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
|
Qiu Y, Liu H, Han C, Yan Z, Lu Y, Ren L, Wang Q, Zhou Q, Xue L. The effect of copper content in Ti-Cu alloy with bone regeneration ability on the phenotypic transformation of macrophages. Colloids Surf B Biointerfaces 2025; 252:114641. [PMID: 40138785 DOI: 10.1016/j.colsurfb.2025.114641] [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: 01/06/2025] [Revised: 03/09/2025] [Accepted: 03/16/2025] [Indexed: 03/29/2025]
Abstract
Titanium (Ti) alloys are widely used in bone repair due to their excellent biocompatibility and mechanical properties. However, managing post-implantation inflammatory responses in the defect region and accelerating the healing process remain major challenges in the design of such materials. As a bridge between the innate and adaptive immune systems, macrophages play a pivotal role in bone defect healing through their M2 polarization, which facilitates the secretion of tissue repair-promoting cytokines. Research on the role of copper ions (Cu²⁺) in regulating inflammatory responses at injury sites suggests their potential as active ions for incorporation into alloys as a secondary phase to modulate macrophage polarization. However, the effective concentration and mechanisms in this process remain unclear. Here, we synthesized Ti-xCu (x = 3, 5, 7 wt%) alloys and investigated the effects of copper concentration on macrophage M1/M2 polarization and the underlying mechanisms. In an 8-week rat mandibular bone regeneration experiment, Ti-5Cu demonstrated superior performance compared to pure titanium. At the early stage (2 weeks), Ti-5Cu promoted the dominance of M1 macrophages and upregulated inflammatory cytokines, facilitating the initial inflammatory response. Subsequently, a timely M1-to-M2 phenotype transition was observed, accompanied by elevated expression of the repair-related cytokine IL-10, ultimately leading to improved bone healing. This study provides a theoretical foundation for the development of titanium-copper composite materials with anti-inflammatory and pro-healing properties, paving the way for innovative solutions to promote bone defect repair.
Collapse
Affiliation(s)
- Yueyang Qiu
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| | - Hui Liu
- Shi-changxu Innovation Center for Advanced Materials, Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, China
| | | | - Zhuoqun Yan
- Liaoning Upcera Co., Ltd, Benxi 117004, China
| | - Yanjin Lu
- Key Laboratory of Opto-Electronic Science and Technology for Medicine of Ministry of Education, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou 350117, China.
| | - Ling Ren
- Shi-changxu Innovation Center for Advanced Materials, Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, China
| | - Qiang Wang
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| | - Qing Zhou
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China.
| | - Lei Xue
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China.
| |
Collapse
|
3
|
Guan Q, Chen J, Liu Z, Xin J, Chen Y, Wang P, Liu J. Biocompatible cloaking of bacteria for effective tumor imaging and therapy. Mater Today Bio 2025; 32:101788. [PMID: 40321697 PMCID: PMC12049847 DOI: 10.1016/j.mtbio.2025.101788] [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: 02/20/2025] [Revised: 04/08/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025] Open
Abstract
Bacteria possess distinctive characteristics, such as tropism, motility, and genetic editability, which make them highly attractive for biomedical applications, such as tumor imaging and therapy. However, the immunogenicity triggered by endotoxins may lead to severe adverse effects and prompt quick elimination in the body, thus restricting their clinical applications. In this study, we described a double-layer coating technique employing tannic acid (TA) and albumin (BSA) for bacteria encapsulation. This compact coating effectively shields endotoxin exposure and inhibits endotoxin leakage from bacteria, exhibiting a favorable safety profile. Moreover, bacteria coated with BSA have superior biocompatibility with their surroundings, which prevents phagocytes from eliminating the bacteria, hence prolonging the reservation in vivo. As bacteria grow, the BSA-TA layer progressively detaches after reaching targeted sites, allowing free bacteria to exploit their own advantages. Importantly, the BSA-TA coating strategy protects bacteria against various environmental assaults without compromising their growth, proliferation, or motility, maintaining their inherent characteristics. In murine tumor models, the BSA-TA-coated bacteria demonstrated enhanced long-term tumor imaging and therapeutic efficacy against tumors. Together, the BSA-TA coating strategy improves the biocompatibility of bacteria and has the capacity to expand the range of bacteria for biomedical applications.
Collapse
Affiliation(s)
- Qing Guan
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Junjie Chen
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Zhaonan Liu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Jianfeng Xin
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Yu Chen
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Peng Wang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Jun Liu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| |
Collapse
|
4
|
Yu SY, Wu T, Xu KH, Liu RY, Yu TH, Wang ZH, Zhang ZT. 3D bioprinted biomimetic MOF-functionalized hydrogel scaffolds for bone regeneration: Synergistic osteogenesis and osteoimmunomodulation. Mater Today Bio 2025; 32:101740. [PMID: 40270888 PMCID: PMC12018039 DOI: 10.1016/j.mtbio.2025.101740] [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: 02/03/2025] [Revised: 03/30/2025] [Accepted: 04/06/2025] [Indexed: 04/25/2025] Open
Abstract
Critical-size bone defects remain a significant clinical challenge. The lack of endogenous stem cells with osteogenic differentiation potential in the defect area, combined with the inflammatory responses induced by scaffold implantation, highlights the need for biomaterials that can deliver stem cells and possess inflammatory regulation properties. In this study, we developed a 3D bioprinted gelatin methacrylate (GelMA) hydrogel scaffold modified with luteolin-loaded ZIF-8 (LUT@ZIF-8) nanoparticles, designed to deliver bone marrow mesenchymal stem cells (BMSCs) to the defect site and release bioactive components that promote osteogenesis and modulate the immune microenvironment. The LUT@ZIF-8/GelMA hydrogel scaffolds demonstrated excellent physical properties and biocompatibility. The sustained release of luteolin and zinc ions from the LUT@ZIF-8 nanoparticles conferred antibacterial, osteoinductive, and inflammatory regulation effects. The immune microenvironment modulated by LUT@ZIF-8/GelMA hydrogel scaffolds facilitated osteogenic differentiation of BMSCs. Furthermore, in vivo experiments confirmed the osteogenic and inflammatory regulation capabilities of the LUT@ZIF-8/GelMA hydrogel scaffolds. In conclusion, the 3D bioprinted LUT@ZIF-8/GelMA hydrogel scaffolds exhibit osteoimmunomodulatory properties, presenting a promising strategy for the treatment of bone defects.
Collapse
Affiliation(s)
- San-yang Yu
- The VIP Department, School and Hospital of Stomatology, China Medical University, Shenyang, 110002, PR China
| | - Ting Wu
- The VIP Department, School and Hospital of Stomatology, China Medical University, Shenyang, 110002, PR China
| | - Kai-hao Xu
- The VIP Department, School and Hospital of Stomatology, China Medical University, Shenyang, 110002, PR China
| | - Ru-yue Liu
- The VIP Department, School and Hospital of Stomatology, China Medical University, Shenyang, 110002, PR China
| | - Tian-hao Yu
- The VIP Department, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, 110002, PR China
| | - Zhen-hua Wang
- Department of Physiology, School of Life Sciences, China Medical University, Shenyang, 110122, PR China
| | - Zhong-ti Zhang
- The VIP Department, School and Hospital of Stomatology, China Medical University, Shenyang, 110002, PR China
| |
Collapse
|
5
|
Martkamjan C, Lerdsudkanung K, Tipay PS, Rezgui R, Teo JCM, Sapudom J. Machine learning-based label-free macrophage phenotyping in immune-material interactions. J Mater Chem B 2025; 13:5858-5870. [PMID: 40289902 DOI: 10.1039/d5tb00365b] [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
The rapid advancement of implantable biomedical materials necessitates a comprehensive understanding of macrophage interactions to optimize implant immunocompatibility. Macrophages, key immune regulators, exhibit phenotypic plasticity by polarizing into pro-inflammatory (M1) or anti-inflammatory (M2) subtypes. Conventional phenotyping techniques, such as flow cytometry and immunostaining, provide insights but have limitations related to fixation and endpoint analysis. This study presents a high-throughput, label-free macrophage phenotyping approach integrating AI-driven image classification with quantitative phase imaging (QPI). THP-1-derived macrophages were differentiated into M0, M1, M2a, and M2c phenotypes, and their morphological and refractive index properties were analyzed using QPI. Although QPI alone could not fully distinguish phenotypes, deep learning models, including GoogLeNet, ShuffleNet, VGG-16, and ResNet-18, were evaluated, with ResNet-18 achieving over 90% accuracy. Additionally, macrophage responses to collagen coatings (types I, III, and IV) were assessed using machine learning-based phenotyping and cytokine profiling. Collagen I induced an M1 response, collagen III supported a balanced M1/M2 profile, and collagen IV promoted a controlled immune environment. These findings demonstrate the potential of AI-driven QPI as a non-invasive tool for macrophage characterization, offering insights into biomaterial immunocompatibility and informing implant design strategies.
Collapse
Affiliation(s)
- Chawalwat Martkamjan
- International School of Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok, Thailand
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates.
| | - Kornlavit Lerdsudkanung
- International School of Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok, Thailand
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates.
| | - Paul Sean Tipay
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates.
- Independent Researcher, Abu Dhabi, United Arab Emirates
| | - Rachid Rezgui
- Core Technology Platform - Light Microscopy, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Jeremy C M Teo
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates.
| | - Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
6
|
Sun T, Wang M, Zhang L, Gong M, Xie Q, Yang X, Xiao S, Zhang W, Liu X, Zhao Y, Zhang Z, Zhou J, Zhang D, Zhou C. Engineered Bacterial Biohybrid-Mediated CD47-SIRPα Blockade and HSP90 Inhibition for Enhanced Immuno-Photothermal Therapy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:29183-29197. [PMID: 40331355 DOI: 10.1021/acsami.5c01645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Macrophage phagocytosis of tumor cells shows significant promise in cancer treatment. However, it faces great challenges due to the upregulation of antiphagocytosis molecules, such as CD47, on the surface of tumor cells. Merely reducing the level of CD47 is insufficient to induce phagocytosis of tumor cells because it lacks enough "eat me" signals. Here, we have developed an engineered bacterial biohybrid system (eVNP@AuNFs) to decrease the expression of CD47 and HSP90 proteins, achieving an enhanced immuno-photothermal combination therapy. The attenuated Salmonella VNP20009, capable of selectively accumulating in hypoxic tumor regions, was intracellularly genetically engineered with CD47 and HSP90 shRNA plasmids and an extracellularly adsorbed flower-like gold nanoparticle (AuNF) photothermal agent, forming an eVNP@AuNF bacterial hybrid. After administration into 4T1 tumor-bearing mice intravenously, the eVNP@AuNF bacterial hybrid could effectively accumulate in tumor tissues and release CD47 and HSP90 shRNA plasmids to reduce the expression of CD47 and HSP90 protein, leading to enhanced macrophage phagocytosis to tumor cells and an improved photothermal effect. Under further NIR-II laser irradiation, extracellular AuNFs of eVNP@AuNFs could photothermally induce immunogenic cell death, including surface calreticulin exposure and high-mobility group box 1 translocation, facilitating the infiltration of the "eat me" signal and multiple immune cells and enhancing tumor immunogenicity. The eVNP@AuNF bacterial hybrid could eradicate the primary tumor and elicit a systemic antitumor immunity response, inhibiting the recurrence of the tumor. This study presents a hybrid system involving bacteria, plasmids, and nanomaterials for tumor therapy, opening an avenue for hierarchical modulation of the tumor immune response.
Collapse
Affiliation(s)
- Tao Sun
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
- Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Miaomiao Wang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
- School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Liang Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Mingfu Gong
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Qian Xie
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Xiaofeng Yang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Shilin Xiao
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Wansu Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Xu Liu
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Yue Zhao
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Zhipeng Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Jun Zhou
- Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Dong Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Chunyu Zhou
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| |
Collapse
|
7
|
Jia H, Zhang H, Mo D, Xie B, Qiao H, Chen T, Song H, Xu X, Yang S. UTX Responds to Nanotopography to Suppress Macrophage Inflammatory Response by Remodeling H3K27me3 Modification. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e05723. [PMID: 40386877 DOI: 10.1002/advs.202505723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2025] [Revised: 04/28/2025] [Indexed: 05/20/2025]
Abstract
Peri-implantitis is the leading cause of implant failure, primarily due to weak defense at the implant-soft tissue interface, which disrupts the local immune microenvironment. As an integral part of this microenvironment, the implant-tissue interface plays a critical role in shaping immune cell function. Thus, engineering the surface topography of implants has emerged as a novel strategy for sustained immunomodulation following implantation. This study investigated the mechanical regulation of macrophage function by nanopatterned topographies. Titanium nanotubes (TNTs) surfaces reduce the expression of phosphorylated myosin light chain (pMLC) and promote the retention of the UTX histone methyltransferase in the nucleus. This process attenuates the enrichment of the repressive H3K27me3 histone marker at the Abca1 gene locus, increasing Abca1 expression and suppressing inflammation. This study reveals the mechanosensitivity of UTX and provides a new target for the development of therapeutic strategies that integrate mechanical signaling and immune modulation.
Collapse
Affiliation(s)
- Hengji Jia
- College of Stomatology, Chongqing Medical University, Chongqing, China
| | - He Zhang
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, China
- Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, China
| | - Dingqiang Mo
- College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Bo Xie
- College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Hongdou Qiao
- College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Tao Chen
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, China
- Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, China
| | - Haoyue Song
- College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Xinxin Xu
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, China
- Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, China
| | - Sheng Yang
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, China
- Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, China
| |
Collapse
|
8
|
Ma S, Zhao Z, Shang Y, Qi M, Xu S, Yao S, Li Y, Wang X, Tong T, Zheng H, Ma B, Yang Y, Wu J, Liu Z, Deng J. An Asymmetric Zn Membrane with Degradability, Antimicrobial, and Bone Immunomodulation for Guided Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40375688 DOI: 10.1021/acsami.5c06421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
As a biodegradable metal, zinc (Zn) offers a promising material option for barrier membranes in the field of bone regeneration due to its suitable degradation rate and mechanical properties. An ideal barrier membrane not only blocks the growth of epithelial fibers but also promotes bone regeneration. Therefore, we prepared an asymmetric Zn membrane with micrometer-sized pores on one side by laser etching, with the pore side facilitating cell adhesion and proliferation, and the smooth side facilitating the blocking of epithelial cell growth entry. And the antimicrobial peptide GL13K (P1) was loaded onto the smooth surface of Zn by Zn-specific binding peptide (NCS) to resist postoperative bacterial infections, and the small intestinal submucosal hydrogel complex (SIS-P2), which was specifically loaded with Substance P (SP), was placed in the pores on the pore side to modulate the immunity and promote osteogenesis. This innovative asymmetric zinc barrier membrane (Zn-P1-SIS-P2 membrane) exhibited excellent mechanical, antimicrobial, and biocompatibility properties. More importantly, the Zn-P1-SIS-P2 membrane promotes macrophage polarization toward the M2 type, thereby promoting osteogenic differentiation and providing a good immune microenvironment for bone regeneration. In addition, the Zn-P1-SIS-P2 membrane inhibited RANKL-induced osteoclast formation and suppressed bone resorption at the site of bone defects. In conclusion, the Zn-P1-SIS-P2 membrane demonstrated all the desirable qualities of a GBR therapeutic barrier membrane.
Collapse
Affiliation(s)
- Shiqing Ma
- Department of Stomatology, The Second Hospital of Tianjin Medical University, Tianjin 300211, PR China
| | - Zhezhe Zhao
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Yuli Shang
- Department of Stomatology, The Second Hospital of Tianjin Medical University, Tianjin 300211, PR China
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Mengyue Qi
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Shendan Xu
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Shiyu Yao
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Yumeng Li
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Xiaojing Wang
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Tianyi Tong
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Hong Zheng
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Beibei Ma
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Yilin Yang
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Jie Wu
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Zihao Liu
- Zhongnuo Dental Hospital, Tianjin Nankai District, Tianjin 300101, PR China
| | - Jiayin Deng
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| |
Collapse
|
9
|
Zhang C, Fu Z, Liu Q, Guo X, Li Z, Song W, Kong Y, Du J, Su Y, Yu B, Kong Y, Tian F, Fu X, Du X, Huang S. Bioprinted M2 macrophage-derived extracellular vesicle mimics attenuate foreign body reaction and enhance vascularized tissue regeneration. Biofabrication 2025; 17:035007. [PMID: 40328275 DOI: 10.1088/1758-5090/add49f] [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: 01/24/2025] [Accepted: 05/06/2025] [Indexed: 05/08/2025]
Abstract
Foreign body reaction (FBR) and insufficient vascularization greatly hinder the integration of 3D-bioprinted tissue substitutes with host tissues. Previous studies have shown that these problems are exacerbated by the stiffness of the 3D-bioprinted constructions, which is highly associated with the abnormal polarization of macrophages. Therefore, we developed an engineering strategy using membrane extrusion to prepare macrophage-derived extracellular vesicle mimics (EVMs). The EVMs derived from M1 and M2 macrophages (M1-EVMs and M2-EVMs) were rich in functional proteins. In the 2D environment, M1-EVMs promoted the fibrotic phenotype of fibroblasts, vascularization, and the M1 polarization of macrophages. In contrast, M2-EVMs effectively avoided the fibrotic trend, showed stronger angiogenic capabilities, and prevented excessive M1 polarization, demonstrating their potential to inhibit FBR and promote neovascularization. After bioprinting the EVMs loaded by gelatin-alginate bioink, the basic physical properties of the bioink were not significantly affected, and the biological functions of EVMs remain stable, indicating their potential as bioink additives. In the subcutaneous implantation model, unlike the FBR-aggravating effects of M1-EVMs, 3D-bioprinted M2-EVMs successfully reduced the immune response, prevented fibrous capsule formation, and increased vascular density. When applied to skin wound treatment, 3D-bioprinted M2-EVMs not only inhibited inflammatory levels but also exhibited pleiotropic pro-regenerative effects, effectively promoting vascularization, re-epithelialization, and appendage regeneration. As an innovative additive for bioinks, M2-EVMs present a promising approach to enhance the survival of bioengineered tissues and can further serve as a targeted drug loading system, promoting the development of regenerative medicine and improving clinical outcomes.
Collapse
Affiliation(s)
- Chao Zhang
- School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Ze Fu
- Chinese PLA Medical School, Beijing 100853, People's Republic of China
- Department of General Surgery, First Medical Center of Chinese PLA General Hospital, Beijing 100853, People's Republic of China
| | - Qinghua Liu
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Xu Guo
- College of Graduate, Tianjin Medical University, Tianjin 300203, People's Republic of China
| | - Zhao Li
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Wei Song
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Yi Kong
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Jinpeng Du
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Yanlin Su
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Bingyang Yu
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Yue Kong
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Feng Tian
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Xiaobing Fu
- School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
- Chinese PLA Medical School, Beijing 100853, People's Republic of China
- College of Graduate, Tianjin Medical University, Tianjin 300203, People's Republic of China
| | - Xiaohui Du
- Chinese PLA Medical School, Beijing 100853, People's Republic of China
- Department of General Surgery, First Medical Center of Chinese PLA General Hospital, Beijing 100853, People's Republic of China
| | - Sha Huang
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
- Chinese PLA Medical School, Beijing 100853, People's Republic of China
| |
Collapse
|
10
|
Dong L, Zhang G, Shen Z, Hong X, Xing Y, Wu Y, Yang W, Zhang B, Shi Z. Degradation of WE43 Magnesium Alloy in Vivo and Its Degradation Products on Macrophages. ACS OMEGA 2025; 10:17280-17295. [PMID: 40352546 PMCID: PMC12059945 DOI: 10.1021/acsomega.4c09349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 04/07/2025] [Accepted: 04/16/2025] [Indexed: 05/14/2025]
Abstract
Due to their biocompatibility, biodegradability, and suitable mechanical properties, magnesium-based biodegradable implants are emerging as a promising alternative to traditional metal implants. The Mg-4Y-3RE (WE43) biodegradable alloy is among the most extensively studied and widely utilized magnesium alloys in clinical applications. As an absorbable and degradable metallic material, magnesium alloys undergo gradual degradation, wear, and fracture within the body. These alloys reduce the long-term risks associated with permanent implants but generate insoluble byproducts that accumulate in surrounding tissues. Following the implantation of magnesium alloys, granulation tissue and fibrous encapsulation typically form around the material. However, limited research has addressed the interaction between insoluble byproducts of magnesium alloys and macrophages. This study focused on the biological effects of macrophages during the second stage of the host inflammatory response in the degradation process of magnesium alloy. Using subcutaneous implantation of WE43 magnesium alloy sheets, observations were made regarding the degradation components, morphological changes in surrounding tissues, and the biological effects of macrophages upon phagocytosis of insoluble byproducts. The primary degradation products of WE43 in vivo were identified as Ca3 (PO4)2, Mg3(PO4)2, Na3PO4, NaCa (PO4), MgSO4, MgCO3, NaCl, Mg24Y5, and Mg12YNd. Postimplantation, levels of IL-1β and IL-18 in adjacent tissues significantly increased (p < 0.05). By 8 weeks, compared to nitinol alloy, significant thickening of the fibrous capsule (p < 0.05) was observed, accompanied by substantial inflammatory cell infiltration, vascularization, and the presence of macrophages and multinucleated giant cells. Macrophages were observed extending pseudopodia to enclose and phagocytose particles, forming phagosomes and creating a relatively isolated microenvironment around the engulfed substances, where further particle degradation occurred. Following the phagocytosis of degradation products, macrophages exhibited increased lysosome numbers, mitochondrial swelling and damage, phagolysosome formation, and autophagosome development. Furthermore, the degradation products were observed to induce elevated reactive oxygen species (ROS) production in macrophages, activation of P2X7 receptors, enhanced IL-6 secretion, endoplasmic reticulum stress, autophagy, and activation of the NLRP3 inflammasome pathway. This study provides novel insights and contributes a theoretical foundation for a more comprehensive understanding of magnesium alloy degradation in vivo.
Collapse
Affiliation(s)
- Li Dong
- Department
of Cardiology, The Fourth Affiliated Hospital
of Harbin Medical University, Harbin 150001, China
| | - Guangde Zhang
- Department
of Cardiology, The Fourth Affiliated Hospital
of Harbin Medical University, Harbin 150001, China
| | - Zhiyuan Shen
- Department
of Cardiology, The Fourth Affiliated Hospital
of Harbin Medical University, Harbin 150001, China
| | - Xiaojian Hong
- Department
of Cardiology, The Fourth Affiliated Hospital
of Harbin Medical University, Harbin 150001, China
| | - Yongli Xing
- Department
of Medical Imaging, Second Hospital of Harbin, Harbin 150056, China
| | - Yue Wu
- Department
of Cardiology, The Fourth Affiliated Hospital
of Harbin Medical University, Harbin 150001, China
| | - Wei Yang
- Department
of Cardiology, The Fourth Affiliated Hospital
of Harbin Medical University, Harbin 150001, China
| | - Binmei Zhang
- Department
of Cardiology, The Fourth Affiliated Hospital
of Harbin Medical University, Harbin 150001, China
| | - Zhiyu Shi
- Department
of Cardiology, The First Affiliated Hospital
of Harbin Medical University, Harbin 150007, China
| |
Collapse
|
11
|
Liu X, Zhao Y, Wu X, Zhou Y, Liu Y, Wang S, Zhang Y, Yang H, Song F, Huang C. Spatiotemporally Programming Microenvironment to Recapitulate Endochondral Ossification via Greenhouse-Inspired Bionic Niche. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2504057. [PMID: 40317581 DOI: 10.1002/adma.202504057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/31/2025] [Indexed: 05/07/2025]
Abstract
Various biomaterials have been developed to address challenging critical-sized bone defects. However, most of them focus on intramembranous ossification (IMO) rather than endochondral ossification (ECO), often resulting in suboptimal therapeutic outcomes. Drawing inspiration from the functionality of the greenhouse ecosystem, herein a bionic niche is innovatively crafted to recapitulate the ECO process. This niche consists of three hierarchical components: an embedded microchannel network that facilitates cell infiltration and matter exchange, a polydopamine surface modification layer with immunomodulatory functions, and an ECO-targeted delivery system based on mesoporous silica nanoparticles. Through spatiotemporally programming of the microenvironment, the bionic niche effectively recapitulates the key stages of ECO. Notably, even in the rat calvaria, a region well-known for IMO, the bionic niche is capable of initiating ECO, evident by cartilage template formation, leading to efficient bone regeneration. Taken together, this study introduces prospective concepts for designing next-generation ECO-driven biomaterials for bone tissue engineering.
Collapse
Affiliation(s)
- Xuzheng Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yaning Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Xiaoyi Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yueli Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yingheng Liu
- Dental Materials Science, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong
| | - Shilei Wang
- Key Laboratory of Resources and Compound of Traditional Chinese Medicine, Ministry of Education, Hubei University of Traditional Chinese Medicine, Wuhan, 430065, China
| | - Yufeng Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Hongye Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Fangfang Song
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Cui Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
12
|
Summer M, Riaz S, Ali S, Noor Q, Ashraf R, Khan RRM. Understanding the Dual Role of Macrophages in Tumor Growth and Therapy: A Mechanistic Review. Chem Biodivers 2025; 22:e202402976. [PMID: 39869825 DOI: 10.1002/cbdv.202402976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/29/2025]
Abstract
Macrophages are heterogeneous cells that are the mediators of tissue homeostasis. These immune cells originated from monocytes and are classified into two basic categories, M1 and M2 macrophages. M1 macrophages exhibit anti-tumorous inflammatory reactions due to the behavior of phagocytosis. M2 macrophages or tumor-associated macrophages (TAMs) are the most abundant immune cells in the tumor microenvironment (TME) and have a basic role in tumor progression by interacting with other immune cells in TME. By the expression of various cytokines, chemokines, and growth factors, TAMs lead to strengthening tumor cell proliferation, angiogenesis, and suppression of the immune system which further support invasion and metastasis. This review discusses recent and updated mechanisms regarding tumor progression by M2 macrophages. Moreover, the current therapeutic approaches targeting TAMs, their advantages, and limitations are also summarized, and further treatment approaches are outlined along with an elaboration of the tumor regression role of macrophages. This comprehensive review article possibly helps to understand the mechanisms underlying the tumor progression and regression role of macrophages in a comparative way from a basic level to the advanced one.
Collapse
Affiliation(s)
- Muhammad Summer
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Saima Riaz
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Shaukat Ali
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Qudsia Noor
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Rimsha Ashraf
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Rana Rashad Mahmood Khan
- Faculty of Chemistry and Life Sciences, Department of Chemistry, Government College University Lahore, Lahore, Pakistan
| |
Collapse
|
13
|
Ma YX, Lei C, Ye T, Wan QQ, Wang KY, Zhu YN, Li L, Liu XF, Niu LZ, Tay FR, Mu Z, Jiao K, Niu LN. Silicon Enhances Functional Mitochondrial Transfer to Improve Neurovascularization in Diabetic Bone Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415459. [PMID: 40125794 DOI: 10.1002/advs.202415459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/15/2025] [Indexed: 03/25/2025]
Abstract
Diabetes mellitus is a metabolic disorder associated with an increased risk of fractures and delayed fracture healing, leading to a higher prevalence of bone defects. Recent advancements in strategies aim at regulating immune responses and enhancing neurovascularization have not met expectations. This study demonstrates that a silicon-based strategy significantly enhances vascularization and innervation, thereby optimizing the repair of diabetic bone defects. Silicon improves mitochondrial function and modulates mitochondrial fission dynamics in macrophages via the Drp1-Mff signaling pathway. Subsequently, functional mitochondria are transferred from macrophages to endothelial and neuronal cells through microvesicles, providing a protective mechanism for blood vessels and peripheral nerves during early wound healing. On this basis, an optimized strategy combining a silicified collagen scaffold with a Drp1-Fis1 interaction inhibitor is used to further regulate mitochondrial fission in macrophages and enhance the trafficking of functional mitochondria into stressed receptor cells. In diabetic mice with critical-sized calvarial defects, the silicon-based treatment significantly promotes vessel formation, nerve growth, and mineralized tissue development. These findings provide therapeutic insights into the role of silicon in promoting diabetic bone regeneration and highlight the importance of intercellular communication in diabetic conditions.
Collapse
Affiliation(s)
- Yu-Xuan Ma
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Chen Lei
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Tao Ye
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Qian-Qian Wan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Kai-Yan Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yi-Na Zhu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Ling Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xu-Fang Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Long-Zhang Niu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Franklin R Tay
- The Dental College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Zhao Mu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Kai Jiao
- Department of Stomatology, Tangdu hospital, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Li-Na Niu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
14
|
Teng Y, Zhang X, Song L, Yang J, Li D, Shi Z, Guo X, Wang S, Fan H, Jiang L, Hou S, Ramakrishna S, Lv Q, Shi J. Construction of anti-calcification small-diameter vascular grafts using decellularized extracellular matrix/poly (L-lactide-co-ε-caprolactone) and baicalin-cathepsin S inhibitor. Acta Biomater 2025; 197:184-201. [PMID: 40120837 DOI: 10.1016/j.actbio.2025.03.033] [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: 09/23/2024] [Revised: 03/07/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
The long-term transplantation of small-diameter vascular grafts (SDVGs) is associated with a risk of calcification, which is a key factor limiting the clinical translation of SDVG. Hence, there is an urgency attached to the development of new SDVGs with anti-calcification properties. Here, we used decellularized extracellular matrix (dECM) and poly (L-lactide-co-ε-caprolactone) (PLCL) as base materials and combined these with baicalin, cathepsin S (Cat S) inhibitor to prepare PBC-SDVGs by electrospinning. Baicalin contains carboxyl and hydroxyl groups that can interact with chemical groups in dECM powder, potentially blocking calcium nucleation sites. Cat S inhibitor prevents elastin degradation and further reduces the risk of calcification. PBC-SDVGs were biocompatible and when implanted in rat abdominal aorta, accelerated endothelialization, enhanced vascular tissue regeneration, inhibited elastin degradation, and promoted macrophage polarization M2 phenotype to regulate inflammation. After 3 months of implantation, the results of Doppler ultrasound, MicroCT, and histological staining revealed a significant reduction in calcification. In summary, the developed anti-calcification SDVGs offer a promising strategy for long-term implantation with significant clinical application potential. STATEMENT OF SIGNIFICANCE: The dECM and PLCL were used as base materials, connected with baicalin, and loaded with Cat S inhibitor to prepare PBC-SDVGs. The baicalin and dECM powder formed hydrogen bonds to crosslink together reducing the calcium deposition. In vitro, the vascular graft downregulated the expression level of osteogenic genes and promoted macrophage polarization toward an anti-inflammatory M2 phenotype, thereby reducing calcification. The PBC-SDVGs implanted in rat abdominal aorta can accelerate endothelialization, enhance vascular tissue regeneration, inhibit elastin degradation, reduce inflammation response and calcification.
Collapse
Affiliation(s)
- Yanjiao Teng
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China
| | - Xiaohai Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210000, PR China
| | - Lin Song
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210000, PR China
| | - Jianing Yang
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China
| | - Duo Li
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China
| | - Ziqi Shi
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China
| | - Xiaoqin Guo
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China
| | - Shufang Wang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Haojun Fan
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China
| | - Li Jiang
- Tianjin Eye Hospital, Nankai University Affiliated Eye Hospital, Tianjin 300021, PR China
| | - Shike Hou
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China.
| | - Seeram Ramakrishna
- Department of Mechanical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117575, Singapore.
| | - Qi Lv
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China.
| | - Jie Shi
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China.
| |
Collapse
|
15
|
Shi X, Askari Rizvi SF, Yang Y, Liu G. Emerging nanomedicines for macrophage-mediated cancer therapy. Biomaterials 2025; 316:123028. [PMID: 39693782 DOI: 10.1016/j.biomaterials.2024.123028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/22/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Tumor-associated macrophages (TAMs) contribute to tumor progression by promoting angiogenesis, remodeling the tumor extracellular matrix, inducing tumor invasion and metastasis, as well as immune evasion. Due to the high plasticity of TAMs, they can polarize into different phenotypes with distinct functions, which are primarily categorized as the pro-inflammatory, anti-tumor M1 type, and the anti-inflammatory, pro-tumor M2 type. Notably, anti-tumor macrophages not only directly phagocytize tumor cells, but also present tumor-specific antigens and activate adaptive immunity. Therefore, targeted regulation of TAMs to unleash their potential anti-tumor capabilities is crucial for improving the efficacy of cancer immunotherapy. Nanomedicine serves as a promising vehicle and can inherently interact with TAMs, hence, emerging as a new paradigm in cancer immunotherapy. Due to their controllable structures and properties, nanomedicines offer a plethora of advantages over conventional drugs, thus enhancing the balance between efficacy and toxicity. In this review, we provide an overview of the hallmarks of TAMs and discuss nanomedicines for targeting TAMs with a focus on inhibiting recruitment, depleting and reprogramming TAMs, enhancing phagocytosis, engineering macrophages, as well as targeting TAMs for tumor imaging. We also discuss the challenges and clinical potentials of nanomedicines for targeting TAMs, aiming to advance the exploitation of nanomedicine for cancer immunotherapy.
Collapse
Affiliation(s)
- Xueying Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China
| | - Syed Faheem Askari Rizvi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China; Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, 54000, Punjab, Pakistan
| | - Yinxian Yang
- School of Pharmaceutical Sciences, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| |
Collapse
|
16
|
Brigi C, Aghila Rani K, Selvakumar B, Hamad M, Abou Neel EA, Samsudin A. Decoding biomaterial-associated molecular patterns (BAMPs): influential players in bone graft-related foreign body reactions. PeerJ 2025; 13:e19299. [PMID: 40292103 PMCID: PMC12024449 DOI: 10.7717/peerj.19299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
Bone grafts frequently induce immune-mediated foreign body reactions (FBR), which hinder their clinical performance and result in failure. Understanding biomaterial-associated molecular patterns (BAMPs), including physicochemical properties of biomaterial, adsorbed serum proteins, and danger signals, is crucial for improving bone graft outcomes. Recent studies have investigated the role of BAMPs in the induction and maintenance of FBR, thereby advancing the understanding of FBR kinetics, triggers, stages, and key contributors. This review outlines the stages of FBR, the components of BAMPs, and their roles in immune activation. It also discusses various bone grafting biomaterials, their physicochemical properties influencing protein adsorption and macrophage modulation, and the key mechanisms of protein adsorption on biomaterial surfaces. Recent advancements in surface modifications and immunomodulatory strategies to mitigate FBR are also discussed. Furthermore, the authors look forward to future studies that will focus on a comprehensive proteomic analysis of adsorbed serum proteins, a crucial component of BAMPs, to identify proteins that promote or limit inflammation. This understanding could facilitate the design of biomaterials that selectively adsorb beneficial proteins, thereby reducing the risk of FBR and enhancing bone regeneration.
Collapse
Affiliation(s)
- Carel Brigi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
| | - K.G. Aghila Rani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
| | - Balachandar Selvakumar
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
| | - Mawieh Hamad
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
- Department of Medical Laboratory Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Ensanya Ali Abou Neel
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
- Department of Preventive and Restorative Dentistry, College of Dental Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - A.R. Samsudin
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
- Oral and Craniofacial Health Sciences Department, College of Dental Medicine, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
17
|
Zhu Z, Jin L, Wang Q, Shi H, Cheng K, Mao Z. Inhalable Ce Nanozyme-Backpacked Phage Aims at Ischemic Cerebral Injury by M1-Microglia Hitchhiking. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2419903. [PMID: 40231579 DOI: 10.1002/adma.202419903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 04/03/2025] [Indexed: 04/16/2025]
Abstract
There is a desperate need for precise nanomedications to treat ischemic cerebral injury. Yet, the drawbacks of poor delivery efficiency and off-target toxicity in pathologic parenchyma for traditional antioxidants against ischemic stroke result in inadequate brain accumulation. M13 bacteriophages are highly phagocytosed by M1-polarized microglia and can be carried toward the neuroinflammatory sites. Here, a bio-active, inhalable, Ce0.9Zr0.1O2-backpacked-M13 phage (abbreviated as CZM) is developed and demonstrates how M13 bacteriophages are taken up by different phenotypes' microglia. With the M1 microglia's proliferating and migrating, CZM can be extensively and specifically delivered to the site of the ischemic core and penumbra, where the surviving nerve cells need to be shielded from secondary oxidative stress and inflammatory cascade initiated by reactive oxygen species (ROS). With non-invasive administration, CZM effectively alleviates oxidative damage and apoptosis of neurons by eliminating ROS generated by hyperactive M1-polarized microglia. Here, a secure and effective strategy for the targeted therapy of neuroinflammatory maladies is offered by this research.
Collapse
Affiliation(s)
- Zhixin Zhu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Lulu Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Qiaoxuan Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Haifei Shi
- Department of Orthopedics, 1st Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 31000, China
| | - Ke Cheng
- Department of Biomedical Engineering, Columbia University, New York, NY, 10032, USA
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
| |
Collapse
|
18
|
Ren S, Lv H, Chen S, Zhou J, Chen S, Chen J, Luo J, Guo Y, Wang H, Zhai J, Zhou Y. Photoresponsive Blood-Derived Protein Hydrogels Packed with Bioactive Carbon Dots Modulate Mitochondrial Homeostasis and Reprogram Metabolism for Chronic Wound Healing in Diabetes. ACS APPLIED MATERIALS & INTERFACES 2025; 17:20885-20900. [PMID: 40148098 DOI: 10.1021/acsami.5c00635] [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: 03/29/2025]
Abstract
Autologous platelet concentrates (APC) represent a class of personalized regenerative materials for vascularized tissue regeneration. However, shortcomings including poor controllability of gel formation, lack of reactive oxygen species (ROS) scavenging ability, and deficient anti-inflammatory capacity restrict the tissue healing outcomes of APC. This study proposes an APC-based synergistic platform (CurCDs@iPRF-MA) for the treatment of chronic wounds in diabetes. Such a platform is composed of injectable platelet-rich fibrin (iPRF), gelatin methacryloyl (GelMA), and a carbogenic nanodrug from curcumin (CurCDs) that is injectable before the light-induced gel formation process, greatly facilitating the clinical applications of APC. Significantly, CurCDs@iPRF-MA can modulate the mitochondrial homeostasis under inflammatory conditions, activate the oxidative phosphorylation (OXPHOS) program, and regulate the diabetic microenvironment through metabolic reprogramming to achieve macrophage phenotype regulation and ROS elimination, as well as promote vascularization by releasing autologous growth factors, dramatically improving the healing efficacy of the chronic wounds in diabetes. This study offers a practical and effective approach to developing spatiotemporally controllable and multifunctional APC-based hydrogels for highly effective tissue regeneration.
Collapse
Affiliation(s)
- Sicong Ren
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Changchun 130021, Jilin, China
| | - Huixin Lv
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Changchun 130021, Jilin, China
| | - Sheng Chen
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Changchun 130021, Jilin, China
| | - Jing Zhou
- School of Stomatology, Jilin University, Changchun 130021, Jilin, China
| | - Siyu Chen
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Changchun 130021, Jilin, China
| | - Jingxia Chen
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Changchun 130021, Jilin, China
| | - Jiaxin Luo
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Changchun 130021, Jilin, China
| | - Yuanxin Guo
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Changchun 130021, Jilin, China
| | - Huan Wang
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Jingjie Zhai
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Changchun 130021, Jilin, China
| | - Yanmin Zhou
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Changchun 130021, Jilin, China
| |
Collapse
|
19
|
Su Y, Ju J, Shen C, Li Y, Yang W, Luo X, Wang Z, Zeng J, Liu L. In situ 3D bioprinted GDMA/Prussian blue nanozyme hydrogel with wet adhesion promotes macrophage phenotype modulation and intestinal defect repair. Mater Today Bio 2025; 31:101636. [PMID: 40161927 PMCID: PMC11950758 DOI: 10.1016/j.mtbio.2025.101636] [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: 12/21/2024] [Revised: 02/20/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Developing hydrogels with wet-adhesion, immunomodulation and regenerative repair capabilities in intestinal repair remains a formidable challenge. In the present study, the development of an anti-inflammatory, wet-adhesive, decellularized extracellular matrix hydrogel produced using three-dimensional (3D) -printing technology is presented. This hydrogel, which integrates gelatin and dopamine, was demonstrated to display excellent wet-adhesion properties, fully harnessing the outstanding regenerative potential of the decellularized small-intestine matrix. Furthermore, the integration of Prussian Blue nanozymes imparted significant anti-inflammatory and antioxidant properties. Through modulating macrophage polarization, the hydrogel was not only found to enhance tissue repair, but also to substantially mitigate inflammation. In vivo experiments (namely, histopathological analyses using a rat model) demonstrated that this hydrogel was able to effectively enhance tissue regeneration and healing in models of intestinal damage. In conclusion, through the utilization of 3D-printing technology, the present study has shown that the precise manufacturing and customization of the hydrogel to various shapes and sizes of intestinal defects may be executed, thereby providing an innovative strategy for intestinal repair. This advanced hydrogel has therefore been shown to hold significant promise as a bioadhesive for both emergency repair and regenerative therapy.
Collapse
Affiliation(s)
- Yang Su
- Department of Gastrointestinal Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Molecular Medicine center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jingyi Ju
- Department of Plastic Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chentao Shen
- Department of Plastic Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yanqi Li
- Department of Gastrointestinal Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Molecular Medicine center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wangshuo Yang
- Department of Gastrointestinal Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Molecular Medicine center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xuelai Luo
- Department of Gastrointestinal Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Molecular Medicine center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhenxing Wang
- Department of Plastic Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jinhao Zeng
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Lu Liu
- Department of Gastrointestinal Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Molecular Medicine center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
20
|
Liu D, Chen J, Zhou J, Dai J, Qin H, Wan G, Qian J, Li P, Li J. Macrophage-related inflammatory responses to degradation products of biodegradable molybdenum implants. Mater Today Bio 2025; 31:101519. [PMID: 39990739 PMCID: PMC11846932 DOI: 10.1016/j.mtbio.2025.101519] [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: 11/12/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 02/25/2025] Open
Abstract
Metallic molybdenum (Mo) has been increasingly recognized as a potential biodegradable metal for biomedical implants. However, the macrophage-mediated inflammatory responses to Mo-based implants remain underexplored. This study examined the in vitro inflammatory reactions of macrophages to the degradation products of biodegradable Mo implants. The short-term and long-term biodegradation behavior and the subsequent impact on cytotoxicity, metabolism, and macrophage polarization were assessed. Both Mo and its degradation products were shown to be non-toxic within macrophage tolerance limits. Nevertheless, morphological changes and pro-inflammatory polarization were observed in cells around Mo-based specimen. Notably, matrix metalloproteinase 9 (Mmp9) was identified as a key gene influencing macrophage polarization in proximity to Mo. Additionally, pre-treating the Mo specimens in culture medium for 24 h significantly mitigated its stimulatory effects on cells. These results demonstrated the significance of optimizing Mo pre-treatment methods to prevent localized inflammation associated with its degradation. Specifically, pre-treatment of Mo can effectively mitigate the adverse impacts of its early degradation on macrophages and the surrounding immune environment. Our research into these early degradation phases introduces new avenues for studying molybdenum's immunomodulatory properties, potentially through precise control of its release and the targeted expression of pivotal genes.
Collapse
Affiliation(s)
- Danyang Liu
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou, 510182, PR China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, 510182, PR China
| | - Jiahao Chen
- Department of Prosthodontics, Geriatric Dentistry and Craniomandibular Disorders, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Aßmannshauser Str. 4-6, 14197, Berlin, Germany
| | - Jiannan Zhou
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou, 510182, PR China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, 510182, PR China
| | - Jingtao Dai
- Department of Orthodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510515, PR China
| | - Haotian Qin
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, PR China
| | - Guojiang Wan
- Institute of Biomedical Engineering, College of Medicine, Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, 610031, PR China
| | - Junyu Qian
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, PR China
| | - Ping Li
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou, 510182, PR China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, 510182, PR China
| | - Jiang Li
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou, 510182, PR China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, 510182, PR China
| |
Collapse
|
21
|
Chen W, Pan Y, Chu CH, Dong S, Wang M, Wang L, Wang L, Lin X, Tang C. Microenvironment-responsive nanoparticles functionalized titanium implants mediate redox balance and immunomodulation for enhanced osseointegration. Mater Today Bio 2025; 31:101628. [PMID: 40124346 PMCID: PMC11930443 DOI: 10.1016/j.mtbio.2025.101628] [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: 12/08/2024] [Revised: 02/27/2025] [Accepted: 03/01/2025] [Indexed: 03/25/2025] Open
Abstract
Various pathological conditions (e.g., diabetes, osteoporosis) are accompanied by persistent oxidative stress, which compromises the immune microenvironment and poses substantial challenges for osseointegration. Reactive oxygen species (ROS) play a "double-edged sword" role in bone tissue. Therefore, developing responsive biomaterials to maintain redox balance dynamically is crucial for enhanced osseointegration. Herein, the microenvironment-responsive coordination nanoparticles (C-Ca-SalB NPs) composed of salvianolic acid B (SalB), catechol-conjugated chitosan (CS-C), and Ca2+ are constructed and further covalently immobilized onto titanium implant surfaces. The resulting implants achieve on-demand antioxidant and immunomodulatory effects in a microenvironment-responsive manner, thus facilitating bone regeneration under both normal and oxidative conditions. Under physiological conditions, the functionalized implants display modest immunomodulatory properties without affecting oxidative balance, while C-Ca-SalB NPs remain relatively stable. However, the modified implants enable rapid decomposition of C-Ca-SalB NPs under acidic oxidative conditions, displaying robust ROS-scavenging, anti-inflammatory, and osteoinductive capacities, ultimately remodeling the pathological microenvironment into a regenerative one. Overall, smart implants with controlled bioactive agent release in this study present a comprehensive solution for enhancing bone-implant integration, particularly in the challenging context of oxidative stress.
Collapse
Affiliation(s)
- Wei Chen
- Department of Oral Implantology, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
- State Key Laboratory Cultivation Base of Research, Prevention, and Treatment for Oral Diseases, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210029, China
| | - Yifei Pan
- Department of Oral Implantology, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
- State Key Laboratory Cultivation Base of Research, Prevention, and Treatment for Oral Diseases, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210029, China
| | - Catherine Huihan Chu
- Department of Orthodontic, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Shuo Dong
- Department of Oral Implantology, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
- State Key Laboratory Cultivation Base of Research, Prevention, and Treatment for Oral Diseases, Nanjing, 210029, China
| | - Mingxi Wang
- Department of Oral Implantology, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Long Wang
- Department of Oral Implantology, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Lingxu Wang
- Department of Oral Implantology, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xuyang Lin
- Department of Oral Implantology, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Chunbo Tang
- Department of Oral Implantology, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
- State Key Laboratory Cultivation Base of Research, Prevention, and Treatment for Oral Diseases, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210029, China
| |
Collapse
|
22
|
Zhang H, Hua S, He C, Yin M, Qin J, Liu H, Zhou H, Wu S, Yu X, Jiang H, Wang Y, Qian Y. Application of 4D-Printed Magnetoresponsive FOGS Hydrogel Scaffolds in Auricular Cartilage Regeneration. Adv Healthc Mater 2025; 14:e2404488. [PMID: 39955711 DOI: 10.1002/adhm.202404488] [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: 11/13/2024] [Revised: 02/01/2025] [Indexed: 02/17/2025]
Abstract
3D-printed hydrogel scaffolds are widely utilized in auricular cartilage tissue engineering. However, issues such as graft-related inflammation, poor mechanical properties, and the lack of external modulation of 3D-printed scaffolds in vivo have raised significant concerns. To address these challenges, a "fried egg" structure is designed, consisting of chitosan-coated ferroferric oxide magnetic nanoparticles (Fe3O4@CS MNPs), which are uniformly incorporated into hydrogel. Through 4D printing technology, magnetoresponsive hydrogel scaffolds are constructed to overcome the aforementioned limitations. The results demonstrated that, compared to 3D printing, 4D-printed magnetic hydrogel scaffolds significantly enhanced cartilage tissue regeneration in both in vitro and in vivo environments when subjected to an external magnetic field (MF). Furthermore, the mechanical strength of regenerated cartilage approached to that of natural cartilage. The chitosan coating on the surface of MNPs exhibited anti-inflammatory and antibacterial properties, promoting M2 polarization of macrophages and suppressing graft-related inflammation and bacteria. Transcriptomic analysis confirmed that MNPs modulate macrophage immunity by activating JAK2/STAT3 signaling pathway. Taken together, a magnetoresponsive multifunctional scaffold is designed that can be externally controlled by magnetic fields to promote ear cartilage tissue regeneration. The regenerated cartilage exhibits excellent biocompatibility, anti-inflammatory, antibacterial properties, and mechanical performance, providing new insights for auricular cartilage tissue engineering.
Collapse
Affiliation(s)
- Hongyi Zhang
- Department of Plastic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Medical College, Tongji University, Shanghai, 200331, China
| | - Shan Hua
- Department of Plastic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Medical College, Tongji University, Shanghai, 200331, China
| | - Chenlong He
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
| | - Ming Yin
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
| | - Jingwen Qin
- The Institute for Translational Nanomedicine, Shanghai East Hospital, the Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200092, P. R. China
| | - Huawei Liu
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
| | - Han Zhou
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
| | - Shengming Wu
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
| | - Xingge Yu
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200135, P. R. China
| | - Hua Jiang
- Department of Plastic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Medical College, Tongji University, Shanghai, 200331, China
| | - Yilong Wang
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
| | - Yuxin Qian
- Department of Plastic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Medical College, Tongji University, Shanghai, 200331, China
| |
Collapse
|
23
|
Yan Z, Sun T, Zeng J, He T, He Y, Xu D, Liu R, Tan W, Zang X, Yan J, Deng Y. Enhanced Immune Modulation and Bone Tissue Regeneration through an Intelligent Magnetic Scaffold Targeting Macrophage Mitochondria. Adv Healthc Mater 2025; 14:e2500163. [PMID: 40095440 DOI: 10.1002/adhm.202500163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/28/2025] [Indexed: 03/19/2025]
Abstract
During the bone tissue repair process, the highly dynamic interactions between the host and materials hinder precise, stable, and sustained immune modulation. Regulating the immune response based on potential mechanisms of macrophage phenotypic changes may represent an effective strategy for promoting bone healing. This study successfully constructs a co-dispersed pFe₃O₄-MXene nanosystem by loading positively charged magnetite (pFe₃O₄) nanoparticles onto MXene nanosheets using electrostatic self-assembly. Subsequently, this work fabricates a biomimetic porous bone scaffold (PFM) via selective laser sintering, which exhibit superior magnetic properties, mechanical performance, hydrophilicity, and biocompatibility. Further investigations demonstrate that the PFM scaffold could precisely and remotely modulate macrophage polarization toward the M2 phenotype under a static magnetic field, significantly enhancing osteogenesis and angiogenesis. Proteomic analysis reveal that the scaffold upregulates Arg2 expression, enhancing mitochondrial function and accelerating oxidative phosphorylation, thereby inducing the M2 transition. In vivo experiments validated the scaffold's immune regulatory capacity in subcutaneous and cranial defect repairs in rats, effectively promoting new bone formation. Overall, this strategy of immune modulation targeting macrophage metabolism and mitochondrial function offers novel insights for material design in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Zuyun Yan
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Tianshi Sun
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Jin Zeng
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Tao He
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Yiwen He
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Dongcheng Xu
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Renfeng Liu
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Wei Tan
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Xiaofang Zang
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Jinpeng Yan
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, 410017, P. R. China
| | - Youwen Deng
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| |
Collapse
|
24
|
Guan Y, Zhang M, Song J, Negrete M, Adcock T, Kandel R, Racioppi L, Gerecht S. CaMKK2 Regulates Macrophage Polarization Induced by Matrix Stiffness: Implications for Shaping the Immune Response in Stiffened Tissues. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417778. [PMID: 40036145 PMCID: PMC12021110 DOI: 10.1002/advs.202417778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/13/2025] [Indexed: 03/06/2025]
Abstract
Macrophages are essential for immune responses and maintaining tissue homeostasis, exhibiting a wide range of phenotypes depending on their microenvironment. The extracellular matrix (ECM) is a vital component that provides structural support and organization to tissues, with matrix stiffness acting as a key regulator of macrophage behavior. Using physiologically relevant 3D stiffening hydrogel models, it is found that increased matrix stiffness alone promoted macrophage polarization toward a pro-regenerative phenotype, mimicking the effect of interleukin-4(IL-4) in softer matrices. Blocking Calcium/calmodulin-dependent kinase kinase 2 (CaMKK2) selectively inhibited stiffness-induced macrophage polarization without affecting IL-4-driven pro-regenerative pathways. In functional studies, CaMKK2 deletion prevented M2-like/pro-tumoral polarization caused by matrix stiffening, which in turn hindered tumor growth. In a murine wound healing model, loss of CaMKK2 impaired matrix stiffness-mediated macrophage accumulation, ultimately disrupting vascularization. These findings highlight the critical role of CaMKK2 in the macrophage mechanosensitive fate determination and gene expression program, positioning this kinase as a promising therapeutic target to selectively modulate macrophage responses in pathologically stiff tissues.
Collapse
Affiliation(s)
- Ya Guan
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Min Zhang
- Division of Hematological Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27708USA
| | - Jiyeon Song
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Marcos Negrete
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Tyler Adcock
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Reeva Kandel
- Division of Hematological Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27708USA
| | - Luigi Racioppi
- Division of Hematological Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27708USA
- Department of Molecular Medicine and Medical BiotechnologyUniversity of Naples Federico IINaplesItaly
| | - Sharon Gerecht
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| |
Collapse
|
25
|
Chen Y, Feng D, Cheng Y, Jiang X, Qiu L, Zhang L, Shi D, Wang J. Research progress of metal-CpG composite nanoadjuvants in tumor immunotherapy. Biomater Sci 2025; 13:1605-1623. [PMID: 39998438 DOI: 10.1039/d4bm01399a] [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: 02/26/2025]
Abstract
The practical benefits and therapeutic potential of tumor vaccines in immunotherapy have drawn significant attention in the field of cancer treatment. Among the available vaccines, nanovaccines that utilize nanoparticles as carriers or adjuvants have demonstrated considerable effectiveness in combating cancer. Cytosine-phosphate-guanine oligodeoxynucleotide (CpG ODN), a common adjuvant in tumor nanovaccines, activates both humoral and cellular immunity by recognizing toll-like receptor 9 (TLR9), thereby aiding in the prevention and treatment of cancer. Metal nanoparticles hold great promise in tumor immunotherapy due to their adjustable size, surface functionalization, ability to regulate innate immunity, and capacity for controlled delivery of antigens or immunomodulators. Consequently, composite nanoadjuvants, formed by combining metal nanoparticles with CpG ODNs, can be customized to meet the specific performance requirements of different application scenarios, effectively overcoming the limitations of conventional immunotherapy approaches. This review provides a comprehensive analysis of the critical role of metal-CpG composite nanoadjuvants in advancing vaccine adjuvants for cancer therapy and prevention, highlighting their efficacy in preclinical settings.
Collapse
Affiliation(s)
- Yifan Chen
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| | - Danna Feng
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| | - Yilin Cheng
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| | - Xianmeng Jiang
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| | - Lin Qiu
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| | - Li Zhang
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Dongjian Shi
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Jianhao Wang
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| |
Collapse
|
26
|
Yu H, Wei D, Liao W, Shang X, Li D, Liu C, Deng Q, Huangfu H. Exosome-mediated effects of BRCA1 on cardiovascular artery disease. Cell Biol Toxicol 2025; 41:59. [PMID: 40080209 PMCID: PMC11906578 DOI: 10.1007/s10565-025-09996-4] [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: 05/20/2024] [Accepted: 01/28/2025] [Indexed: 03/15/2025]
Abstract
The progression of coronary artery disease atherosclerosis (CAD) is closely associated with cardiomyocyte apoptosis and inflammatory responses. This study focused on investigating the impact of BRCA1 in exosomes (Exo) derived from M1 macrophages on CAD. Through the analysis of single-cell RNA-seq datasets, significant communication between macrophages and cardiomyocytes in CAD patients was observed. BRCA1, identified as a significant apoptosis-related gene, was pinpointed through the assessment of differential gene expression and weighted gene co-expression network analysis (WGCNA). Experimental procedures involved BRCA1 lentivirus transfection of M1 macrophages, isolation of Exo for application to cardiomyocytes and smooth muscle cells, cell viability assessments, and characterization of Exo. The results showed that BRCA1-Exo from M1 macrophages induced cardiomyocyte apoptosis and affected smooth muscle cell behavior. In vivo studies further supported the exacerbating effects of BRCA1-Exo on CAD progression. Overall, the involvement of Exo carrying BRCA1 from M1 macrophages is evident in the induction of cardiomyocyte apoptosis and the regulation of smooth muscle cell behaviors, thereby contributing to CAD atherosclerosis progression. These findings unveil novel molecular targets that could have potential implications for CAD treatment strategies.
Collapse
Affiliation(s)
- Hairui Yu
- Department of Preventive Medicine, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Shenzhen, 518000, China
| | - Dong Wei
- Department of Preventive Medicine, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Shenzhen, 518000, China
| | - Weiqian Liao
- Department of Cardiology, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Shenzhen, 518000, China
| | - Xiaoming Shang
- Department of Cardiology, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Shenzhen, 518000, China
| | - Dandan Li
- Department of Cardiology, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Shenzhen, 518000, China
| | - Chunzhao Liu
- Department of Preventive Medicine, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Shenzhen, 518000, China
| | - Qimei Deng
- Department of Preventive Medicine, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Shenzhen, 518000, China
| | - Haiquan Huangfu
- Department of Cardiology, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Shenzhen, 518000, China.
| |
Collapse
|
27
|
Jiang R, Fang Q, Liu W, Chen L, Yang H. Recent Progress in Radiosensitive Nanomaterials for Radiotherapy-Triggered Drug Release. ACS APPLIED MATERIALS & INTERFACES 2025; 17:14801-14821. [PMID: 40014050 DOI: 10.1021/acsami.4c23023] [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: 02/28/2025]
Abstract
Benefiting from the unique properties of ionizing radiation, such as high tissue penetration, spatiotemporal resolution, and clinical relevance compared with other external stimuli, radiotherapy-induced drug release strategies are showing great promise in developing effective and personalized cancer treatments. However, the requirement of high doses of X-ray irradiation to break chemical bonds for drug release limits the application of radiotherapy-induced prodrug activation in clinics. Recent advances in nanomaterials offer a promising approach for radiotherapy sensitization as well as integrating multiple modalities for improved therapy outcomes. In particular, the catalytic radiosensitization that utilizes electrons and energy generated by nanomaterials upon X-ray irradiation has demonstrated excellent potential for enhanced radiotherapy. In this Review, we summarize the design principles of X-ray-responsive chemical bonds for controlled drug release, strategies for catalytic radiosensitization, and recent progress of X-ray-responsive nanoradiosensitizers for enhanced radiotherapy by integration with chemotherapy, chemodynamic therapy, photodynamic therapy, photothermal therapy, gas therapy, and immunotherapy. Finally, we discuss the challenges of X-ray-responsive nanoradiosensitizers heading toward possible clinical translation. We expect that emerging strategies based on radiotherapy-triggered drug release will facilitate a frontier in accurate and effective cancer therapy in the near future.
Collapse
Affiliation(s)
- Renfeng Jiang
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Qiong Fang
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Wenjun Liu
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Lanlan Chen
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Huanghao Yang
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| |
Collapse
|
28
|
Zhang N, Yang G, Wu Y, Hu L, Zhao C, Liu HH, Wu L, Pan J, Liu X. Controlled Release of Cold Atmospheric Plasma by Gelatin Scaffold Enhances Wound Healing via Macrophage Modulation. ACS APPLIED MATERIALS & INTERFACES 2025; 17:15050-15066. [PMID: 40013441 DOI: 10.1021/acsami.4c21635] [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: 02/28/2025]
Abstract
Cold atmospheric plasma (CAP) has emerged as a promising therapeutic tool for wound healing and tissue regeneration. However, the clinical application of CAP faces challenges, particularly the need for repeated treatments to maintain therapeutic efficacy and the difficulty in controlling the release of reactive oxygen and nitrogen species (ROS/RNS), which can cause tissue damage at high concentrations. In this study, we developed a controlled-release system by integrating CAP with a skin bionics gelatin scaffold (CAP-GS), enabling the sustained, localized release of CAP without the need for repeated applications. Our results demonstrate that CAP-GS significantly accelerates skin wound healing by modulating the immune microenvironment and reducing inflammation. The gelatin scaffold effectively regulates the release of ROS/RNS, maintaining levels conducive to M2 macrophage polarization and minimizing oxidative damage. Furthermore, CAP-GS enhances macrophage metabolism, including oxidative phosphorylation (OXPHOS) and fatty acid oxidation (FAO), metabolic pathways characteristic of M2 polarization. The increased secretion of growth factors by CAP-GS-treated macrophages contributes to cell proliferation, migration, and tissue regeneration. These findings highlight CAP-GS as a promising, clinically applicable system for wound healing and tissue repair, overcoming the limitations of traditional CAP treatments and offering an approach to regenerative medicine.
Collapse
Affiliation(s)
- Nian 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, Sichuan 610041, China
| | - Guang Yang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Yan Wu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Liru Hu
- 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, Sichuan 610041, China
| | - Chengzhi Zhao
- 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, Sichuan 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, Sichuan 610041, China
| | - Li Wu
- College of Electronics and Information Engineering, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jian Pan
- 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, Sichuan 610041, China
| | - Xian 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, Sichuan 610041, China
| |
Collapse
|
29
|
Zheng S, Sun X, Chen K, Zhang M, Zou C, Wang L, Guo Z, Jin Z, Ma Z, Li G, Wu G. Metal-Phenolic Modified Coaxial Electrospun Biomembrane Combined with the Photothermal Effect Enhances Bone Regeneration by Ameliorating Oxidative Stress and Mitochondrial Dysfunction via the PI3K/Akt Signaling Pathway. ACS APPLIED MATERIALS & INTERFACES 2025; 17:15019-15034. [PMID: 40016904 DOI: 10.1021/acsami.4c21265] [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: 03/01/2025]
Abstract
Critical-sized bone defect regeneration remains a significant clinical challenge due to the complex cascade of biological processes involved. To address this, we developed a sophisticated hierarchical biomembrane (PCS@MPN10) designed to modulate the osteogenic microenvironment. Using coaxial electrospinning, we fabricated a core-shell structure with polylactic acid (PLA) as the membrane base, incorporating simvastatin in the core and chitosan in the shell. The membrane surface was further modified with a tannic acid-iron metal-polyphenol network coating. Our results demonstrated that the biomembrane exhibits excellent biocompatibility, photothermal properties, and significant antibacterial activity. Additionally, the membrane regulates the microenvironment by promoting M1-to-M2 macrophage polarization, showing strong osteogenic potential both in vitro and in vivo. Furthermore, PCS@MPN10+NIR modulates mitochondrial function through the PI3K-AKT pathway, clears mitochondrial reactive oxygen species (ROS), and alleviates cellular oxidative stress, thereby enhancing bone regeneration. Overall, these findings suggest that this biomembrane holds great promise as a strategy for improving bone regeneration in critical-sized defects.
Collapse
Affiliation(s)
- Shikang Zheng
- Department of Oral, Plastic, and Aesthetic Surgery, The Bethune Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, China
| | - Xiumei Sun
- Department of Orthodontics, Hospital of Stomatology, The Bethune Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, China
| | - Kai Chen
- Department of Oral, Plastic, and Aesthetic Surgery, The Bethune Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, China
| | - Mingjun Zhang
- Department of Oral, Plastic, and Aesthetic Surgery, The Bethune Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, China
| | - Chentong Zou
- Department of Oral, Plastic, and Aesthetic Surgery, The Bethune Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, China
| | - Lin Wang
- Department of Oral, Plastic, and Aesthetic Surgery, The Bethune Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, China
| | - Zhipeng Guo
- Faculty of Materials Science, Shenzhen MSU-BIT University, Shenzhen 518172, P. R. China
| | - Zhaoyi Jin
- Department of Oral, Plastic, and Aesthetic Surgery, The Bethune Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, China
| | - Ziyi Ma
- Department of Oral, Plastic, and Aesthetic Surgery, The Bethune Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, China
| | - Guanyu Li
- Department of Oral, Plastic, and Aesthetic Surgery, The Bethune Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, China
| | - Guomin Wu
- Department of Oral, Plastic, and Aesthetic Surgery, The Bethune Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
30
|
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
|
31
|
Lou T, Wang X, Li J, Wang W, Han P, Yu S, Fan C, Zhou C, Ruan H. Chirality Regulates Bone Regeneration through Mechanoresponse and Immunoregulation. ACS NANO 2025; 19:7767-7783. [PMID: 39967389 DOI: 10.1021/acsnano.4c13164] [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: 02/20/2025]
Abstract
As an omnipresent occurrence in the natural world, chirality plays a crucial role in numerous biological and physiological processes. Therefore, incorporation of chirality into biointerface materials has been emerging as a research hotspot in the development of regenerative biomaterials. Nevertheless, how chiral biointerface materials interact with biological organisms remains poorly understood. In the current study, a bioinspired chiral self-assembled nanohydroxyapatite material is developed for bone regeneration using a distraction osteogenesis model. We found that the left-handed chirality, rather than the right-handed chirality, possessed the greatest heightened ability for bone regeneration. Molecular mechanism research reveals that the left-handed chirality can activate mechanosensitive pathways to promote Ca2+ influx, subsequently enhancing macrophage type-2 polarization through phosphorylation of signal transducer and activator of transcription (STAT6), which eventually stimulates bone regeneration by harnessing the synergistic effects of angiogenesis and osteogenesis. These findings provide additional insights into the underlying mechanism of chiral recognition between biological systems and biointerface materials, offering alternative pathways for the development of biomaterials.
Collapse
Affiliation(s)
- Tengfei Lou
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xu Wang
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai 200233, China
| | - Juehong Li
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Wei Wang
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Pei Han
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Shiyang Yu
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Cunyi Fan
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Chao Zhou
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 201306, China
| | - Hongjiang Ruan
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| |
Collapse
|
32
|
Li XL, Zhao YQ, Miao L, An YX, Wu F, Han JY, Han JY, Tay FR, Mu Z, Jiao Y, Wang J. Strategies for promoting neurovascularization in bone regeneration. Mil Med Res 2025; 12:9. [PMID: 40025573 PMCID: PMC11874146 DOI: 10.1186/s40779-025-00596-1] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 01/26/2025] [Indexed: 03/04/2025] Open
Abstract
Bone tissue relies on the intricate interplay between blood vessels and nerve fibers, both are essential for many physiological and pathological processes of the skeletal system. Blood vessels provide the necessary oxygen and nutrients to nerve and bone tissues, and remove metabolic waste. Concomitantly, nerve fibers precede blood vessels during growth, promote vascularization, and influence bone cells by secreting neurotransmitters to stimulate osteogenesis. Despite the critical roles of both components, current biomaterials generally focus on enhancing intraosseous blood vessel repair, while often neglecting the contribution of nerves. Understanding the distribution and main functions of blood vessels and nerve fibers in bone is crucial for developing effective biomaterials for bone tissue engineering. This review first explores the anatomy of intraosseous blood vessels and nerve fibers, highlighting their vital roles in bone embryonic development, metabolism, and repair. It covers innovative bone regeneration strategies directed at accelerating the intrabony neurovascular system over the past 10 years. The issues covered included material properties (stiffness, surface topography, pore structures, conductivity, and piezoelectricity) and acellular biological factors [neurotrophins, peptides, ribonucleic acids (RNAs), inorganic ions, and exosomes]. Major challenges encountered by neurovascularized materials during their clinical translation have also been highlighted. Furthermore, the review discusses future research directions and potential developments aimed at producing bone repair materials that more accurately mimic the natural healing processes of bone tissue. This review will serve as a valuable reference for researchers and clinicians in developing novel neurovascularized biomaterials and accelerating their translation into clinical practice. By bridging the gap between experimental research and practical application, these advancements have the potential to transform the treatment of bone defects and significantly improve the quality of life for patients with bone-related conditions.
Collapse
Affiliation(s)
- Xin-Ling Li
- 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, China
| | - Yu-Qing 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, China
| | - Li Miao
- Department of Stomatology, The Seventh Medical Center of PLA General Hospital, Beijing, 100700, China
| | - Yan-Xin An
- Department of General Surgery, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, 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, China
| | - Jin-Yu Han
- 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, China
| | - Jing-Yuan Han
- 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, China
| | - Franklin R Tay
- Graduate School of Augusta University, Augusta, GA, 30912, USA
| | - Zhao Mu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Yang Jiao
- Department of Stomatology, The Seventh Medical Center of PLA General Hospital, Beijing, 100700, 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, China.
| |
Collapse
|
33
|
Wang W, Wu H, Zhang X, Hong Y, Tao S, Cao X, Wang S, Zha L, Zha Z. Whole-Component Antigen Nanovaccines Combined With aTIGIT for Enhanced Innate and Adaptive Anti-tumor Immunity. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2412800. [PMID: 39967373 DOI: 10.1002/smll.202412800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/27/2025] [Indexed: 02/20/2025]
Abstract
Using entire tumor cells or tissues that display both common and patient-specific antigens can potentially trigger a comprehensive and long-lasting anti-tumor immune response. However, the limited immunogenicity, low uptake efficiency, and susceptibility to degradation of whole-component antigens present significant challenges. In this study, we employed tumor lysates (TLs) as whole-component antigens, in conjunction with MgAl-layered double hydroxide (MA) as nanoadjuvants and Mn2+ as immunostimulants, to create personalized MMAT (Mn2+-MA-TLs) nanovaccines. After subcutaneous injection of MMAT nanovaccines, the high local concentrations of TLs and Mn2+ facilitated the recruitment and activation of antigen-presenting cells (APCs), thereby inducing a robust adaptive immune response. Remarkably, MMAT nanovaccines enabled lysosomal escape, enhanced antigen cross-presentation, and activated the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway in APCs. Furthermore, MMAT nanovaccines, when combined with the anti-TIGIT monoclonal antibody (aTIGIT), an immune checkpoint inhibitor, not only stimulated T-cell-based adaptive anti-tumor immune responses but also activated the NK-cell-based innate anti-tumor immunity, effectively suppressing tumor growth, recurrence, and metastasis. Thus, the ternary MMAT nanovaccines developed here introduced a pioneered paradigm for the rapid preparation of whole-component tumor antigens with nanoadjuvants and immunostimulants into nanovaccines, offering new prospects for clinical immunotherapies.
Collapse
Affiliation(s)
- Weitao Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Haitao Wu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Xuan Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Yang Hong
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Shi Tao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Xiangjing Cao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Shipeng Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Lisha Zha
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
- School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Zhengbao Zha
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| |
Collapse
|
34
|
Wang Z, Chu Y, Du J, Hu Y, Wang H, Liu H, Yang C, Wang M, Ran J, Yu A. Accelerating repair of infected bone defects through post-reinforced injectable hydrogel mediated antibacterial/immunoregulatory microenvironment at bone-hydrogel interface. Carbohydr Polym 2025; 351:123082. [PMID: 39779005 DOI: 10.1016/j.carbpol.2024.123082] [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: 09/14/2024] [Revised: 11/04/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025]
Abstract
Functional injectable hydrogel (IH) is promising for infected bone defects (IBDs) repair, but how to endow it with desired antibacterial/immunoregulatory functions as well as avoid mechanical failures during its manipulation has posed as main challenges. Herein, rosmarinic acid (RosA), a natural product with antibacterial/immunoregulatory activities, was utilized to develop a FCR IH through forming phenylboronic acid ester bonds with 4-formylphenyl phenylboronic acid (4-FPBA) grafted chitosan (CS) (FC). After being applied to the IBD site, the FCR IH was then injected with tobramycin (Tob) solution, another alkaline antibacterial drug, to induce in situ crystallization of the FC, endowing the resultant FCRT hydrogel with adaptively enhanced mechanical strength and structural stability. Owing to the specific structural composition, the FCRT hydrogel could sustainedly release Tob and RosA molecules at the IBD interface, effectively eliminating in situ bacterial infection. In addition, the released RosA molecules also induced the M2 polarization of in situ macrophages (Mφ), which was identified to be related to the NF-κB and PI3K-AKT pathways, therefore promoting the osteogenic differentiation of in situ bone marrow stromal cells (BMSCs). Due to the simultaneous antibacterial/osteo-immunoregulatory microenvironment at the IBD interface, the repair of IBDs was proved to be greatly accelerated by the FCRT hydrogel.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan 430000, China; Hubei Clinical Medical Research Center of Trauma and Microsurgery, Wuhan 430000, China
| | - Ying Chu
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Jingyi Du
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Yan Hu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan 430000, China; Hubei Clinical Medical Research Center of Trauma and Microsurgery, Wuhan 430000, China
| | - Huimin Wang
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Hanghang Liu
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Changying Yang
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Man Wang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Jiabing Ran
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China; Medicine & Engineering & Informatics Fusion and Transformation Key Laboratory of LuZhou City, Luzhou, China. 646000.
| | - Aixi Yu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan 430000, China; Hubei Clinical Medical Research Center of Trauma and Microsurgery, Wuhan 430000, China.
| |
Collapse
|
35
|
Zhao Y, Hang R, Li H, Sun Y, Yao R, Huang X, Zhang X, Yao X, Wang H, Xiao Y, Huang D, Han Y, Wang X, Hang R. Biomaterial Surface-Mediated Macrophages Exert Immunomodulatory Roles by Exosomal CCL2-Induced Membrane Integrin β1 Trafficking in Recipient Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409809. [PMID: 39836488 PMCID: PMC11905086 DOI: 10.1002/advs.202409809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 12/29/2024] [Indexed: 01/23/2025]
Abstract
The interaction between biomaterials and immune system is a critical area of research, especially in tissue engineering and regenerative medicine. A fascinating and less explored aspect involves the immunomodulatory behaviors of macrophage (MΦ)-derived exosomes induced by biomaterial surfaces. Herein, untreated surface, nanostructured surface, and type I collagen (Col-I)-decorated nanostructured surface of titanium implants are chosen to culture MΦs, followed by extraction of MΦ-derived exosomes and investigation of their immunomodulatory functions and mechanisms. The results show that the exosomes in the untreated group carried plenty of inflammatory cytokines, predominantly C─C motif chemokine ligand 2 (CCL2). After targeting recipient cells, the CCL2 on the exosomes can specifically bind to its receptor C─C motif chemokine receptor 2, triggering downstream signaling pathways to induce internalization of membrane integrin β1 and targeted lysosomal degradation, consequently suppressing the functions of recipient cells. In contrast, the exosomes in the nanostructured group, especially Col-I-decorated nanostructured group carried few CCL2, moderating their inhibition on the functions of recipient cells. These findings not only clearly show that CCL2 is a key constituent of exosomes involved in the interaction between biomaterials and host immune system, but also potentially a key target for designing advanced biomaterials to promote tissue repair and regeneration.
Collapse
Affiliation(s)
- Yuyu Zhao
- Shanxi Key Laboratory of Biomedical Metal MaterialsCollege of Materials Science and EngineeringTaiyuan University of TechnologyTaiyuan030024China
| | - Ruiyue Hang
- Shanxi Key Laboratory of Biomedical Metal MaterialsCollege of Materials Science and EngineeringTaiyuan University of TechnologyTaiyuan030024China
| | - Huifei Li
- School and Hospital of StomatologyShanxi Medical UniversityTaiyuan030001China
| | - Yonghua Sun
- Shanxi Key Laboratory of Biomedical Metal MaterialsCollege of Materials Science and EngineeringTaiyuan University of TechnologyTaiyuan030024China
| | - Runhua Yao
- Shanxi Key Laboratory of Biomedical Metal MaterialsCollege of Materials Science and EngineeringTaiyuan University of TechnologyTaiyuan030024China
| | - Xiaobo Huang
- Shanxi Key Laboratory of Biomedical Metal MaterialsCollege of Materials Science and EngineeringTaiyuan University of TechnologyTaiyuan030024China
| | - Xiangyu Zhang
- Shanxi Key Laboratory of Biomedical Metal MaterialsCollege of Materials Science and EngineeringTaiyuan University of TechnologyTaiyuan030024China
| | - Xiaohong Yao
- Shanxi Key Laboratory of Biomedical Metal MaterialsCollege of Materials Science and EngineeringTaiyuan University of TechnologyTaiyuan030024China
| | - Huaiyu Wang
- Center for Human Tissues and Organs DegenerationShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518055China
| | - Yin Xiao
- School of Medicine and DentistryGriffith UniversityGold CoastQLD4222Australia
| | - Di Huang
- Research Center for Nano‐Biomaterials & Regenerative MedicineDepartment of Biomedical EngineeringCollege of Biomedical EngineeringTaiyuan University of TechnologyTaiyuan030024China
| | - Yong Han
- State‐Key Laboratory for Mechanical Behavior of MaterialsXi'an Jiaotong UniversityXi'an710049China
| | - Xing Wang
- School and Hospital of StomatologyShanxi Medical UniversityTaiyuan030001China
| | - Ruiqiang Hang
- Shanxi Key Laboratory of Biomedical Metal MaterialsCollege of Materials Science and EngineeringTaiyuan University of TechnologyTaiyuan030024China
- State‐Key Laboratory for Mechanical Behavior of MaterialsXi'an Jiaotong UniversityXi'an710049China
| |
Collapse
|
36
|
Poudel K, Vithiananthan T, Kim JO, Tsao H. Recent progress in cancer vaccines and nanovaccines. Biomaterials 2025; 314:122856. [PMID: 39366184 DOI: 10.1016/j.biomaterials.2024.122856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
Vaccine science, nanotechnology, and immunotherapy are at the forefront of cancer treatment strategies, each offering significant potential for enhancing tumor-specific immunity and establishing long-lasting immune memory to prevent tumor recurrence. Despite the promise of these personalized and precision-based anti-cancer approaches, challenges such as immunosuppression, suboptimal immune activation, and T-cell exhaustion continue to hinder their effectiveness. The limited clinical success of cancer vaccines often stems from difficulties in identifying effective antigens, efficiently targeting immune cells, lymphoid organs, and the tumor microenvironment, overcoming immune evasion, enhancing immunogenicity, and avoiding lysosomal degradation. However, numerous studies have demonstrated that integrating nanotechnology with immunotherapeutic strategies in vaccine development can overcome these challenges, leading to potent antitumor immune responses and significant progress in the field. This review highlights the critical components of cancer vaccine and nanovaccine strategies for immunomodulatory antitumor therapy. It covers general vaccine strategies, types of vaccines, antigen forms, nanovaccine platforms, challenges faced, potential solutions, and key findings from preclinical and clinical studies, along with future perspectives. To fully unlock the potential of cancer vaccines and nanovaccines, precise immunological monitoring during early-phase trials is essential. This approach will help identify and address obstacles, ultimately expanding the available options for patients who are resistant to conventional cancer immunotherapies.
Collapse
Affiliation(s)
- Kishwor Poudel
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tulasi Vithiananthan
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Hensin Tsao
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
37
|
Kang N, Hwang J, Jeong D, Choi JH, Thangam R, Min S, Hong H, Kim D, Rha H, Lee S, Jung H, Kim T, Zare I, Jung HJ, Najafabadi AH, Jung HD, Zhang K, Zhao P, Bian L, Kim HK, Kim JS, Song G, Yoon J, Park SG, Jang WY, Kang H. Ligand Inter-Relation Analysis Via Graph Theory Predicts Macrophage Response. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2414356. [PMID: 39719659 DOI: 10.1002/adma.202414356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/10/2024] [Indexed: 12/26/2024]
Abstract
Graph theory has been widely used to quantitatively analyze complex networks of molecules, materials, and cells. Analyzing the dynamic complex structure of extracellular matrix can predict cell-material interactions but has not yet been demonstrated. In this study, graph theory-based mathematical modeling of RGD ligand graph inter-relation is demonstrated by differentially cutting off RGD-to-RGD interlinkages with flexibly conjugated magnetic nanobars (MNBs) with tunable aspect ratio. The RGD-to-RGD interlinkages are less effectively cut off by MNBs with a lower aspect ratio, which decreases the shortest path while increasing the number of instances thereof, thereby augmenting RGD nano inter-relation. This facilitates integrin recruitment of macrophages and thus actin fiber assembly and vinculin expression, which mediates pro-regenerative polarization, involving myosin II, actin polymerization, and rho-associated protein kinase. Unidirectional pre-aligning or reversibly lifting highly elongated MNBs both increase RGD nano inter-relation, which promotes host macrophage adhesion and switches their polarization from pro-inflammatory to pro-regenerative phenotype. The latter approach produces nano-spaces through which macrophages can penetrate and establish RGD links thereunder. Using graph theory, this study presents the example of mathematically modeling the functionality of extracellular-matrix-mimetic materials, which can help elucidate complex dynamics of the interactions occurring between host cells and materials via versatile geometrical nano-engineering.
Collapse
Affiliation(s)
- Nayeon Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Jangsun Hwang
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul, 02841, Republic of Korea
| | - Daun Jeong
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul, 02841, Republic of Korea
| | - Ji Hye Choi
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul, 02841, Republic of Korea
| | - Ramar Thangam
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Sunhong Min
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hyunsik Hong
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Dahee Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hyunji Rha
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Sungkyu Lee
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hwapyung Jung
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Taeeon Kim
- Department of Nano-Bio Convergence, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, 51508, Republic of Korea
- Department of Future Convergence Materials, Korea University, Seoul, 02841, Republic of Korea
| | - Iman Zare
- Research and Development Department, Sina Medical Biochemistry Technologies Co. Ltd., Shiraz, 7178795844, Iran
| | - Hee Joon Jung
- Interdisciplinary Materials Measurements Institute, Korea Research Institute of Standards and Science (KRISS), Daejeon, 34113, Republic of Korea
| | | | - Hyun-Do Jung
- Division of Materials Science and Engineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Kunyu Zhang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Pengchao Zhao
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Hong-Kyu Kim
- Advanced Analysis Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul, 02841, Republic of Korea
| | - Guosheng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Sung-Gyu Park
- Department of Nano-Bio Convergence, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, 51508, Republic of Korea
- Department of Future Convergence Materials, Korea University, Seoul, 02841, Republic of Korea
| | - Woo Young Jang
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul, 02841, Republic of Korea
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
- Department of Future Convergence Materials, Korea University, Seoul, 02841, Republic of Korea
- College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
38
|
Zhang B, Qu H, Zhang Z, Wang X, Dou Z, Li X, Cao R, Zhang K, Zhang J, Zhang Q. Eu-Doped TiO 2 Coatings via One-Step In Situ Preparation Enhance Macrophage Polarization and Osseointegration of Implants. ACS APPLIED MATERIALS & INTERFACES 2025; 17:8886-8900. [PMID: 39885805 DOI: 10.1021/acsami.4c17495] [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: 02/01/2025]
Abstract
The controllable regulation of immune and osteogenic processes plays a critical role in the modification of biocompatible materials for tissue regeneration. In this study, titanium dioxide-europium coatings (MAO/Eu) were prepared on the surface of a titanium alloy (Ti-6Al-4V) via a one-step process combining microarc oxidation (MAO) and in situ doping. The incorporation of Eu significantly improved the hydrophilic and mechanical properties of the TiO2 coatings without altering their morphology. The presence of Eu effectively stimulated calcium influx in macrophages and activated β-catenin through the wnt/β-catenin signaling pathway. Consequently, macrophage M2 polarization was accelerated through the overexpression of prostaglandin E2 (PGE2). Additionally, Ca2+ promoted the osteogenic differentiation of MC3T3-E1 cells through the synergistic upregulation of transcription factors (e.g., AP-1, BMP-2). In vivo studies demonstrated that MAO/Eu coatings significantly enhanced osseointegration compared with the titanium alloy group. Therefore, MAO/Eu shows promising potential as an ideal coating for implants that offers effective immunomodulatory strategies and improves bone integration.
Collapse
Affiliation(s)
- Baoping Zhang
- School (Hospital) of Stomatology, Lanzhou University, Lanzhou 730000, China
- Gansu Province Key Lab of Maxillofacial Reconstruction and Intelligent Manufacturing, Lanzhou University, Lanzhou 730000, China
| | - Huidan Qu
- School (Hospital) of Stomatology, Lanzhou University, Lanzhou 730000, China
| | - Zhidong Zhang
- School (Hospital) of Stomatology, Lanzhou University, Lanzhou 730000, China
| | - Xinyu Wang
- School (Hospital) of Stomatology, Lanzhou University, Lanzhou 730000, China
| | - Zhihao Dou
- School (Hospital) of Stomatology, Lanzhou University, Lanzhou 730000, China
| | - Xinjie Li
- School (Hospital) of Stomatology, Lanzhou University, Lanzhou 730000, China
| | - Rui Cao
- School (Hospital) of Stomatology, Lanzhou University, Lanzhou 730000, China
| | - Kailiang Zhang
- School (Hospital) of Stomatology, Lanzhou University, Lanzhou 730000, China
- Gansu Province Key Lab of Maxillofacial Reconstruction and Intelligent Manufacturing, Lanzhou University, Lanzhou 730000, China
| | - Jingxiang Zhang
- School (Hospital) of Stomatology, Lanzhou University, Lanzhou 730000, China
- Gansu Province Key Lab of Maxillofacial Reconstruction and Intelligent Manufacturing, Lanzhou University, Lanzhou 730000, China
- College of Civil Engineering and Mechanics of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Mechanics on Disaster and Environment in Western China and the Ministry of Education of China, Lanzhou University, Lanzhou 730000, China
| | - Qiangqiang Zhang
- College of Civil Engineering and Mechanics of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Mechanics on Disaster and Environment in Western China and the Ministry of Education of China, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
39
|
So Y, Yim D, Kim HK, Lee S, Lee H, Yu Y, Choi C, Choi Y, Kim H, Yang CS, Kim JH. Functional Nanosheet Immunoswitches Reprogramming Innate Macrophages for Immunotherapy of Colorectal Cancer and Sepsis. ACS NANO 2025; 19:5165-5177. [PMID: 39898465 DOI: 10.1021/acsnano.4c08828] [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: 02/04/2025]
Abstract
Macrophages are involved in the immunopathogenesis of cancer and inflammatory diseases and are a primary target for immunotherapy to reprogram the M1 and M2 phenotypes in tumor and inflammatory microenvironments. Herein, functional nanosheet immunoswitches that can bidirectionally polarize macrophages in tumor and inflammatory microenvironments are designed for effective immunotherapy of colorectal cancer and sepsis. WSe2 nanosheets are functionalized with palmitic acid to obtain an M1 immunoswitch (PA-WSe2) that promotes the polarization of macrophages toward the M1 phenotype in the tumor microenvironment by activating the STAT1 signaling pathway. WS2 nanosheets bearing linoleic acid are synthesized as an M2 immunoswitch (LA-WS2) that effectively polarizes macrophages to the M2 phenotype in the septic microenvironment by activating the STAT3 signaling pathway. The PA-WSe2 M1 immunoswitch upregulates the secretion of pro-inflammatory cytokines and reactive oxygen and nitrogen species (ROS and RNS) via M1 polarization, leading to the effective immunotherapy for colorectal cancer in vivo. In contrast, the LA-WS2 M2 immunoswitch induces the elevated production of anti-inflammatory cytokines and scavenging of ROS and RNS through M2 polarization, resulting in superior immunotherapy for severe sepsis in mice. These nanosheet immunoswitches can provide a route to immunotherapy for various cancers and inflammatory diseases.
Collapse
Affiliation(s)
- Yoonhee So
- Department of Materials Science and Chemical Engineering, Hanyang University, Ansan 15588, Republic of Korea
| | - DaBin Yim
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California 94720, United States
| | - Hyo Keun Kim
- Department of Molecular and Life Science, and Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Sin Lee
- Department of Materials Science and Chemical Engineering, Hanyang University, Ansan 15588, Republic of Korea
| | - Hyunji Lee
- Department of Materials Science and Chemical Engineering, Hanyang University, Ansan 15588, Republic of Korea
| | - Yejoo Yu
- Department of Materials Science and Chemical Engineering, Hanyang University, Ansan 15588, Republic of Korea
| | - Chanhee Choi
- Department of Materials Science and Chemical Engineering, Hanyang University, Ansan 15588, Republic of Korea
| | - Yujin Choi
- Department of Materials Science and Chemical Engineering, Hanyang University, Ansan 15588, Republic of Korea
| | - Hongwon Kim
- Department of Materials Science and Chemical Engineering, Hanyang University, Ansan 15588, Republic of Korea
| | - Chul-Su Yang
- Department of Molecular and Life Science, and Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Jong-Ho Kim
- Department of Materials Science and Chemical Engineering, Hanyang University, Ansan 15588, Republic of Korea
| |
Collapse
|
40
|
Hofbauer SI, Fink LA, Young RE, Vijayakumar T, Nelson KM, Bellopede N, Alameh MG, Weissman D, Gleghorn JP, Riley RS. Cytokine mRNA Delivery and Local Immunomodulation in the Placenta using Lipid Nanoparticles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.637086. [PMID: 39974923 PMCID: PMC11839073 DOI: 10.1101/2025.02.07.637086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
During pregnancy, the maternal immune system adapts to balance tolerance of the semi-allogenic fetus while protecting the fetus from pathogens. Dysregulated immune activity at the maternal-fetal interface contributes to pregnancy complications, such as recurrent pregnancy loss and preeclampsia. Compared to healthy placentas, preeclamptic placentas exhibit increased pro-inflammatory signaling, including a predominance of inflammatory macrophages, leading to impaired tissue remodeling and restricted blood flow. However, the precise mechanisms driving this immune imbalance remain poorly understood, in part due to the lack of tools to probe individual pathways. Here, we use lipid nanoparticles (LNPs) to deliver cytokine-encoded mRNA to placental cells, called trophoblasts, enabling local immunomodulation. LNP-mediated delivery of IL-4 and IL-13 mRNA induced cytokine secretion by trophoblasts, leading to polarization of primary human monocytes toward anti-inflammatory phenotypes. Notably, lowering the mRNA dose increased expression of alternatively-activated macrophage markers, revealing an inverse relationship between dose and polarization status. Intravenous injection of LNPs in pregnant mice achieved placental secretion of IL-4 and IL-13 with minimal changes to pro-inflammatory cytokines in the serum. These findings establish LNPs as a tool for local immunomodulation in the placenta, offering a strategy to study and treat immune dysfunction in pregnancy and in other inflammatory conditions.
Collapse
|
41
|
Hou L, Meng Z, Zhang J, Jiao Y, Chang K, He J, Zhang J. Effect of 3D-Printed Polycaprolactone Scaffold With Powdery/Smooth Micromorphology on Local Immune Environments. J Biomed Mater Res A 2025; 113:e37869. [PMID: 39893554 DOI: 10.1002/jbm.a.37869] [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: 07/30/2024] [Revised: 11/24/2024] [Accepted: 12/30/2024] [Indexed: 02/04/2025]
Abstract
Selective laser sintering (SLS) has become a viable approach for producing biodegradable medical implants in various clinical applications. The resulting scaffolds typically exhibit a powdery microstructure, which may potentially impact the behavior of immune cells and immune responses in surrounding tissues. However, limited research has been conducted to understand the effect of surface morphology in SLS-fabricated scaffolds on local immune environments. This study aims to compare the effect of SLS-fabricated polycaprolactone (PCL) scaffolds with powdery and smooth surface morphologies on immune-related biological responses. Compared with those on the powdery micromorphology, RAW264.7 macrophages displayed greater dispersion and adopted a spread and elongated morphology on the scaffolds with smooth surface. The expression levels of arginase-1 and CD206 were found to be upregulated in macrophages adhering to the PCL scaffolds with smooth surface, accompanied by an augmented secretion of anti-inflammatory cytokines TGF-β and IL-10. Conversely, there was a decrease in the secretion of pro-inflammatory cytokines TNF-α and IL-12. When implanted in vivo, the SLS-derived scaffolds were completely covered by host tissues, Withing increased collagen deposition, indicating good histocompatibility. At 1-week post-implantation, there was a significantly higher presence of M2-type macrophages surrounding the scaffold compared to M1 macrophages in both groups. By 3 weeks post-implantation, the overall level of macrophages had decreased in both groups. However, a significant higher level of M1 macrophages were observed in the powdery scaffold group. At the same time, the number of neutrophils around the powder scaffold increased significantly, demonstrating long-term local inflammatory responses. The results suggested that post-treated scaffolds with smooth surfaces can effectively reduce local inflammation, making them more suitable for clinical implantation.
Collapse
Affiliation(s)
- Lan Hou
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, China
| | - Zijie Meng
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, China
- National Medical Products Administration (NMPA) key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, China
- State Industry-Education Integration Center for Medical Innovations, Xi'an Jiaotong University, Xi'an, China
| | - Jiawei Zhang
- The First Clinical Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Yangchi Jiao
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, China
| | - Kexin Chang
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, China
| | - Jiankang He
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, China
- National Medical Products Administration (NMPA) key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, China
- State Industry-Education Integration Center for Medical Innovations, Xi'an Jiaotong University, Xi'an, China
| | - Juliang Zhang
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, China
| |
Collapse
|
42
|
Gangadaran P, Onkar A, Rajendran RL, Goenka A, Oh JM, Khan F, Nagarajan AK, Muthu S, Krishnan A, Hong CM, Ahn BC. Noninvasive in vivo imaging of macrophages: understanding tumor microenvironments and delivery of therapeutics. Biomark Res 2025; 13:20. [PMID: 39865337 PMCID: PMC11770947 DOI: 10.1186/s40364-025-00735-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/21/2025] [Indexed: 01/28/2025] Open
Abstract
Macrophages are pivotal in the body's defense and response to inflammation. They are present in significant numbers and are widely implicated in various diseases, including cancer. While molecular and histological techniques have advanced our understanding of macrophage biology, their precise function within the cancerous microenvironments remains underexplored. Enhancing our knowledge of macrophages and the dynamics of their extracellular vesicles (EVs) in cancer development can potentially improve therapeutic management. Notably, macrophages have also been harnessed to deliver drugs. Noninvasive in vivo molecular imaging of macrophages is crucial for investigating intricate cellular processes, comprehending the underlying mechanisms of diseases, tracking cells and EVs' migration, and devising macrophage-dependent drug-delivery systems in living organisms. Thus, in vivo imaging of macrophages has become an indispensable tool in biomedical research. The integration of multimodal imaging approaches and the continued development of novel contrast agents hold promise for overcoming current limitations and expanding the applications of macrophage imaging. This study comprehensively reviews several methods for labeling macrophages and various imaging modalities, assessing the merits and drawbacks of each approach. The review concludes by offering insights into the applicability of molecular imaging techniques for real time monitoring of macrophages in preclinical and clinical scenarios.
Collapse
Affiliation(s)
- Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Akanksha Onkar
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Ramya Lakshmi Rajendran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Anshika Goenka
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| | - Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Fatima Khan
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - ArulJothi Kandasamy Nagarajan
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, 603203, Tamilnadu, India
| | - Sathish Muthu
- Department of Orthopaedics, Government Medical College, Tamil Nadu, 639004, Karur, India
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Tamil Nadu, 641021, Coimbatore, India
| | - Anand Krishnan
- Precision Medicine and Integrated Nano-Diagnostics (P-MIND) Research Group, Office of the Dean, Faculty of Health Sciences, University of the Free State, Bloemfontein, 9300, South Africa
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea.
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea.
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Korea.
| | - Byeong-Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Korea.
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea.
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea.
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Korea.
| |
Collapse
|
43
|
Ghattas M, Dwivedi G, Chevrier A, Horn-Bourque D, Alameh MG, Lavertu M. Chitosan immunomodulation: insights into mechanisms of action on immune cells and signaling pathways. RSC Adv 2025; 15:896-909. [PMID: 39802469 PMCID: PMC11719903 DOI: 10.1039/d4ra08406c] [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: 11/28/2024] [Accepted: 12/22/2024] [Indexed: 01/16/2025] Open
Abstract
Chitosan, a biodegradable and biocompatible natural polymer composed of β-(1-4)-linked N-acetyl glucosamine (GlcNAc) and d-glucosamine (GlcN) and derived from crustacean shells, has been widely studied for various biomedical applications, including drug delivery, cartilage repair, wound healing, and tissue engineering, because of its unique physicochemical properties. One of the most promising areas of research is the investigation of the immunomodulatory properties of chitosan, since the biopolymer has been shown to modulate the maturation, activation, cytokine production, and polarization of dendritic cells and macrophages, two key immune cells involved in the initiation and regulation of innate and adaptive immune responses, leading to enhanced immune responses. Several signaling pathways, including the cGAS-STING, STAT-1, and NLRP3 inflammasomes, are involved in chitosan-induced immunomodulation. This review provides a comprehensive overview of the current understanding of the in vitro immunomodulatory effects of chitosan. This information may facilitate the development of chitosan-based therapies and vaccine adjuvants for various immune-related diseases.
Collapse
Affiliation(s)
- Majed Ghattas
- Department of Chemical Engineering, Polytechnique Montreal Montreal QC Canada
- Institute of Biomedical Engineering, Polytechnique Montreal Montreal QC Canada
| | - Garima Dwivedi
- Perelman School of Medicine, University of Pennsylvania Philadelphia PA USA
| | - Anik Chevrier
- Department of Chemical Engineering, Polytechnique Montreal Montreal QC Canada
| | - Delano Horn-Bourque
- Department of Chemical Engineering, Polytechnique Montreal Montreal QC Canada
- Institute of Biomedical Engineering, Polytechnique Montreal Montreal QC Canada
| | - Mohamad-Gabriel Alameh
- Perelman School of Medicine, University of Pennsylvania Philadelphia PA USA
- Penn Institute for RNA Innovation, University of Pennsylvania Philadelphia PA USA
| | - Marc Lavertu
- Department of Chemical Engineering, Polytechnique Montreal Montreal QC Canada
- Institute of Biomedical Engineering, Polytechnique Montreal Montreal QC Canada
| |
Collapse
|
44
|
Li J, Zhai X, Yu C. Spatial distribution-based progression of spinal cord injury pathology: a key role for neuroimmune cells. Front Immunol 2025; 15:1505755. [PMID: 39850888 PMCID: PMC11754049 DOI: 10.3389/fimmu.2024.1505755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 12/19/2024] [Indexed: 01/25/2025] Open
Abstract
An external trauma, illness, or other pathological cause can harm the structure and function of the spinal cord, resulting in a significant neurological disorder known as spinal cord injury (SCI). In addition to impairing movement and sensory functions, spinal cord injury (SCI) triggers complex pathophysiological responses, with the spatial dynamics of immune cells playing a key role. The inflammatory response and subsequent healing processes following SCI are profoundly influenced by the spatial distribution and movement of immune cells. Despite significant advances in both scientific and clinical research, SCI therapy still faces several challenges. These challenges primarily stem from our limited understanding of the spatial dynamics of immune cell distribution and the processes that regulate their interactions within the microenvironment following injury. Therefore, a comprehensive investigation into the spatial dynamics of immune cells following SCI is essential to uncover their mechanisms in neuroinflammation and repair, and to develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Jian Li
- Shanghai TCM-Integrated Hospital, Shanghai University of TCM, Department of Neurosurgery, Shanghai, China
| | - Xiaolei Zhai
- Department of Neurosurgery, Shuyang Hospital of Traditional Chinese Medicine, Affiliated Shuyang Hospital of Nanjing University of Chinese Medicine, Shuyang, China
| | - Chaochun Yu
- Shanghai TCM-Integrated Hospital, Shanghai University of TCM, Department of Neurosurgery, Shanghai, China
| |
Collapse
|
45
|
Yin J, Liao Y, Liu S, Che B, Zhu H, Yang B, Shi B. Titanium nanotubes modulate immunophenotyping and cytokine secretion of T cells via IL-17A: a bioinformatic analysis and experimental validation. Front Immunol 2025; 15:1381158. [PMID: 39840051 PMCID: PMC11747796 DOI: 10.3389/fimmu.2024.1381158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 12/02/2024] [Indexed: 01/23/2025] Open
Abstract
Object We aim to explore the immunomodulatory properties of T cells on different titanium nanotubes and the key immunological factors involved in this process. Methods Transcriptome data from GEO database of healthy people and healthy implants were used to analyze cell infiltration and factor distribution of adaptive immune using bioinformatics tools. T cells from activated rat were cultured on titanium nanotubes that were prepared by anodization with different diameters (P-0, NT15-30 nm, NT40-100 nm, NT70-200 nm). The proliferation and expressions of the main transcription factors and cytokines of T-cells were detected. Magnetic bead sorting of CD3+ T cells and transcriptome sequencing were performed to explore the signaling pathways and key immune factors that may influence the related immune responses. Results Bioinformatics analysis showed that healthy peri-implant tissues were enriched by the most of T-cell subtypes. T-cell-mediated adaptive immunological responses involved IL-17A. On the third day, the NT15 and NT40 groups showed significantly higher pro-proliferative effects than the NT70 group (P<0.05). Notably, the NT40 group exhibited the lowest T-bet expression (P<0.05) along with the highest levels of Rorγt, Gata3, and Foxp3(P<0.05), followed by the NT15 group. Additionally, the NT40 group demonstrated reduced RANKL, TNF-α, and IL-6 (P<0.05) and increased OPG and IL-10 (P<0.05). Meanwhile, the NT15 group had lower IFN-γ expression(P>0.05) but higher IL-4, and TGF-β1 expressions(P<0.05). Differential expressed genes (DGEs) of T-cell related to the morphologies of titanium nanotubes were mostly enriched in the IL-17 signaling pathway mediated by IL-17A/F. Gene and protein expressions indicated that the NT40 group had the highest secretion in IL-17A of T cells. Conclusion Titanium nanotube morphologies in medium (100 nm) and small (30 nm) sizes significantly influence T cell differentiation and immune factor secretion, with T-cell-derived IL-17A likely playing a key regulatory role.
Collapse
Affiliation(s)
- Jingju Yin
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Oral Medicine Center, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- School of Stomatology, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Oral Disease, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yunyang Liao
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Oral Medicine Center, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- School of Stomatology, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Oral Disease, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Shaofeng Liu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Oral Medicine Center, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- School of Stomatology, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Oral Disease, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Bangwei Che
- Department of Urology & Andrology, The First Affiliated of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Hanghang Zhu
- School of Stomatology, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Oral Disease, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Bingbing Yang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Oral Medicine Center, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- School of Stomatology, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Oral Disease, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Bin Shi
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Oral Medicine Center, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
46
|
Acharya AB, Hegde U, Acharya S. Nanosystems for modulation of immune responses in periodontal therapy: a mini-review. FRONTIERS IN DENTAL MEDICINE 2025; 5:1509775. [PMID: 39917718 PMCID: PMC11797770 DOI: 10.3389/fdmed.2024.1509775] [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: 10/11/2024] [Accepted: 12/13/2024] [Indexed: 02/09/2025] Open
Abstract
Periodontitis is one of the most common oral diseases. It is generally treated by non-surgical and/or surgical therapy with adjunctive approaches for prevention and control. The current understanding of the pathogenesis of periodontitis has unraveled the importance of the inflammatory and immune reactions to combat periodontitis whose etiology is an overlap of microbial, genetic, and environmental factors in a susceptible host. Based on this premise, many therapeutic modalities have been investigated or attempted to resolve this inflammatory disease. Amongst these, nanomedicine has been shown to have therapeutic applications in periodontitis, especially focused on immunomodulation because periodontitis is characterized by over-reactive immune response. This mini-review explores the potential of nanosystems in treating periodontitis by providing an overview of the research efforts in this field of therapeutics. The unique physicochemical and targeting properties of nanosystems seem to be potentially effective platforms for treating periodontitis.
Collapse
Affiliation(s)
- Anirudh B. Acharya
- Department of Restorative Dentistry, College of Dental Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Usha Hegde
- Department of Oral Pathology, JSS Dental College and Hospital, JSS Academy of Higher Education and Research (JSSAHER), Mysore, Karnataka, India
| | - Swetha Acharya
- Department of Oral Pathology, JSS Dental College and Hospital, JSS Academy of Higher Education and Research (JSSAHER), Mysore, Karnataka, India
| |
Collapse
|
47
|
Tang Q, Fan Y, Sun J, Fan W, Zhao B, Yin Z, Cao Y, Han Y, Su B, Yang C, Yu P, Ning C, Chen L. Remodel Heterogeneous Electrical Microenvironment at Nano-Scale Interface Optimizes Osteogenesis by Coupling of Immunomodulation and Angiogenesis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406090. [PMID: 39692158 DOI: 10.1002/smll.202406090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/02/2024] [Indexed: 12/19/2024]
Abstract
Immunomodulation is essential for implants to regulate tissue regeneration, while bioelectricity plays a fundamental role in regulating immune activities. Under natural preferences, the bone matrix electrical microenvironment is heterogeneous in the nanoscale, which provides fundamental electrical cues to regulate bone immunity and regenerative repair. However, remodeling bone nanoscale heterogeneous electrical microenvironment remains a challenge, and the underlying immune modulation mechanism remains to be explored. In this research, in situ discretely distributed nano-heterojunctions are constructed on titanium oxide nanofibers to mimic the heterogeneous electrical microenvironment exhibited by bone collagen fibers. The material is identified to directly regulate calcium ion channeling for anti-inflammatory polarization of macrophages. Surprisingly, the highly biomimetic heterogeneous electrical microenvironment can induce a pro-angiogenic phenotypic transformation of macrophages, leading to enhanced neo-vascularization at the early stage of osteogenesis. Mechanistic exploration identifies that PI3K signaling pathway-mediated FGF2 secretion may partially explain for strengthened coupling of immunomodulation and angiogenesis, which optimizes subsequent bone regeneration. These findings highlight the significance of biomimetic heterogeneous electrical cues on immune-modulation and provide a design principle for future electroactive implant materials.
Collapse
Affiliation(s)
- Qingming Tang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Youzhun Fan
- School of Material Science and Engineering, National Engineering Research Center for Tissue Restoration and Reconstruction, Metallic Materials Surface Functionalization Engineering Research Center of Guangdong Province, South China University of Technology, Guangzhou, 510641, P. R. China
| | - Jiwei Sun
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Wenjie Fan
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Baoying Zhao
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Zhaoyi Yin
- Faculty of Materials Science and Engineering, Kunming University of Science and Technology, Kunming, 650093, China
| | - Yaru Cao
- Department of Neurobiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yunyun Han
- Department of Neurobiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bin Su
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Cheng Yang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Peng Yu
- School of Material Science and Engineering, National Engineering Research Center for Tissue Restoration and Reconstruction, Metallic Materials Surface Functionalization Engineering Research Center of Guangdong Province, South China University of Technology, Guangzhou, 510641, P. R. China
| | - Chengyun Ning
- School of Material Science and Engineering, National Engineering Research Center for Tissue Restoration and Reconstruction, Metallic Materials Surface Functionalization Engineering Research Center of Guangdong Province, South China University of Technology, Guangzhou, 510641, P. R. China
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| |
Collapse
|
48
|
Kong J, Yao Z, Stelzel JL, Yang Y, Chen J, Feng H, Schmidt C, Zhang C, Krishnan K, Chen L, Pan J, Ding K, Zhu Y, Li X, Doloff JC, Mao H, Reddy SK. Granular Nanofiber-Hydrogel Composite-Programmed Regenerative Inflammation and Adipose Tissue Formation. Adv Healthc Mater 2025; 14:e2403094. [PMID: 39580666 PMCID: PMC11773126 DOI: 10.1002/adhm.202403094] [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: 11/04/2024] [Indexed: 11/26/2024]
Abstract
The interplay between biomaterials and host immune responses critically determines outcomes in tissue restoration. Recent studies suggest that physicochemical properties of materials can dictate pro-regenerative versus pro-fibrotic responses and have begun to define the key immune cell types and signals governing these divergent effects. This emerging understanding enables the engineering of regenerative biomaterials capable of functional restoration in situ. An injectable nanofiber-hydrogel composite (NHC) microparticles are designed and constructed from cross-linked electrospun collagen nanofiber fragments surface-bonded to the hyaluronic acid hydrogel network via covalent conjugation during the cross-linking process. The collagen nanofiber fragments, acting as the structural reinforcement component, increased the overall storage modulus of the NHC to a level comparable to native soft tissues while maintaining a sufficiently high degree of porosity of the hydrogel phase to allow host cell infiltration following subcutaneous injection of the NHC microparticles. More importantly, the NHC promoted macrophage/monocyte infiltration, migration, and spreading, sustained cell recruitment over time, and enhanced the proangiogenic effect and recruitment of PDGFRα+ perivascular progenitor cells, leading to extensive adipose tissue remodeling. This study demonstrates the regenerative potential of the injectable NHC microgels as an off-the-shelf solution for devastating soft tissue losses.
Collapse
|
49
|
Khodaei A, Nawaz Q, Zhu Z, Amin Yavari S, Weinans H, Boccaccini AR. Biomolecule and Ion Releasing Mesoporous Nanoparticles: Nonconvergent Osteogenic and Osteo-immunogenic Performance. ACS APPLIED MATERIALS & INTERFACES 2024; 16:67491-67503. [PMID: 39576881 DOI: 10.1021/acsami.4c17540] [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: 11/24/2024]
Abstract
Immune-involved cell communications have recently been introduced as key role players in the fate of mesenchymal stem cells in making bone tissue. In this study, a drug delivery system for bone (re)generation based on copper-doped mesoporous bioactive glass nanoparticles (BGNPs) was developed to codeliver copper as a biologically active ion and icariin as an anti-inflammatory agent. This design was based on temporal inflammation fluctuations from proinflammatory to anti-inflammatory during bone generation. Three in vitro models were performed with human mesenchymal stem cells (hMSCs) to verify the osteo-immunomodulatory effects of released copper ions and icariin: nonstimulated, co-conditioned with macrophage medium and co-cultured with macrophages. Both icariin and copper showed increased levels of alkaline phosphatase activation, indicating a direct osteogenic effect. Copper-doped BGNPs showed the highest increase of osteo-immunogenic properties in a mineralization assay and also induced short-term inflammation. However, the mineralization dropped in copper doped BGNPs after loading with icariin due to copper-icariin chelate formation and inhibition of the early inflammatory phase in the immune-stimulated in vitro models. In the absence of copper, the direct osteogenic properties of icariin overtook its osteo-immunogenic inhibition and increased calcification. Overall, BGNPs doped with 5 mol % copper and no icariin showed the highest bone-forming capacity.
Collapse
Affiliation(s)
- Azin Khodaei
- Department of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
- Department of Orthopedics, University Medical Center Utrecht, 3508GA Utrecht, The Netherlands
| | - Qaisar Nawaz
- Department of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Zhengqing Zhu
- Department of Orthopedics, University Medical Center Utrecht, 3508GA Utrecht, The Netherlands
| | - Saber Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, 3508GA Utrecht, The Netherlands
- Regenerative Medicine Centre Utrecht, Utrecht University, 3508GA Utrecht, The Netherlands
| | - Harrie Weinans
- Department of Orthopedics, University Medical Center Utrecht, 3508GA Utrecht, The Netherlands
| | - Aldo R Boccaccini
- Department of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| |
Collapse
|
50
|
Zhang N, Zhao L, Li J, Li H, Chen Y. Harnessing Nanotechnology for Gout Therapy: Colchicine-Loaded Nanoparticles Regulate Macrophage Polarization and Reduce Inflammation. Biomater Res 2024; 28:0089. [PMID: 39665079 PMCID: PMC11632155 DOI: 10.34133/bmr.0089] [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: 03/25/2024] [Revised: 09/13/2024] [Accepted: 09/24/2024] [Indexed: 12/13/2024] Open
Abstract
Gout is a disease caused by hyperuricemia, characterized by inflammation reactions triggered by macrophage polarization. Colchicine is a commonly used drug for gout treatment, but its mechanism of action remains unclear. The aim of this study was to investigate the regulatory effect of colchicine on macrophage polarization to enhance the therapeutic effectiveness against gout inflammation. To accomplish this, a mouse model was established, and peripheral blood mononuclear cell samples were collected. Single-cell RNA sequencing was employed to reveal cellular heterogeneity and identify key genes. Molecular docking and experimental validation were performed to confirm the binding between the key genes and colchicine. Lentiviral intervention and biochemical indicator detection were conducted to assess the impact of key genes on gout mice. Additionally, the therapeutic effect of colchicine incorporated into neutrophil membrane-coated nanoparticles was investigated. The study found that macrophage polarization plays a critical role in gout, and AHNAK was identified as the key gene through which colchicine affects macrophage polarization. Lentiviral intervention to decrease AHNAK expression was shown to alleviate joint swelling in gout mice and regulate macrophage polarization. Colchicine encapsulated in R4F peptide-modified neutrophil membrane-coated Pluronic F127 nanoparticle (R4F-NM@F127) nanocarriers inhibited M1 macrophage polarization, induced M2 macrophage polarization, alleviated gout, and minimized toxicity to normal tissues. Colchicine suppressed M1 macrophage polarization and induced M2 macrophage polarization by binding to AHNAK protein, thereby alleviating gout. Colchicine incorporated into R4F-NM@F127 nanocarriers can serve as a targeted therapeutic drug to regulate macrophage polarization, alleviate gout, and reduce toxicity to normal tissues.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Rheumatology and Immunology,
Shengjing Hospital Affiliated to China Medical University, Shenyang 110000, China
| | - Lanqing Zhao
- Department of Sleep Medicine Center, The Shengjing Affiliated Hospital,
China Medical University, Shenyang 110000, Liaoning, China
| | - Jinwei Li
- Department of Neurology/Stroke Center, the First Affiliated Hospital ofChina Medical University, China Medical University, Shenyang 110000, Liaoning, China
| | - Hongxi Li
- Department of Pain Management,
Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Yu Chen
- Department of The Fourth Otolaryngology Head and Neck Surgery,
Shengjing Hospital of China Medical University, Shenyang 110000, China
| |
Collapse
|