differentiate macrophage-like cells. Herein, protein secretion and gene expression markers for various macrophage subsets cultivated on a 3D topographical substrate are investigated. The results show that macrophages differentiate into anti-inflammatory M2-type macrophages, secreting increased IL-10 levels compared to the controls. Remarkably, these macrophage cells are differentiated into the M2d subset, making up the main component of tumour-associated macrophages (TAMs), as measured by upregulated Il-10 and Vegf mRNA. M2d subset differentiation is attributed to the topographical substrates with 3D fractal-like geometries arrayed over the surface, else primarily achieved by tumour-associated factors in vivo. From a broad perspective, this work paves the way for implementing 3D topographical inorganic surfaces for drug discovery programs, harnessing the advantages of in vitro assays without external stimulation and allowing the rapid characterisation of therapeutic modalities in physiologically relevant environments.
Collapse
Affiliation(s)
- Stefania C. Carrara
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
- Centre for Synthetic Biology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Amanda Davila-Lezama
- Facultad de Ciencias de la Salud (FACISALUD), Universidad Autónoma de Baja California, Blvd. Universitario 1000, Valle de las Palmas, 22260 Tijuana, Mexico
- Mesoscale Chemical Systems, MESA+ Institute, University of Twente, P.O. Box 217, 7500AE Enschede, the Netherlands
| | - Clément Cabriel
- Institut Langevin, ESPCI Paris, CNRS, Université PSL, 75005 Paris, France
| | - Erwin J.W. Berenschot
- Mesoscale Chemical Systems, MESA+ Institute, University of Twente, P.O. Box 217, 7500AE Enschede, the Netherlands
| | - Silke Krol
- Mesoscale Chemical Systems, MESA+ Institute, University of Twente, P.O. Box 217, 7500AE Enschede, the Netherlands
- Encytos B.V., Piet Heinstraat 12, Enschede, the Netherlands
| | - J.G.E. Gardeniers
- Mesoscale Chemical Systems, MESA+ Institute, University of Twente, P.O. Box 217, 7500AE Enschede, the Netherlands
| | - Ignacio Izeddin
- Institut Langevin, ESPCI Paris, CNRS, Université PSL, 75005 Paris, France
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
- Centre for Synthetic Biology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Arturo Susarrey-Arce
- Mesoscale Chemical Systems, MESA+ Institute, University of Twente, P.O. Box 217, 7500AE Enschede, the Netherlands
| |
Collapse
|
39
|
Majidi M, Pakzad S, Salimi M, Azadbakht A, Hajighasemlou S, Amoupour M, Nokhbedehghan Z, Bonakdar S, Sineh Sepehr K, Pal Singh Chauhan N, Gholipourmalekabadi M. Macrophage cell morphology-imprinted substrates can modulate mesenchymal stem cell behaviors and macrophage M1/M2 polarization for wound healing applications. Biotechnol Bioeng 2023; 120:3638-3654. [PMID: 37668186 DOI: 10.1002/bit.28546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 08/09/2023] [Accepted: 08/24/2023] [Indexed: 09/06/2023]
Abstract
Mesenchymal stem cells and macrophages (MQ) are two very important cells involved in the normal wound healing process. It is well understood that topological cues and mechanical factors can lead to different responses in stem cells and MQ by influencing their shape, cytoskeleton proliferation, migration, and differentiation, which play an essential role in the success or failure of biomaterial implantation and more importantly wound healing. On the other hand, the polarization of MQ from proinflammatory (M1) to prohealing (M2) phenotypes has a critical role in the acceleration of wound healing. In this study, the morphology of different MQ subtypes (M0, M1, and M2) was imprinted on a silicon surface (polydimethylsiloxane [PDMS]) to prepare a nano-topography cell-imprinted substrate with the ability to induce anti-inflammatory effects on the mouse adipose-derived stem cells (ADSCs) and RAW264.7 monocyte cell line (MO). The gene expression profiles and flow cytometry of MQ revealed that the cell shape microstructure promoted the MQ phenotypes according to the specific shape of each pattern. The ELISA results were in agreement with the gene expression profiles. The ADSCs on the patterned PDMS exhibited remarkably different shapes from no-patterned PDMS. The MOs grown on M2 morphological patterns showed a significant increase in expression and section of anti-inflammatory cytokine compared with M0 and M1 patterns. The ADSCs homing in niches heavily deformed the cytoskeletal, which is probably why the gene expression and phenotype unexpectedly changed. In conclusion, wound dressings with M2 cell morphology-induced surfaces are suggested as excellent anti-inflammatory and antiscarring dressings.
Collapse
Affiliation(s)
- Mohammad Majidi
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Saeedreza Pakzad
- Food and Drug Laboratory Research Center, Food and Drug Administration, Iran Ministry of Health and Medical Education, Tehran, Iran
| | - Maryam Salimi
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abdolnaser Azadbakht
- Department of Biomedical Engineering, Central Tehran Branch, Islamic Azad University, Tehran, Iran
- Stem Cell Research Center, Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Saieh Hajighasemlou
- Food and Drug Administration, Iran Ministry of Health and Medical Education, Tehran, Iran
| | - Moein Amoupour
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Nokhbedehghan
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Koushan Sineh Sepehr
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Mazaher Gholipourmalekabadi
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
Li J, Long J, Zhao Z, Wang Q, Bo W, Ren L, Fan Y, Wang P, Cheng Y, Liu B, Cheng X, Xi H. Procedural Promotion of Multiple Stages in the Wound Healing Process by Graphene-Spiky Silica Heterostructured Nanoparticles. Int J Nanomedicine 2023; 18:6585-6599. [PMID: 38026527 PMCID: PMC10644860 DOI: 10.2147/ijn.s426552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/28/2023] [Indexed: 12/01/2023] Open
Abstract
Background Multiple stages including hemostasis, inflammation, proliferation, and remodeling were involved in the wound healing process. The increase in nanomaterials in recent years has extended the scope of tools for wound healing; however, it is still difficult to achieve the four multistage procedures simultaneously. Materials and Methods In this study, graphene-spiky silica heterostructured nanoparticles (GS) were synthesized for the procedural acceleration of the multistage in wound healing process. The nanobridge effect of GS was analyzed through the adhesion of two skins, the antibacterial effect was assessed in Gram-negative Escherichia coli (E. coli) and Gram-positive Staphylococcus aureus (S. aureus) bacteria, cell proliferation and migration were investigated in mouse embryonic fibroblast (NIH-3T3) cells, and the in vivo wound healing effect was examined in female BALB/c mice with a cutting wound and E. coli or S. aureus bacteria infection on the back. Results First, GS has a strong nanobridge effect on the rapid closure of wounds because the spiky architecture on the surface of GS facilitates the adhesion of skins, promoting the hemostasis stage. Second, graphene exhibits antimicrobial activities both in chemical and physical interactions, especially under simulated sunlight irradiation. Third, graphene plays an important role in scaffolding function, together with the spiky topographical architecture of GS, accelerating the proliferation and maturation stages. Conclusion By periodically promoting every stage of wound healing, GS combined with simulated sunlight irradiation could significantly accelerate wound healing. With a simple composition and compact structure but multiple functions, this strategy will be the guideline for the development of ideal wound-healing nanomaterials.
Collapse
Affiliation(s)
- Jie Li
- Department of Orthopedics, Children’s Hospital Affiliated to Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Jiangtao Long
- Department of Orthopedics, Children’s Hospital Affiliated to Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Zheng Zhao
- Department of General Surgery, Children’s Hospital Affiliated to Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Qianqian Wang
- Department of Orthopedics, Children’s Hospital Affiliated to Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Wang Bo
- Department of General Surgery, Children’s Hospital Affiliated to Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Liang Ren
- Department of Orthopedics, Children’s Hospital Affiliated to Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Yan Fan
- Department of Burn and Plastic Surgery, Children’s Hospital affiliated to Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Peng Wang
- Department of Orthopedics, Children’s Hospital Affiliated to Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Yi Cheng
- Department of Orthopedics, Children’s Hospital Affiliated to Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Binbin Liu
- Department of Orthopedics, Children’s Hospital Affiliated to Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Xinkui Cheng
- Department of Orthopedics, Children’s Hospital Affiliated to Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Hongwei Xi
- Department of General Surgery, Children’s Hospital Affiliated to Shanxi Medical University, Taiyuan, People’s Republic of China
| |
Collapse
|
41
|
Li P, Tian X, Zhou X, Xun Q, Zheng J, Mu Y, Liao J. A novel porous hydroxyapatite scaffold (pHAMG) enhances angiogenesis and osteogenesis around dental implants by regulating the immune microenvironment. Clin Oral Investig 2023; 27:6879-6889. [PMID: 37843634 DOI: 10.1007/s00784-023-05304-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 10/02/2023] [Indexed: 10/17/2023]
Abstract
OBJECTIVE The purpose was to evaluate whether a novel porous hydroxyapatite (HA) scaffold with a 25-30-µm groove structure (pHAMG) may improve bone osteogenesis, angiogenesis, and bone integration of titanium dental implants in animal models. METHODS The pHAMG was prepared by chemical precipitation method and its elemental composition and crystal structure were evaluated. The ability of the scaffolds to induce ectopic osteogenesis and the ability of scaffolds combined with titanium dental implants to induce orthotopic peri-implant angiogenesis, osteogenesis, and osteointegration were tested after implantation into the femur muscle pocket in rats and the mandibular defects in beagle dogs, respectively. The elemental composition was evaluated by SEM-EDS; the expression of the relevant osteogenic/inflammation marker and the anti-/pro-inflammation markers was evaluated by immunostaining and immunofluorescence, respectively. RESULTS In animal experiments with ectopic and peri-implant osteogenesis, pHAMG resulted in significantly larger neovascularization by hematoxylin-eosin staining, as well as deposition of collagen fibers by Masson staining than HA. Meanwhile, microgrooves in pHAMG upregulate more bone morphogenetic protein (BMP) 2 and interleukin-4 (IL-4) and -10 (IL-10) and downregulate more IL-1β and tumor necrosis factor-α (TNF-α) than that in HA. The pHAMG showed greater expression of arginase (Arg)-1 and lower expression of inducible nitric oxide synthase (iNOS) than HA. CONCLUSION The novel pHAMG can better repair bone defects in ectopic and orthotopic model. It also transfers macrophages to anti-inflammatory phenotypes, promoting angiogenic and osteogenesis in scaffolds, and bone integration in implants. CLINICAL RELEVANCE The novel pHAMG induce greater osteogenesis and angiogenesis which could be utilized in the clinical treatment.
Collapse
Affiliation(s)
- Peng Li
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xi Tian
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xinzhu Zhou
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qiongyu Xun
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Junwen Zheng
- Southwest Medical University, Luzhou, Sichuan, China
| | - Yandong Mu
- Department of Stomatology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Juan Liao
- Department of Stomatology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
42
|
Gonthier A, Botvinick EL, Grosberg A, Mohraz A. Effect of Porous Substrate Topographies on Cell Dynamics: A Computational Study. ACS Biomater Sci Eng 2023; 9:5666-5678. [PMID: 37713253 PMCID: PMC10565724 DOI: 10.1021/acsbiomaterials.3c01008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 08/31/2023] [Indexed: 09/16/2023]
Abstract
Controlling cell-substrate interactions via the microstructural characteristics of biomaterials offers an advantageous path for modulating cell dynamics, mechanosensing, and migration, as well as for designing immune-modulating implants, all without the drawbacks of chemical-based triggers. Specifically, recent in vivo studies have suggested that a porous implant's microscale curvature landscape can significantly impact cell behavior and ultimately the immune response. To investigate such cell-substrate interactions, we utilized a 3D computational model incorporating the minimum necessary physics of cell migration and cell-substrate interactions needed to replicate known in vitro behaviors. This model specifically incorporates the effect of membrane tension, which was found to be necessary to replicate in vitro cell behavior on curved surfaces. Our simulated substrates represent two classes of porous materials recently used in implant studies, which have markedly different microscale curvature distributions and pore geometries. We found distinct differences between the overall migration behaviors, shapes, and actin polymerization dynamics of cells interacting with the two substrates. These differences were correlated to the shape energy of the cells as they interacted with the porous substrates, in effect interpreting substrate topography as an energetic landscape interrogated by cells. Our results demonstrate that microscale curvature directly influences cell shape and migration and, therefore, is likely to influence cell behavior. This supports further investigation of the relationship between the surface topography of implanted materials and the characteristic immune response, a complete understanding of which would broadly advance principles of biomaterial design.
Collapse
Affiliation(s)
- Alyse
R. Gonthier
- Department
of Materials Science & Engineering, University of California, Irvine, Irvine, California 92697, United States
| | - Elliot L. Botvinick
- Department
of Biomedical Engineering, University of
California, Irvine, Irvine, California 92697, United States
- Center
for Complex Biological Systems, University
of California, Irvine, Irvine, California 92697, United States
- Beckman
Laser Institute and Medical Clinic, University
of California, Irvine, Irvine, California 92697, United States
- Department
of Surgery,University of California, Irvine, Irvine, California 92697, United States
- Edwards
Lifesciences
Foundation Cardiovascular Innovation & Research Center, University of California, Irvine, Irvine, California 92697, United States
| | - Anna Grosberg
- Department
of Biomedical Engineering, University of
California, Irvine, Irvine, California 92697, United States
- Center
for Complex Biological Systems, University
of California, Irvine, Irvine, California 92697, United States
- Edwards
Lifesciences
Foundation Cardiovascular Innovation & Research Center, University of California, Irvine, Irvine, California 92697, United States
- Department
of Chemical & Biomolecular Engineering, University of California, Irvine, Irvine, California 92697, United States
- The
NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California 92697, United States
- Sue
and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California 92697, United States
| | - Ali Mohraz
- Department
of Materials Science & Engineering, University of California, Irvine, Irvine, California 92697, United States
- Department
of Chemical & Biomolecular Engineering, University of California, Irvine, Irvine, California 92697, United States
| |
Collapse
|
43
|
Li R, Feng D, Han S, Zhai X, Yu X, Fu Y, Jin F. Macrophages and fibroblasts in foreign body reactions: How mechanical cues drive cell functions? Mater Today Bio 2023; 22:100783. [PMID: 37701130 PMCID: PMC10494263 DOI: 10.1016/j.mtbio.2023.100783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/14/2023] Open
Abstract
Biomaterials, when implanted in the human body, can induce a series of cell- and cytokine-related reactions termed foreign body reactions (FBRs). In the progression of FBRs, macrophages regulate inflammation and healing by polarizing to either a pro-inflammatory or pro-healing phenotype and recruit fibroblasts by secreting cytokines. Stimulated by the biomaterials, fibrotic capsule is formed eventually. The implant, along with its newly formed capsule, introduces various mechanical cues that influence cellular functions. Mechanosensing proteins, such as integrins or ion channels, transduce extracellular mechanical signals into cytoplasm biochemical signals in response to mechanical stimuli. Consequently, the morphology, migration mode, function, and polarization state of the cells are affected. Modulated by different intracellular signaling pathways and their crosstalk, the expression of fibrotic genes increases with fibroblast activation and fibroblast to myofibroblast transition under stiff or force stimuli. However, summarized in most current studies, the outcomes of macrophage polarization in the effect of different mechanical cues are inconsistent. The underlying mechanisms should be investigated with more advanced technology and considering more interfering aspects. Further research is needed to determine how to modulate the progression of fibrotic capsule formation in FBR artificially.
Collapse
Affiliation(s)
- Rihan Li
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
- Department of Breast and Reconstructive Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Dongdong Feng
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
- Department of Breast and Reconstructive Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Siyuan Han
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
- Department of Breast and Reconstructive Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Xiaoyue Zhai
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, Liaoning, 110000, China
| | - Xinmiao Yu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
- Department of Breast and Reconstructive Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Yuanyuan Fu
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, Liaoning, 110000, China
| | - Feng Jin
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| |
Collapse
|
44
|
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
|
45
|
Tao YB, Shi KQ, Xiao J. [Research progress of biomaterials with ordered micro-nano structure in wound repair]. ZHONGHUA SHAO SHANG YU CHUANG MIAN XIU FU ZA ZHI 2023; 39:896-900. [PMID: 37805808 DOI: 10.3760/cma.j.cn501225-20221017-00455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 10/09/2023]
Abstract
Many injury-causing factors, including burns and surgery, etc., can lead to the destruction of structure and function of skin. Suitable wound dressing or implant is the material basis to promote wound healing and regeneration. Biomaterials with micro-nano structure can affect cell behavior, promote orderly growth of cell in accordance with the structure. Their application in wound healing can promote angiogenesis, regulate immune response, and reduce scar area. In recent years, the application of biomaterials with ordered micro-nano structure in tissue engineering has attracted extensive attention. This paper introduces the structure and preparation methods of several biomaterials with ordered micro-nano structure, and focuses on how the surface microstructure of biomaterials affects the process of wound healing and its molecular mechanism, in order to find and develop medical biomaterials that are closer to the skin tissue structure for clinical wound treatment.
Collapse
Affiliation(s)
- Y B Tao
- Laboratory of Translational Medicine, Wound Repair and Regeneration Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - K Q Shi
- Laboratory of Translational Medicine, Wound Repair and Regeneration Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - J Xiao
- Laboratory of Translational Medicine, Wound Repair and Regeneration Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
46
|
Wahab R, Hasan MM, Azam Z, Grippo PJ, Al-Hilal TA. The role of coagulome in the tumor immune microenvironment. Adv Drug Deliv Rev 2023; 200:115027. [PMID: 37517779 PMCID: PMC11099942 DOI: 10.1016/j.addr.2023.115027] [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: 01/31/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/01/2023]
Abstract
The rising incidence and persistent thrombosis in multiple cancers including those that are immunosuppressive highlight the need for understanding the tumor coagulome system and its role beyond hemostatic complications. Immunotherapy has shown significant benefits in solid organ tumors but has been disappointing in the treatment of hypercoagulable cancers, such as glioblastoma and pancreatic ductal adenocarcinomas. Thus, targeting thrombosis to prevent immunosuppression seems a clinically viable approach in cancer treatment. Hypercoagulable tumors often develop fibrin clots within the tumor microenvironment (TME) that dictates the biophysical characteristics of the tumor tissue. The application of systems biology and single-cell approaches highlight the potential role of coagulome or thrombocytosis in shaping the tumor immune microenvironment (TIME). In-depth knowledge of the tumor coagulome would provide unprecedented opportunities to better predict the hemostatic complications, explore how thrombotic stroma modulates tumor immunity, reexamine the significance of clinical biomarkers, and enable steering the stromal versus systemic immune response for boosting the effectiveness of immune checkpoint inhibitors in cancer treatment. We focus on the role of coagulation factors in priming a suppressive TIME and the huge potential of existing anticoagulant drugs in the clinical settings of cancer immunotherapy.
Collapse
Affiliation(s)
- Riajul Wahab
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Md Mahedi Hasan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79968, USA; Department of Environmental Science & Engineering, College of Science, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Zulfikar Azam
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Paul J Grippo
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Taslim A Al-Hilal
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79968, USA; Department of Environmental Science & Engineering, College of Science, University of Texas at El Paso, El Paso, TX 79968, USA.
| |
Collapse
|
47
|
Rosado-Galindo H, Domenech M. Substrate topographies modulate the secretory activity of human bone marrow mesenchymal stem cells. Stem Cell Res Ther 2023; 14:208. [PMID: 37605275 PMCID: PMC10441765 DOI: 10.1186/s13287-023-03450-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 08/11/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) secrete a diversity of factors with broad therapeutic potential, yet current culture methods limit potency outcomes. In this study, we used topographical cues on polystyrene films to investigate their impact on the secretory profile and potency of bone marrow-derived MSCs (hBM-MSCs). hBM-MSCs from four donors were cultured on topographic substrates depicting defined roughness, curvature, grooves and various levels of wettability. METHODS The topographical PS-based array was developed using razor printing, polishing and plasma treatment methods. hBM-MSCs from four donors were purchased from RoosterBio and used in co-culture with peripheral blood mononuclear cells (PBMCs) from Cell Applications Inc. in an immunopotency assay to measure immunosuppressive capacity. Cells were cultured on low serum (2%) for 24-48 h prior to analysis. Image-based analysis was used for cell quantification and morphology assessment. Metabolic activity of BM-hMSCs was measured as the mitochondrial oxygen consumption rate using an extracellular flux analyzer. Conditioned media samples of BM-hMSCs were used to quantify secreted factors, and the data were analyzed using R statistics. Enriched bioprocesses were identify using the Gene Ontology tool enrichGO from the clusterprofiler. One-way and two-way ANOVAs were carried out to identify significant changes between the conditions. Results were deemed statistically significant for combined P < 0.05 for at least three independent experiments. RESULTS Cell viability was not significantly affected in the topographical substrates, and cell elongation was enhanced at least twofold in microgrooves and surfaces with a low contact angle. Increased cell elongation correlated with a metabolic shift from oxidative phosphorylation to a glycolytic state which is indicative of a high-energy state. Differential protein expression and gene ontology analyses identified bioprocesses enriched across donors associated with immune modulation and tissue regeneration. The growth of peripheral blood mononuclear cells (PBMCs) was suppressed in hBM-MSCs co-cultures, confirming enhanced immunosuppressive potency. YAP/TAZ levels were found to be reduced on these topographies confirming a mechanosensing effect on cells and suggesting a potential role in the immunomodulatory function of hMSCs. CONCLUSIONS This work demonstrates the potential of topographical cues as a culture strategy to improve the secretory capacity and enrich for an immunomodulatory phenotype in hBM-MSCs.
Collapse
Affiliation(s)
- Heizel Rosado-Galindo
- Bioengineering Program, University of Puerto Rico-Mayagüez, Road 108, KM 1.1., Mayagüez, PR, 00680, USA
| | - Maribella Domenech
- Bioengineering Program, University of Puerto Rico-Mayagüez, Road 108, KM 1.1., Mayagüez, PR, 00680, USA.
- Department of Chemical Engineering, University of Puerto Rico-Mayagüez, Road 108, KM 1.1., Mayagüez, PR, 00680, USA.
| |
Collapse
|
48
|
Wilson HM. Modulation of macrophages by biophysical cues in health and beyond. DISCOVERY IMMUNOLOGY 2023; 2:kyad013. [PMID: 38567062 PMCID: PMC10917218 DOI: 10.1093/discim/kyad013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/13/2023] [Accepted: 08/09/2023] [Indexed: 04/04/2024]
Abstract
Macrophages play a key role in tissue development and homeostasis, innate immune defence against microbes or tumours, and restoring homeostasis through tissue regeneration following infection or injury. The ability to adopt such diverse functions is due to their heterogeneous nature, which is driven largely by their developmental origin and their response to signals they encounter from the microenvironment. The most well-characterized signals driving macrophage phenotype and function are biochemical and metabolic. However, the way macrophages sense and respond to their extracellular biophysical environment is becoming increasingly recognized in the field of mechano-immunology. These biophysical cues can be signals from tissue components, such as the composition and charge of extracellular matrix or topography, elasticity, and stiffness of the tissue surrounding cells; and mechanical forces such as shear stress or stretch. Macrophages are important in determining whether a disease resolves or becomes chronic. Ageing and diseases such as cancer or fibrotic disorders are associated with significant changes in the tissue biophysical environment, and this provides signals that integrate with those from biochemical and metabolic stimuli to ultimately dictate the overall function of macrophages. This review provides a brief overview of macrophage polarization, followed by a selection of commonly recognized physiological and applied biophysical stimuli impacting macrophage activity, and the potential signalling mechanisms driving downstream responses. The effects of biophysical cues on macrophages' function in homeostasis and disease and the associated clinical implications are also highlighted.
Collapse
Affiliation(s)
- Heather M Wilson
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
49
|
Xu Y, Ying L, Lang JK, Hinz B, Zhao R. Modeling Mechanical Activation of Macrophages During Pulmonary Fibrogenesis for Targeted Anti-Fibrosis Therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.19.549794. [PMID: 37503121 PMCID: PMC10370161 DOI: 10.1101/2023.07.19.549794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Pulmonary fibrosis, as seen in idiopathic pulmonary fibrosis (IPF) and COVID-induced pulmonary fibrosis, is an often-fatal lung disease. Increased numbers of immune cells such as macrophages were shown to accumulate in the fibrotic lung, but it is unclear how they contribute to the development of fibrosis. To recapitulate the macrophage mechanical activation in the fibrotic lung tissue microenvironment, we developed a fibrotic microtissue model with cocultured human macrophages and fibroblasts. We show that profibrotic macrophages seeded on topographically controlled stromal tissue constructs become mechanically activated. The resulting co-alignment of macrophages, collagen fibers and fibroblasts promote widespread fibrogenesis in micro-engineered lung tissues. Anti-fibrosis treatment using pirfenidone disrupts the polarization and mechanical activation of profibrotic macrophages, leading to fibrosis inhibition. Pirfenidone inhibits the mechanical activation of macrophages by suppressing integrin αMβ2 (CD11b/CD18) and Rho-associated kinase 2, which is a previously unknown mechanism of action of the drug. Together, these results demonstrate a potential pulmonary fibrogenesis mechanism at the tissue level contributed by mechanically activated macrophages. We propose the coculture, force-sensing microtissue model as a powerful tool to study the complex immune-stromal cell interactions and the mechanism of action of anti-fibrosis drugs.
Collapse
|
50
|
Qiang H, Hou C, Zhang Y, Luo X, Li J, Meng C, Liu K, Lv Z, Chen X, Liu F. CaP-coated Zn-Mn-Li alloys regulate osseointegration via influencing macrophage polarization in the osteogenic environment. Regen Biomater 2023; 10:rbad051. [PMID: 37324238 PMCID: PMC10267298 DOI: 10.1093/rb/rbad051] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 06/17/2023] Open
Abstract
Immune response is an important factor in determining the fate of bone replacement materials, in which macrophages play an important role. It is a new idea to design biomaterials with immunomodulatory function to reduce inflammation and promote bone integration by regulating macrophages polarization. In this work, the immunomodulatory properties of CaP Zn-Mn-Li alloys and the specific mechanism of action were investigated. We found that the CaP Zn0.8Mn0.1Li alloy promoted the polarization of macrophages toward M2 and reduced inflammation, which could effectively upregulate osteogenesis-related factors and promote new bone formation, indicating the important role of macrophages polarization in biomaterial induction of osteogenesis. In vivo studies further demonstrated that CaP Zn0.8Mn0.1Li alloy could stimulate osteogenesis better than other Zn-Mn-Li alloys implantations by regulating macrophages polarization and reducing inflammation. In addition, transcriptome results showed that CaP Zn0.8Mn0.1Li played an important regulatory role in the life process of macrophages, activating Toll-like receptor signaling pathway, which participated in the activation and attenuation of inflammation, and accelerated bone integration. Thus, by preparing CaP coatings on the surface of Zn-Mn-Li alloys and combining the bioactive ingredient with controlled release, the biomaterial will be imbibed with beneficial immunomodulatory properties that promote bone integration.
Collapse
Affiliation(s)
| | | | - Yujue Zhang
- Liaocheng People’s Hospital, Liaocheng Dongchangfu People’s Hospital, Liaocheng 252000, China
- School of Materials Science and Engineering, University of Science and Technology, Beijing 100083, China
| | - Xin Luo
- Liaocheng People’s Hospital, Liaocheng Dongchangfu People’s Hospital, Liaocheng 252000, China
| | - Jun Li
- Liaocheng People’s Hospital, Liaocheng Dongchangfu People’s Hospital, Liaocheng 252000, China
| | - Chunxiu Meng
- Liaocheng People’s Hospital, Liaocheng Dongchangfu People’s Hospital, Liaocheng 252000, China
| | - Kun Liu
- Liaocheng People’s Hospital, Liaocheng Dongchangfu People’s Hospital, Liaocheng 252000, China
| | - Zhaoyong Lv
- Correspondence address. E-mail: (Z.L.); (X.C.); (F.L.)
| | - Ximeng Chen
- Correspondence address. E-mail: (Z.L.); (X.C.); (F.L.)
| | - Fengzhen Liu
- Correspondence address. E-mail: (Z.L.); (X.C.); (F.L.)
| |
Collapse
|