1
|
Yang L, Wang P, Zhang Y, Zhou J, Bi X, Qian Z, Hou S, Li L, Fan Y. Hybrid cell membrane coating orchestrates foreign-body reactions, anti-adhesion, and pro-regeneration in abdominal wall reconstruction. Biomaterials 2025; 321:123289. [PMID: 40154120 DOI: 10.1016/j.biomaterials.2025.123289] [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: 11/30/2024] [Revised: 03/10/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Tension-free synthetic meshes are the clinical standard for hernia repair, but they often trigger immune response-mediated complications such as severe foreign-body reactions (FBR), visceral adhesions, and fibrotic healing, increasing the risk of recurrence. Herein, we developed a hybrid cell membrane coating for macroscale mesh fibers that acts as an immune orchestrator, capable of balancing immune responses with tissue regeneration. Cell membranes derived from red blood cells (RBCs) and platelets (PLTs) were covalently bonded to fiber surfaces using functionalized-liposomes and click chemistry. The fusion of clickable liposomes with cell membranes significantly improved coating efficiency, coverage uniformity, and in vivo stability. Histological and flow cytometric analyses of subcutaneous implantation in rats and mice demonstrated significant biofunctional heterogeneity among various cell membrane coatings in FBR. Specifically, the RBC-PLT-liposome hybrid cell membrane coating markedly mitigated FBR, facilitated host cell infiltration, and promoted M2-type macrophage polarization. Importantly, experimental results of abdominal wall defect repairs in rats indicate that the hybrid cell membrane coating effectively prevented visceral adhesions, promoted muscle regenerative healing, and enhanced the recruitment of Pax7+/MyoD+ muscle satellite cells. Our findings suggest that the clickable hybrid cell membrane coating offers a promising approach to enhance clinical outcomes of hernia mesh in abdominal wall reconstruction.
Collapse
Affiliation(s)
- Lingbing Yang
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, Hangzhou, 311115, China; Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100191, China; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), Key Laboratory of Innovation and Transformation of Advanced Medical Devices of Ministry of Industry and Information Technology, Beihang University, Beijing, 100191, China
| | - Pu Wang
- Department of Hernia and Abdominal Wall Surgery, Peking University People's Hospital, Beijing, 100044, China
| | - Yilin Zhang
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, Hangzhou, 311115, China; Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100191, China; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), Key Laboratory of Innovation and Transformation of Advanced Medical Devices of Ministry of Industry and Information Technology, Beihang University, Beijing, 100191, China
| | - Jin Zhou
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, Hangzhou, 311115, China; Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100191, China; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), Key Laboratory of Innovation and Transformation of Advanced Medical Devices of Ministry of Industry and Information Technology, Beihang University, Beijing, 100191, China
| | - Xuewei Bi
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Zhiyong Qian
- Department of Anatomy, Basic Medicine College, Inner Mongolia Medical University, Hohhot, 010000, China
| | - Sen Hou
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, Hangzhou, 311115, China; Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100191, China; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), Key Laboratory of Innovation and Transformation of Advanced Medical Devices of Ministry of Industry and Information Technology, Beihang University, Beijing, 100191, China
| | - Linhao Li
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, Hangzhou, 311115, China; Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100191, China; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), Key Laboratory of Innovation and Transformation of Advanced Medical Devices of Ministry of Industry and Information Technology, Beihang University, Beijing, 100191, China.
| | - Yubo Fan
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, Hangzhou, 311115, China; Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100191, China; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), Key Laboratory of Innovation and Transformation of Advanced Medical Devices of Ministry of Industry and Information Technology, Beihang University, Beijing, 100191, China.
| |
Collapse
|
2
|
Shen Q, Chen J, Wang T, Yang Y, Huang C, Zhang W, Tian G, Cheng N, Kai S. Dual functional photocatalytic hydrogel coupled with hydrogen evolution and glucose depletion for diabetic wound therapy. J Colloid Interface Sci 2025; 695:137753. [PMID: 40344732 DOI: 10.1016/j.jcis.2025.137753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/25/2025] [Accepted: 04/29/2025] [Indexed: 05/11/2025]
Abstract
Diabetic foot ulcers (DFUs), a common and serious complication of diabetes mellitus, are exacerbated by hyperglycemia-induced chronic inflammation and oxidative stress, which collectively impede the wound-healing process. Effective management requires coordinated regulation of the pathological microenvironment through localized glucose reduction, anti-inflammatory modulation, and reactive oxygen species (ROS) scavenging. This study developed an injectable functionalized hydrogel incorporating a Bi nanocrystal-decorated bismuth tungstate/hydrogen-doped titanium dioxide (Bi2WO6/H-TiO2) heterojunction with dual photocatalytic properties: glucose degradation and hydrogen evolution. Upon exposure to light, the hydrogel exploits glucose in the wound as the sacrificial substrate to simultaneously decrease local glucose concentrations and facilitate in situ hydrogen production. The released hydrogen exhibits potent antioxidant and anti-inflammatory activities, synergizing with glucose consumption to inhibit cellular apoptosis and accelerate tissue repair. A systematic evaluation revealed enhanced cell proliferation and migration in hyperglycemic in vitro models. In vivo experiments using a diabetic murine model demonstrated 50 % wound closure within 3 days, accompanied by improved angiogenesis and collagen remodeling. This photocatalytic synergistic strategy represents a clinically promising modality for diabetic wound treatment by regulating the microenvironment and restoring redox homeostasis.
Collapse
Affiliation(s)
- Qing Shen
- School of Pharmacy, Shandong Second Medical University, Weifang, Shandong, China
| | - Jiyu Chen
- School of Pharmacy, Shandong Second Medical University, Weifang, Shandong, China
| | - Tao Wang
- School of Pharmacy, Shandong Second Medical University, Weifang, Shandong, China
| | - Yilei Yang
- School of Pharmacy, Shandong Second Medical University, Weifang, Shandong, China
| | - Changbao Huang
- School of Pharmacy, Shandong Second Medical University, Weifang, Shandong, China
| | - Weifen Zhang
- School of Pharmacy, Shandong Second Medical University, Weifang, Shandong, China
| | - Guihua Tian
- Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Road, Xicheng District, Beijing, China.
| | - Ni Cheng
- School of Pharmacy, Shandong Second Medical University, Weifang, Shandong, China.
| | - Shuangshuang Kai
- School of Pharmacy, Shandong Second Medical University, Weifang, Shandong, China.
| |
Collapse
|
3
|
Zhou X, Wang P, Xie L, Chan YK, Jiao Z, Shu R, Bai D, Lai S, Deng Y. Molybdoenzymes-emulating bio-heterojunction hydrogel with rapid disinfection and macrophage reprogramming for wound regeneration. Biomaterials 2025; 320:123284. [PMID: 40121831 DOI: 10.1016/j.biomaterials.2025.123284] [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: 01/27/2025] [Revised: 03/02/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Developing hydrogel dressings with the capabilities to accommodate irregular wounds and provide a cascade disinfective-regenerative microenvironment for wound repair is of great importance to combating pathogenic bacteria-infected wounds but remains an ongoing challenge. To address the conundrum, we devise a molybdoenzymes-emulating bio-heterojunction (M-bioHJ) doped double network (DN) hydrogel dressing for bacterial-infected wound healing. The near-infrared (NIR) photothermal effect of the M-bioHJ facilitates the exchange of multiple dynamic crosslinking sites in the hydrogel, endowing the hydrogel with photo-remote reprocessing capabilities to completely accommodate the encountered irregular wounds and ultimately accomplish the admirable therapeutic effect. Meanwhile, the introduced M-bioHJ shows NIR light-enhanced photodynamic activity to induce a massive engendering of reactive oxygen species (ROS), allowing rapid sterilization without reliance on exogenous hydrogen peroxide. Furthermore, the Mo ions released from the M-bioHJ-encapsulated hydrogel can play a crucial role in reprogramming the macrophage phenotype and determining tissue regeneration. Both in vitro and in vivo evidences authenticate the accelerated healing potential of infected wounds through the synergistic effects of photo-reprocessing, disinfection, and macrophage-reprogramming facilitated by the hydrogel. These findings highlight the promising application prospects of such neoteric M-bioHJ-encapsulated hydrogel dressings for wound disinfection and tissue regeneration.
Collapse
Affiliation(s)
- Xiong Zhou
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China; Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Peiqi Wang
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China; State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Lu Xie
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China; State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yau Kei Chan
- Department of Ophthalmology, The University of Hong Kong, 999077, Hong Kong, China
| | - Zheng Jiao
- Swanson School of Engineering, University of Pittsburgh, Pittsburgh, 15261, USA
| | - Rui Shu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Ding Bai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Shuangquan Lai
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China; Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, Guangdong 519000, China
| | - Yi Deng
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China; State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China; Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
4
|
Bi X, Mao Z, Zhang Y, Ren Z, Yang K, Yu C, Chen L, Zheng R, Guan J, Liu Z, Yu B, Huang Y, Shu X, Zheng Y. Endogenous dual-responsive and self-adaptive silk fibroin-based scaffold with enhancement of immunomodulation for skull regeneration. Biomaterials 2025; 320:123261. [PMID: 40132357 DOI: 10.1016/j.biomaterials.2025.123261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 02/19/2025] [Accepted: 03/16/2025] [Indexed: 03/27/2025]
Abstract
Despite the current biomaterials (e.g. titanium mesh and polyether ether ketone) have been applied to clinical skull repair, the limitations on mechanical match, shape adaptability, bioactivity and osteointegration have greatly limited their clinical application. In this work, we constructed a water and inflammatory microenvironment dual-responsive self-adaptive silk fibroin-magnesium oxide-based scaffold with the matrix metalloproteinase-2-responsive gelatin-methacryloyl-interleukin-4 (IL-4) coating, which presented good mechanical compliance, quickly shape matching and intraoperative reprocessability. With the capability of responding to an acute inflammation microenvironment followed by a triggered on-demand release of the IL-4, the combination of immunoactive IL-4 and Mg2+ co-ordinately facilitated metabolic reprogramming by suppressing glycolysis, promoting mitochondrial oxidative phosphorylation and modulating adenosine 5'-monophosphate-activated protein kinase (AMPK) signalling pathways in macrophages, resulting in significantly facilitating M2 macrophage activation. During the stage of tissue remodelling, the sustained release of Mg2+ further promoted macrophage M2 polarization and the expression of anti-inflammatory cytokines, significantly reduced immune response and improved ectopic osteogenesis ability. Meanwhile, the cranial defect models of male rats demonstrated that this scaffold could significantly enhance biomineralized deposition and vascularisation, and achieve good bone regeneration of cranial defects. Overall, the bioactive scaffold provides a promising biomaterial and alternative repair strategy for critical-size skull defect repair.
Collapse
Affiliation(s)
- Xuewei Bi
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, China; School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Zhinan Mao
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, China; School of Materials Science and Engineering, Peking University, Beijing, 100871, China.
| | - Yilin Zhang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Zeqi Ren
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Kang Yang
- School of Materials Science and Engineering, Anhui University of Technology, Maanshan 243002, China
| | - Chunhao Yu
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China; School of Life, Beijing Institute of Technology, No.5, Zhongguancun South Street, Haidian District, Beijing, China
| | - Lei Chen
- Beijing Research Institute of Orthopedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Rui Zheng
- Beijing Research Institute of Orthopedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Juan Guan
- International Research Center for Advanced Structural and Biomaterials, School of Materials Science & Engineering, Beihang University, Beijing 100191, China
| | - Zhenhai Liu
- Beijing Research Institute of Orthopedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Binsheng Yu
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, China
| | - Yongcan Huang
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, China.
| | - Xiong Shu
- Beijing Research Institute of Orthopedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China.
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China.
| |
Collapse
|
5
|
Wu J, Zhang F, Li Z, Gan L, Cao H, Cao H, Hao C, Sun Z, Wang W. Multiple omics-based machine learning reveals specific macrophage sub-clusters in renal ischemia-reperfusion injury and constructs predictive models for transplant outcomes. Comput Biol Chem 2025; 117:108421. [PMID: 40086342 DOI: 10.1016/j.compbiolchem.2025.108421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/24/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Ischemia-reperfusion injury (IRI) is closely associated with numerous severe postoperative complications, including acute rejection, delayed graft function (DGF) and graft failure. Macrophages are central to modulating the aseptic inflammatory response during the IRI process. The objective of this study is to conduct an analysis of the developmental and differentiation characteristics of macrophages in IRI, identify distinct molecules subtypes of IRI, and establish robust predictive strategies for DGF and graft survival. METHOD We analyzed scRNA-Seq data from GEO database to identify macrophage sub-clusters specific to renal IRI, and use the hdWGCNA algorithm to screen gene modules closely associated with this sub-cluster. Integrating these module genes with the results from bulk RNA-Seq differential analysis to obtain hub genes, and delineating the different IRI molecular subtypes through consensus clustering based on the expression profiles of hub genes. Innovatively, the gene expression matrix was transformed into a unique graphic pixel module and applied advanced computer vision processing algorithms to construct a DGF predictive model. Additionally, we also employed 111 combinations of 10 machine learning algorithms to develop a predictive signature for graft survival. Finally, we validated the expression of the key gene ANXA1 in a mouse IRI model using qRT-PCR, WB, and IHC. RESULT This study successfully identified a subset of macrophages closely associated with renal IRI, and cell communication and pseudo-time analysis implied that they may be instrumental in both the maintenance and exacerbation of the IRI process. Utilizing the expression patterns of hub genes, recipients can be clustered into two subtypes (CI and C2) with unique clinical and molecular features. We innovatively applied deep learning algorithms to construct a model for DGF prediction, which can effectively mitigate batch effects among IRI recipients. Compared to other existing models, our model demonstrated superior performance with AUC of 0.816 and 0.845 in the training and validation set. Furthermore, we also used the random survival forest algorithm to develop a high-precision predictive signature for graft failure. The mouse IRI model confirmed a marked upregulation of ANXA1 mRNA and protein expression in renal tissue following IRI. CONCLUSION This study successfully revealed the macrophage sub-cluster closely associated with renal IRI. Two distinct IRI subgroups with different characteristics were identified and robust strategies were constructed for predicting DGF and graft survival, which can offer potential therapeutic targets for the treatment of IRI and reference for early prevention of various postoperative complications.
Collapse
Affiliation(s)
- Jiyue Wu
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Institute of Urology, Capital Medical University, Beijing, China
| | - Feilong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Institute of Urology, Capital Medical University, Beijing, China
| | - Zhen Li
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Institute of Urology, Capital Medical University, Beijing, China
| | - Lijian Gan
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Institute of Urology, Capital Medical University, Beijing, China
| | - Haoyuan Cao
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Institute of Urology, Capital Medical University, Beijing, China
| | - Huawei Cao
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Institute of Urology, Capital Medical University, Beijing, China
| | - Changzhen Hao
- Department of Urology, Peking University International Hospital, Beijing, China.
| | - Zejia Sun
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Institute of Urology, Capital Medical University, Beijing, China.
| | - Wei Wang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Institute of Urology, Capital Medical University, Beijing, China.
| |
Collapse
|
6
|
Gao B, Ni H, Lai J, Gao N, Luo X, Wang Y, Chen Y, Zhao J, Yu Z, Zhang J, Cai W, Yang G. Macrophage response to fibrin structure mediated by Tgm2-dependent mitochondrial mechanosensing. Bioact Mater 2025; 50:382-395. [PMID: 40331213 PMCID: PMC12051126 DOI: 10.1016/j.bioactmat.2025.04.022] [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: 04/17/2025] [Accepted: 04/17/2025] [Indexed: 05/08/2025] Open
Abstract
Following an injury at the implantation position, blood-material interactions form a fibrin architecture, which serves as the initial activator of foreign body response (FBR). However, there is limited knowledge regarding how the topography of fibrin architectures regulates macrophage behavior in mitigating FBR. Mechanical cues of the microenvironment have been reported to shape immune cell functions. Here, we investigated macrophage mechanobiology at the organelle level by constructing heterogeneous fibrin networks. Based on findings in vivo, we demonstrated that adhesion-mediated differentiation of mitochondrial function modulated macrophage polarization. The finite activation of integrin signaling upregulated transglutaminase 2 (Tgm2) in a trans-manner, augments PGC1α-mediated mitochondrial biogenesis. Our study highlighted the previously overlooked spatial structures of host proteins adsorbed on material surfaces, advocating for a paradigm shift in material design strategies, from focusing solely on physical properties to considering the modification of host proteins.
Collapse
Affiliation(s)
- Bicong Gao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Haifeng Ni
- Zhejiang Key Laboratory of Plastic Modification and Processing Technology, College of Materials Science & Engineering, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Junhong Lai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Ning Gao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Xinxin Luo
- Zhejiang Key Laboratory of Plastic Modification and Processing Technology, College of Materials Science & Engineering, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Yani Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Jiaying Zhao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Zhou Yu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Jing Zhang
- Zhejiang Key Laboratory of Plastic Modification and Processing Technology, College of Materials Science & Engineering, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Wenjin Cai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| |
Collapse
|
7
|
Wu N, Han Z, Lv W, Huang Y, Zhu J, Deng J, Xue Q. Reprogramming peritoneal macrophages with outer membrane vesicle-coated PLGA nanoparticles for endometriosis prevention. Biomaterials 2025; 319:123198. [PMID: 40015004 DOI: 10.1016/j.biomaterials.2025.123198] [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: 10/03/2024] [Revised: 01/23/2025] [Accepted: 02/16/2025] [Indexed: 03/01/2025]
Abstract
Endometriosis is a chronic inflammatory disease that primarily affects women of reproductive age. The current hormonal treatments are unsuitable for women who wish to conceive, highlighting the need for non-hormonal therapeutic alternatives. In this study, we engineered outer membrane vesicle (OMV)-coated poly (lactic-co-glycolic acid) (PLGA) nanoparticles (OMV-NPs) as a potential therapy for endometriosis. These OMV-NPs were internalized by macrophages more efficiently than bacterial OMVs and preserved the immunostimulatory properties of OMVs. In vivo administration of OMV-NPs in mice achieved prolonged retention in the peritoneal cavity, with effective uptake by nearly 80 % of the peritoneal macrophages. Notably, treatment with OMV-NPs reprogrammed macrophages toward the M1 phenotype, resulting in a significant decrease in the M2 to M1 ratio within the peritoneal cavity and in endometriotic lesions. This shift from M2 to M1 was associated with reduced TGF-β1 production and suppressed myofibroblast activation, which led to substantial inhibition of endometriosis progression. Furthermore, immunohistochemical imaging of paired eutopic and ectopic endometrial tissues from endometriosis patients revealed a positive correlation between M2-polarized macrophages and fibrosis. This finding suggests that reprogramming macrophages with OMV-NPs could be a promising therapeutic approach for endometriosis.
Collapse
Affiliation(s)
- Ning Wu
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing 100034, China
| | - Ziwei Han
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenxing Lv
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanjuan Huang
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingwen Zhu
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing 100034, China
| | - Jinqi Deng
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Qing Xue
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing 100034, China.
| |
Collapse
|
8
|
Chen L, Zhu J, Ge N, Liu Y, Yan Z, Liu G, Li Y, Wang Y, Wu G, Qiu T, Dai H, Han J, Guo C. A biodegradable magnesium alloy promotes subperiosteal osteogenesis via interleukin-10-dependent macrophage immunomodulation. Biomaterials 2025; 318:122992. [PMID: 39862617 DOI: 10.1016/j.biomaterials.2024.122992] [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: 05/16/2024] [Revised: 10/13/2024] [Accepted: 12/03/2024] [Indexed: 01/27/2025]
Abstract
In situ bone regeneration and vertical bone augmentation have been huge problems in clinical practice, always imposing a significant economic burden and causing patient suffering. Herein, MgZnYNd magnesium alloy rod implantation in mouse femur resulted in substantial subperiosteal new bone formation, with osteoimmunomodulation playing a pivotal role. Abundant macrophages were attracted to the subperiosteal new bone region and proved to be the most important regulation cells for bone regeneration. Periosteum stripping, macrophage depletion, and interleukin-10 (IL-10) blockade effectively diminished the MgZnYNd alloy-induced subperiosteal osteogenesis. Mechanistically, the degradation products of MgZnYNd alloy promoted M2 macrophage polarization and the secretion of anti-inflammatory cytokine IL-10, which enhanced periosteum-derived stem cells (PDSCs) osteogenesis through the JAK1-STAT3 pathway. An anti-IL-10 neutralizing antibody or STAT3 inhibitor significantly inhibited M2 macrophage-mediated osteogenic differentiation of PDSCs. Transcriptomics and proteomics revealed that periostin is the core regulator of PDSCs osteogenic differentiation. Furthermore, a novel clinical translation application of Mg-induced subperiosteal osteogenesis was developed, demonstrating its ability to preserve the height and width of the alveolar crest in rats and rabbits following tooth extraction. Collectively, these findings unveil a previously undefined role for Mg alloy-induced subperiosteal osteogenesis via macrophage-mediated osteoimmunomodulation, suggesting the therapeutic potential of magnesium alloy in bone regeneration and bone augmentation.
Collapse
Affiliation(s)
- Liangwei Chen
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China; Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Jianhua Zhu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China; Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Na Ge
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China; Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Yan Liu
- Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China; Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Ziyu Yan
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China; Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Guanqi Liu
- Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Yuqi Li
- Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Yifei Wang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China; Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Guanxi Wu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China; Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Tiancheng Qiu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China; Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Hui Dai
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China.
| | - Jianmin Han
- Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China.
| | - Chuanbin Guo
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China; Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China.
| |
Collapse
|
9
|
Zhang L, Lv T, Hou P, Jin Y, Jia F. Sirt5-mediated polarization and metabolic reprogramming of macrophage sustain brain function following ischemic stroke. Brain Res 2025; 1857:149613. [PMID: 40180144 DOI: 10.1016/j.brainres.2025.149613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 03/16/2025] [Accepted: 03/30/2025] [Indexed: 04/05/2025]
Abstract
Ischemic stroke has become the leading cause of morbidity and mortality in adults. Reperfusion may initiate inflammatory response and cause damage to brain. Macrophage is supposed to be the major contributor of neuroinflammation and immune response. Hypersuccinylation correlates with neuropathological process post cerebral ischemia, rendering the possibility of functional role of succinylation in regulating recovery from injury. Here we reported that ischemic stroke causes upregulation of global protein succinylation dramatically. Mechanically, Sirt5 expression is repressed upon ischemic stroke, which exerts a crucial role in orchestrating global protein succinylation level. Furthermore, deficiency of Sirt5 enhances infiltration, M1 polarization and metabolic programming of macrophage in response to stroke via succinylation of Pkm2. Physiologically, depletion of Sirt5 enlarges damage region of brain during stroke. Utilization of Sirt5 agonist resveratrol efficiently ameliorates the destructive effects induced by stroke, thereby supporting recovery from brain injury. Our study not only reveal a heretofore unrecognized mechanism underlying the relation between stroke and protein succinylation, but also shed light on clinical potential for management of stroke injury via targeting protein succinylation.
Collapse
Affiliation(s)
- Linfeng Zhang
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tao Lv
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pinpin Hou
- Shanghai Immune Therapy Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yichao Jin
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Feng Jia
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
10
|
Zaccagnini G, Baci D, Tastsoglou S, Cozza I, Madè A, Voellenkle C, Nicoletti M, Ruatti C, Longo M, Perani L, Gaetano C, Esposito A, Martelli F. miR-210 overexpression increases pressure overload-induced cardiac fibrosis. Noncoding RNA Res 2025; 12:20-33. [PMID: 40034123 PMCID: PMC11874870 DOI: 10.1016/j.ncrna.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 01/23/2025] [Accepted: 01/26/2025] [Indexed: 03/05/2025] Open
Abstract
Aortic stenosis, a common valvular heart disease, can lead to left ventricular pressure overload, triggering pro-fibrotic responses in the heart. miR-210 is a microRNA that responds to hypoxia and ischemia and plays a role in immune regulation and in cardiac remodeling upon myocardial infarction. This study investigated the effects of miR-210 on cardiac fibrosis caused by pressure overload. Using a mouse model with inducible miR-210 over-expression, we subjected mice to transverse aortic constriction (TAC) to induce pressure overload. Mice with miR-210 over-expression developed eccentric hypertrophy, heightened expression of hypertrophic markers (Nppa and Nppb) and increased cross sectional area of cardiomyocytes, impacting the free wall of the left ventricle. These findings suggest that miR-210 worsens cardiac dysfunction. Furthermore, miR-210 over-expression led to a more robust and sustained inflammatory response in the heart, increased interstitial and perivascular fibrosis, and activation of myofibroblasts. miR-210 also promoted angiogenesis. In vitro, cardiac fibroblasts over-expressing miR-210 showed increased adhesion, wound healing and migration capacity. Our results demonstrate that miR-210 contributes to adverse cardiac remodeling in response to pressure overload, including eccentric hypertrophy, inflammation, and fibrosis.
Collapse
Affiliation(s)
- G. Zaccagnini
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
- Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - D. Baci
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
| | - S. Tastsoglou
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
| | - I. Cozza
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
| | - A. Madè
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
| | - C. Voellenkle
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
- Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - M. Nicoletti
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
| | - C. Ruatti
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
| | - M. Longo
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
| | - L. Perani
- Preclinical Imaging Facility, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
| | - C. Gaetano
- Laboratorio di Epigenetica, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, 27100, Italy
| | - A. Esposito
- Preclinical Imaging Facility, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
- Vita-Salute San Raffaele University, Milan, 20132, Italy
| | - F. Martelli
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
- Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| |
Collapse
|
11
|
Mao J, Xu Y, Wang W, Deng X, Hui Y, Rui M, Tang J, Wang W, Huang Y, Wu L, Xi K, Zhu Y, Gu Y, Chen L. Topological cues of microparticles train stem cells for tissue repair via mechanotransduction. Bioact Mater 2025; 48:531-549. [PMID: 40114729 PMCID: PMC11923629 DOI: 10.1016/j.bioactmat.2025.02.032] [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: 10/22/2024] [Revised: 02/15/2025] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
Microspheres (MPs) and porous microspheres (PMPs) are the two most widely used microparticles in tissue engineering and stem cell therapy. However, how stem cells perceive the topological differences between them to regulate cell function remains to be unclear. Here, we systematically studied the changes in stem cell function under the action of MPs and PMPs and elucidated the related mechanisms. Our findings show that the porous structure of PMPs can be sensed by focal adhesions (FAs), which triggers the synthesis of F-actin to inhibit the phosphorylation and degradation of Yes-associated protein (YAP), while also transmitting stress to the nucleus through the contraction of F-actin, thereby enhancing the nuclear translocation of YAP protein. The activation of YAP significantly enhances the proliferation, osteogenesis, paracrine and glucose metabolism of BMSCs, making them exhibit stronger bone repair ability in both in vivo and in vitro experiments. In summary, this study provides a comprehensive and reliable understanding of the behavior of BMSCs in response to MPs and PMPs. It also deepens our understanding of the association between microparticles' topological cues and biological functions, which will provide valuable guidance for the construction of bone tissue engineering (BTE) scaffolds.
Collapse
Affiliation(s)
- Jiannan Mao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
- Department of Orthopaedics, Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, No.163 Shoushan Road, Jiang Yin, 214400, PR China
| | - Yichang Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Wenbo Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Xiongwei Deng
- Department of Orthopaedics, Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, No.163 Shoushan Road, Jiang Yin, 214400, PR China
| | - Yujian Hui
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
- Department of Orthopaedics, Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, No.163 Shoushan Road, Jiang Yin, 214400, PR China
| | - Min Rui
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
- Department of Orthopaedics, Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, No.163 Shoushan Road, Jiang Yin, 214400, PR China
| | - Jincheng Tang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Wei Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Yiyang Huang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Liang Wu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, PR China
| | - Kun Xi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Yunrong Zhu
- Department of Orthopaedics, Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, No.163 Shoushan Road, Jiang Yin, 214400, PR China
| | - Yong Gu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Liang Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| |
Collapse
|
12
|
Chen J, Qu B, Yang D, Wang Y, Zhu H, Wang Z, Zhang X, Ma H, Zhao N, Zhao L, Zhou L, He X, Li P. Combined metabolomics and network pharmacology to elucidate the mechanisms of Huiyang Shengji decoction in treating diabetic skin ulcer mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156569. [PMID: 40120541 DOI: 10.1016/j.phymed.2025.156569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/17/2025] [Accepted: 02/24/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Diabetic skin ulcer is a clinical disorder of glucose metabolism that has a long treatment period and is prone to recurrent episodes. Huiyang Shengji decoction (HYSJD) is an effective traditional Chinese medicine for its clinical treatment, but its metabolic effects in patients with diabetic skin ulcers have not been well studied. PURPOSE Our study aimed to investigate the mechanism of pharmacological treatment of HYSJD in treating diabetic skin ulcers. METHODS The potential mechanism underlying diabetic wound treatment by HYSJD was screened using network pharmacology. Ultra-high performance liquid chromatography-MS/MS metabolomics analysis and correlation analysis were performed to investigate potential target pathways and genes. Furthermore, the db/db diabetic wound tissues and RAW264.7 macrophage inflammation model verified the mechanism using molecular biology experiments. RESULTS In network pharmacology, HYSJD played a mainly therapeutic effect by regulating PI3K/AKT signaling pathway, EGFR tyrosine kinase inhibitor resistance, metabolic pathway, and other related metabolic-related pathways. Metabolomics analysis disclosed that L-lysine content increased, while those of linoleic and deoxycholic acids decreased in plasma between the HYSJD-treated group and the control group, participating in biotin metabolism. Among them, PPARγ played an important role. The experiments conducted in db/db mice indicated that HYSJD facilitates VEGF secretion and PPARγ expression. In vitro experiments have revealed that HYSJD inhibits macrophage ROS production, augments mitochondrial ATP production, elevates mitochondrial membrane potential, and diminishes the mitochondrial ECAR rate. Furthermore, these effects culminate in promoting M2 macrophage polarization through PPARγ activation. The molecular docking results revealed that the active compounds from HYSJD were capable of binding to PPARγ protein primarily through hydrogen bonding interactions. Notably, all binding energies were found to be lower than -3 kcal/mol, indicating strong and favorable interactions between the active compounds and the target receptor. CONCLUSIONS The findings suggested that HYSJD regulates biotin metabolism by reducing excess levels of linoleic and deoxycholic acids and increasing levels of L-lysine, which in turn promotes diabetic wound healing by promoting M2 macrophage polarization through PPARγ up-regulation. These findings indicated that HYSJD is a decoction that can effectively treat diabetic skin ulcers.
Collapse
Affiliation(s)
- Jia Chen
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China; Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, 430022, China
| | - Baoquan Qu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Danyang Yang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Yazhuo Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Haoyue Zhu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Zhengchun Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Xiawei Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Huike Ma
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Ning Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Li Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China; Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, 430022, China
| | - Lijiaming Zhou
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China
| | - Xiujuan He
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China.
| | - Ping Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, No. 23rd Art Museum Back Street, Dongcheng District, Beijing 100010, China.
| |
Collapse
|
13
|
Wu L, He J, Shen N, Chen S. Molecular and cellular mechanisms underlying peripheral nerve injury-induced cellular ecological shifts: Implications for neuroregeneration. IBRO Neurosci Rep 2025; 18:120-129. [PMID: 39877591 PMCID: PMC11773043 DOI: 10.1016/j.ibneur.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/27/2024] [Indexed: 01/31/2025] Open
Abstract
The peripheral nervous system is a complex ecological network, and its injury triggers a series of fine-grained intercellular regulations that play a crucial role in the repair process. The peripheral nervous system is a sophisticated ecological network, and its injury initiates a cascade of intricate intercellular regulatory processes that are instrumental in the repair process. Despite the advent of sophisticated microsurgical techniques, the repair of peripheral nerve injuries frequently proves inadequate, resulting in adverse effects on patients' quality of life. Accordingly, the continued pursuit of more efficacious treatments is of paramount importance. In this paper, a review of the relevant literature from recent years was conducted to identify the key cell types involved after peripheral nerve injury. These included Schwann cells, macrophages, neutrophils, endothelial cells, and fibroblasts. The review was conducted in depth. This paper analyses the phenotypic changes of these cells after injury, the relevant factors affecting these changes, and how they coordinate with neurons and other cell types. In addition, it explores the potential mechanisms that mediate the behaviour of these cells. Understanding the interactions between these cells and their mutual regulation with neurons is of great significance for the discovery of new neuroregenerative treatments and the identification of potential therapeutic targets.
Collapse
Affiliation(s)
- Limao Wu
- School of Clinical Medicine, Hebei University of Engineering, No.81 Congtai Road, Congtai District, Handan City, Hebei Province 056004, China
| | - Jinglan He
- Affiliated Hospital of Hebei University of Engineering, No. 80, Jianshe Street, Fuxing District, Handan City, Hebei Province 056003, China
| | - Na Shen
- Department of Science and Education, Affiliated Hospital of Hebei University of Engineering, No.81 Congtai Road, Congtai District, Handan City, Hebei Province 056004, China
| | - Song Chen
- Orthopaedic Center, Affiliated Hospital of Hebei University of Engineering, No.81 Congtai Road, Congtai District, Handan City, Hebei Province 56004, China
| |
Collapse
|
14
|
Hu L, Yang K, Chen Y, Wang H, Fu Z, Jiang L, Xu J, Tian H, Zhu Y, Dai Z, Li Y, Chen X, Lin X, Chen P, Gu C, Fan S. K +-H + coupling strategy for immune regulation and bone defect repair. Mater Today Bio 2025; 32:101744. [PMID: 40275952 PMCID: PMC12019076 DOI: 10.1016/j.mtbio.2025.101744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/31/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025] Open
Abstract
Ion homeostasis is crucial for maintaining cell function. Potassium ion (K+) is one of the most important cations in the human body, and it plays key role in maintaining biological activities and cellular functions, including the intricate balance of ion homeostasis that underpins both physiological and pathological processes. This study explored a novel role of K+ ions in regulating immune cell function and promoting tissue repair, especially in macrophage-mediated environments after severe tissue injury. We designed and synthesized a platelet-liposome vesicles loaded KHCO3 (KHCO3@PLV) that precisely delivered potassium bicarbonate to the site of injury extracellular after intravenous injection; then, precise ultrasound-triggered K+ release regulated extracellular K+ concentrations in the local macrophage environment. These effects collectively validate the K+-H+ coupling strategy - a novel mechanism whereby extracellular K+ elevation induces intracellular pH modulation, subsequently activating the AMPK/Nrf2 axis to reprogram macrophage metabolism and facilitating tissue regeneration through resolution of chronic inflammation. The main conclusion of the study is that an elevated extracellular K+ environment, which is an innovative treatment, is a potentially effective strategy for regulating immune responses and promoting repair after severe tissue injury.
Collapse
Affiliation(s)
- Lintao Hu
- Department of Orthopaedics Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedics Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China, Hangzhou, Zhejiang 310016, China
| | - Ke Yang
- Department of Orthopaedics Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedics Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China, Hangzhou, Zhejiang 310016, China
| | - Yiyu Chen
- Department of Orthopaedics Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedics Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China, Hangzhou, Zhejiang 310016, China
| | - Haoli Wang
- Department of Orthopaedics Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedics Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China, Hangzhou, Zhejiang 310016, China
| | - Zezhou Fu
- Department of Orthopaedics Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedics Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China, Hangzhou, Zhejiang 310016, China
| | - Lejian Jiang
- Department of Orthopaedics Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedics Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China, Hangzhou, Zhejiang 310016, China
- Department of Orthopaedics Surgery, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China
| | - Jiachen Xu
- Department of Orthopaedics Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedics Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China, Hangzhou, Zhejiang 310016, China
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310000, China
| | - Hongsen Tian
- Department of Orthopaedics Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedics Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China, Hangzhou, Zhejiang 310016, China
| | - Yiwei Zhu
- Department of Orthopaedics Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedics Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China, Hangzhou, Zhejiang 310016, China
| | - Zhanqiu Dai
- Department of Orthopaedics Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedics Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China, Hangzhou, Zhejiang 310016, China
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yijun Li
- Department of Orthopaedics Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedics Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China, Hangzhou, Zhejiang 310016, China
| | - Xianhua Chen
- Zhejiang Institute of Medical Device Supervition and Testing, Hangzhou, Zhejiang 310016, China
| | - Xianfeng Lin
- Department of Orthopaedics Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedics Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China, Hangzhou, Zhejiang 310016, China
- Cixi Institute of Biomedicine, Wenzhou Medical University, Ningbo, Zhejiang 315000, China
| | - Pengfei Chen
- Department of Orthopaedics Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedics Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China, Hangzhou, Zhejiang 310016, China
| | - Chenhui Gu
- Department of Orthopaedics Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedics Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China, Hangzhou, Zhejiang 310016, China
| | - Shunwu Fan
- Department of Orthopaedics Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedics Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China, Hangzhou, Zhejiang 310016, China
| |
Collapse
|
15
|
Fan G, Liu Y, Tao L, Wang D, Huang Y, Yang X. Sodium butyrate alleviates colitis by inhibiting mitochondrial ROS mediated macrophage pyroptosis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167756. [PMID: 40044062 DOI: 10.1016/j.bbadis.2025.167756] [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: 11/20/2024] [Revised: 02/06/2025] [Accepted: 02/26/2025] [Indexed: 04/15/2025]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory bowel disease with unclear causes and limited treatment options. Sodium butyrate (NaB), a byproduct of dietary fiber in the intestine, has demonstrated efficacy in treating inflammation. However, the precise anti-inflammatory mechanisms of NaB in colon inflammation remain largely unexplored. This study aims to investigate the effects of NaB on dextran sulfate sodium (DSS)-induced colitis in rats. The findings indicate that oral administration of NaB effectively prevent colitis and reduce levels of serum or colon inflammatory factors. Additionally, NaB demonstrated in vitro inhibition of RAW264.7 inflammation cytokines induced by LPS, along with suppression of the ERK and NF-κB signaling pathway activation. Moreover, NaB mitigated LPS and Nigericin-induced RAW264.7 pyroptosis by reducing indicators of mitochondrial damage, including increased mitochondrial membrane potential (JC-1) levels and decreased Mito-ROS production. NaB increases ZO-1 and Occludin expression in CaCo2 cells by inhibiting RAW264.7 pyroptosis. These results suggest that NaB could be utilized as a therapeutic agent or dietary supplement to alleviate colitis.
Collapse
Affiliation(s)
- Guoqiang Fan
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yaxin Liu
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Limei Tao
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Danping Wang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yizhu Huang
- Singao Xiamen Company, Xiamen 361006, PR China
| | - Xiaojing Yang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
16
|
Tang P, Liu Y, Peng S, Cai Z, Tang G, Zhou Z, Hu K, Zhong Y. Cerebral [ 18F]AIF-FAPI-42-Based PET Imaging of Fibroblast Activation Protein for Non-invasive Quantification of Fibrosis After Ischemic Stroke. Transl Stroke Res 2025; 16:848-858. [PMID: 38940873 DOI: 10.1007/s12975-024-01269-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/08/2024] [Accepted: 06/17/2024] [Indexed: 06/29/2024]
Abstract
The development of fibrosis after injury to the brain or spinal cord limits the regeneration of the central nervous system in adult mammals. However, the extent of fibrosis in the injured brain has not been systematically investigated in mammals in vivo. This study aimed to assess whether [18F]AlF-FAPI-42-based cerebral positron emission tomography (PET) can be utilized to assess the extent of fibrosis in ischemic regions of the brain in vivo. Sprague-Dawley rats underwent permanent occlusion of the right middle cerebral artery (MCAO). On days 3, 7, 14, and 21 after MCAO, the uptake of [18F]AlF-FAPI-42 in the ischemic region of the brain in the MCAO groups surpassed that in the control group (day 0). The specific expression of fibroblast activation protein-α (FAP) in ischemic regions of the brain was also confirmed in immunohistofluorescence experiments in vitro. [18F]AlF-FAPI-42 intensity correlated with the density of collagen deposition in the ischemic hemisphere (p < 0.001). [18F]AlF-FAPI-42 PET/CT imaging demonstrated a specific uptake of radioactivity in the infarcted area in an ischemic stroke patient. PET imaging by using [18F]AlF-FAPI-42 offers a promising non-invasive method for monitoring the progression of cerebral fibrosis caused by ischemic stroke and may facilitate the clinical management of stroke patients. Trial registration: chictr.org.cn ChiCTR2200059004. Registered April 22, 2022.
Collapse
Affiliation(s)
- Peipei Tang
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yang Liu
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Simin Peng
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhikai Cai
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ganghua Tang
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhou Zhou
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Kongzhen Hu
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Yuhua Zhong
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
17
|
Zheng K, Wang S, Deng M, Luo Y, Li W, Zeng L, Wang Y. Mechanisms and Therapeutic Strategies of Macrophage Polarization in Intervertebral Disc Degeneration. JOR Spine 2025; 8:e70065. [PMID: 40371270 PMCID: PMC12077540 DOI: 10.1002/jsp2.70065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/03/2025] [Accepted: 03/26/2025] [Indexed: 05/16/2025] Open
Abstract
Background Intervertebral disc degeneration (IVDD) is a leading cause of low back pain (LBP), contributing significantly to global disability and productivity loss. Its pathogenesis involves complex processes, including inflammation, cellular senescence, angiogenesis, fibrosis, neural ingrowth, and sensitization. Emerging evidence highlights macrophages as central immune regulators infiltrating degenerated discs, with macrophage polarization implicated in IVDD progression. However, the mechanisms linking macrophage polarization to IVDD pathology remain poorly elucidated. Methods A comprehensive literature review was conducted by searching major databases (PubMed, Web of Science, and Scopus) for studies published in the last decade (2014-2024). Keywords included "intervertebral disc degeneration," "macrophage polarization," "inflammation," "senescence," and "therapeutic strategies." Relevant articles were selected, analyzed, and synthesized to evaluate the role of macrophage polarization in IVDD. Results Macrophage polarization dynamically influences IVDD through multiple pathways. Pro-inflammatory M1 macrophages exacerbate disc degeneration by amplifying inflammatory cytokines (e.g., TNF-α, IL-1β), promoting cellular senescence, and stimulating abnormal angiogenesis and neural ingrowth. In contrast, anti-inflammatory M2 macrophages may mitigate degeneration by suppressing inflammation and enhancing tissue repair. Therapeutic strategies targeting macrophage polarization include pharmacological agents (e.g., cytokines, small-molecule inhibitors), biologic therapies, gene editing, and physical interventions. Challenges persist, such as incomplete understanding of polarization triggers, lack of targeted delivery systems, and limited translational success in preclinical models. Conclusion Macrophage polarization is a pivotal regulator of IVDD pathology, offering promising therapeutic targets. Future research should focus on elucidating polarization mechanisms, optimizing spatiotemporal control of macrophage phenotypes, and developing personalized therapies. Addressing these challenges may advance innovative strategies to halt or reverse IVDD progression, ultimately improving clinical outcomes for LBP patients.
Collapse
Affiliation(s)
- Kaiyuan Zheng
- Department of Rehabilitation Medicine, Intensive Care MedicineAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| | - Siyu Wang
- Department of Rehabilitation Medicine, Intensive Care MedicineAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| | - Meng Deng
- Department of Clinical LaboratoryThe First People's Hospital of GuangyuanGuangyuanChina
| | - Yaomin Luo
- Department of Rehabilitation Medicine, Intensive Care MedicineAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| | - Wen Li
- Department of Rehabilitation Medicine, Intensive Care MedicineAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| | - Lianlin Zeng
- Department of Rehabilitation MedicineSuining Central HospitalSuiningChina
| | - Yinxu Wang
- Department of Rehabilitation Medicine, Intensive Care MedicineAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| |
Collapse
|
18
|
Xu L, Qiu J, Ren Q, Wang D, Guo A, Wang L, Hou K, Wang R, Liu Y. Gold nanoparticles modulate macrophage polarization to promote skeletal muscle regeneration. Mater Today Bio 2025; 32:101653. [PMID: 40151803 PMCID: PMC11937682 DOI: 10.1016/j.mtbio.2025.101653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/20/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025] Open
Abstract
Skeletal muscle regeneration is a complex process that depends on the interplay between immune responses and muscle stem cell (MuSC) activity. Macrophages play a crucial role in this process, exhibiting distinct polarization states-M1 (pro-inflammatory) and M2 (anti-inflammatory)-that significantly affect tissue repair outcomes. Recent advancements in nanomedicine have positioned gold nanoparticles (Au NPs) as promising tools for modulating macrophage polarization and enhancing muscle regeneration. This review examines the role of Au NPs in influencing macrophage behavior, focusing on their physicochemical properties, biocompatibility, and mechanisms of action. We discuss how Au NPs can promote M2 polarization, facilitating tissue repair through modulation of cytokine production, interaction with cell surface receptors, and activation of intracellular signaling pathways. Additionally, we highlight the benefits of Au NPs on MuSC function, angiogenesis, and extracellular matrix remodeling. Despite the potential of Au NPs in skeletal muscle regeneration, challenges remain in optimizing nanoparticle design, developing targeted delivery systems, and understanding long-term effects. Future directions should focus on personalized medicine approaches and combination therapies to enhance therapeutic efficacy. Ultimately, this review emphasizes the transformative potential of Au NPs in regenerative medicine, offering hope for improved treatments for muscle injuries and diseases.
Collapse
Affiliation(s)
- Lining Xu
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Jiahuang Qiu
- Research Center of Nano Technology and Application Engineering, School of Public Health,Dongguan Innovation Institute, Guangdong Medical University, Dongguan, 523808, China
| | - Quanzhong Ren
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Dingding Wang
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Anyi Guo
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Ling Wang
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
- Department of Radiology, National Center for Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Kedong Hou
- Department of Orthopedics, Beijing Pinggu District Hospital, Beijing, 101200, China
| | - Renxian Wang
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Yajun Liu
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
- Department of Spine Surgery, National Center for Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| |
Collapse
|
19
|
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
|
20
|
Kim J, Bong KW, Cho JK, Song SC. Thermo-responsive hydrogel via sustained Co-delivery of TA and PDGF to modulate the diabetic microenvironment and accelerate diabetic wound healing. J Mater Chem B 2025. [PMID: 40400445 DOI: 10.1039/d5tb00563a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
In modern society, the need for diabetic wound healing is increasing due to the increase in the number of diabetic patients. In particular, chronic inflammation is a major problem in diabetic wounds due to excessive accumulation of reactive oxygen species (ROS). Therefore, it is essential to remove ROS and promote angiogenesis for effective diabetic wound healing. In this study, we developed a thermo-responsive poly(organophosphazene) hydrogel system (TSP-TP) designed to deliver antioxidants and growth factors for a long period of time. The TSP-TP hydrogel stably loads and continuously releases tannic acid (TA) and platelet-derived growth factor (PDGF) through various physical interactions. Effective ROS scavenging induced macrophage polarization and alleviated chronic inflammation, while the sustained release of PDGF promoted angiogenesis, ultimately maximizing wound healing efficacy in a diabetic mouse model. Based on these results, the proposed TSP-TP hydrogel demonstrates synergistic effects through sustained delivery of antioxidants and growth factors, demonstrating a promising system with high applicability in diabetic wound treatment.
Collapse
Affiliation(s)
- Jisun Kim
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Ki Wan Bong
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Jung-Kyo Cho
- Nexgel Biotech, Co., Ltd., Hanam, 12939, Republic of Korea
| | - Soo-Chang Song
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
- Nexgel Biotech, Co., Ltd., Hanam, 12939, Republic of Korea
| |
Collapse
|
21
|
Zhang D, Wang X, Li W, Wan D, Zhou Y, Ma C, Yang Z, Zhang Y, Li W, Li Z, Lin H, Jin Z, Wu W, Huang H. A Single-Cell Atlas-Inspired Hitchhiking Therapeutic Strategy for Acute Pancreatitis by Restricting ROS in Neutrophils. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2502200. [PMID: 40395143 DOI: 10.1002/adma.202502200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 05/07/2025] [Indexed: 05/22/2025]
Abstract
Neutrophils can undergo transcriptional and epigenetic reprogramming in disease, thus causing inflammation or modulating tissue repair and fibrosis. A thorough understanding of the neutrophil subpopulation composition and their polarization processes in acute pancreatitis (AP) is essential to open up design of treatments tailored to individual patients. Herein, this study distinct subgroups and two differentiation pathways associated with N1 and N2 polarization during AP by single-cell sequencing. Inspired by this, a hollow manganese dioxide (HMnO2)-based nanoreactor (Pyp@APHM) conjugated with neutrophil-binding Ly-6G antibody and loaded with porphyrin is developed for targeted and in situ modulation of neutrophil polarization. Pyp@APHM can enrich the AP site by hitchhiking on neutrophils and then degrade in response to a weakly acidic environment to simultaneously release manganese ions and porphyrin ligands, enabling in situ synthesis of manganese porphyrin antioxidants. Leveraging this strategy, Pyp@APHM can effectively eliminate reactive oxygen species (ROS) and broadly inhibit both N1 and N2 polarization, as well as enhance tissue oxygenation by generating O2, thereby further mitigating pancreatic inflammation. This study provides a comprehensive single-cell atlas of neutrophils in AP and proposes an innovative hitchhiking therapeutic strategy for AP by restricting ROS in neutrophils.
Collapse
Affiliation(s)
- Deyu Zhang
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Xinyue Wang
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Wanshun Li
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Dongling Wan
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Yuyan Zhou
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Congjia Ma
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Zhenghui Yang
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Yang Zhang
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Wenhao Li
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Zhaoshen Li
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Han Lin
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Zhendong Jin
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Wencheng Wu
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Haojie Huang
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
22
|
Jensen L, Guo Z, Sun X, Jing X, Yang Y, Cao Y. Angiogenesis, signaling pathways, and animal models. Chin Med J (Engl) 2025; 138:1153-1162. [PMID: 40254738 PMCID: PMC12091601 DOI: 10.1097/cm9.0000000000003561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Indexed: 04/22/2025] Open
Abstract
ABSTRACT The vasculature plays a critical role in homeostasis and health as well as in the development and progression of a wide range of diseases, including cancer, cardiovascular diseases, metabolic diseases (and their complications), chronic inflammatory diseases, ophthalmic diseases, and neurodegenerative diseases. As such, the growth of the vasculature mediates normal development and physiology, as well as disease, when pathologically induced vessels are morphologically and functionally altered owing to an imbalance of angiogenesis-stimulating and angiogenesis-inhibiting factors. This review offers an overview of the angiogenic process and discusses recent findings that provide additional interesting nuances to this process, including the roles of intussusception and angiovasculogenesis, which may hold promise for future therapeutic interventions. In addition, we review the methodology, including those of in vitro and in vivo assays, which has helped build the vast amount of knowledge on angiogenesis available today and identify important remaining knowledge gaps that should be bridged through future research.
Collapse
Affiliation(s)
- Lasse Jensen
- Department of Health, Medical and Caring Sciences, Unit of Diagnostics and Specialist Medicine, Linköping University, Linköping SE-58183, Sweden
| | - Ziheng Guo
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaoting Sun
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vison and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325024, China
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna 17165, Sweden
| | - Xu Jing
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna 17165, Sweden
| | - Yunlong Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna 17165, Sweden
| |
Collapse
|
23
|
Yingming W, Jing G, Tianhong W, Zhenyu W. M2 Macrophages Mitigate Ocular Surface Inflammation and Promote Recovery in a Mouse Model of Dry Eye. Exp Eye Res 2025:110439. [PMID: 40403951 DOI: 10.1016/j.exer.2025.110439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 04/09/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025]
Abstract
Dry eye disease (DED) is a chronic, progressive, multifactorial condition characterized by tear film instability and ocular surface damage. Ocular surface inflammation, triggered by multiple pathogenic factors, represents one of the key mechanisms in DED pathogenesis. This study aims to investigate the therapeutic effects of anti-inflammatory M2 macrophages conditioned medium (M2-CM) on ocular surface inflammation and their potential mechanisms in improving dry eye symptoms in a mouse model. Mouse macrophages (RAW264.7) were polarized into M2 macrophages by IL-4 under different osmolarities, and M2-CM was collected. Flow cytometry and ELISA were applied to measure the cytokine expression of the M2 macrophages. Primary mouse corneal epithelial cells (CECs) were co-cultured with RAW264.7 and M2 macrophages using a Transwell system. The viability and migration of CECs were assessed using CCK-8 and scratch assays. Mouse DED was established by subcutaneous injection of scopolamine, and the therapeutic effects of M2-CM were evaluated by phenol red thread test, fluorescein staining, and tear film breakup time (TBUT). PCR and immunofluorescence staining were applied to observe inflammatory factors and cells on the ocular surface. M2 macrophages enhanced CEC viability, proliferation, and migration, but hyperosmolarity inhibited M2 macrophage polarization. In the DED model, M2-CM improved ocular surface conditions, reduced pro-inflammatory cytokine expression, and increased anti-inflammatory factors. Immunofluorescence revealed reduced pro-inflammatory cells (M1 macrophages, Th1, and Th17) and increased M2 macrophages in the ocular tissues after M2-CM treatment. These results suggest that M2-CM ameliorates ocular surface inflammation and promotes recovery in DED, offering a potential therapeutic strategy for DED.
Collapse
Affiliation(s)
- Wang Yingming
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University
| | - Gao Jing
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University
| | - Wu Tianhong
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University
| | - Wang Zhenyu
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University.
| |
Collapse
|
24
|
Schröder A, Engelhardt H, Nogueira A, Clausen B, Kirschneck C, Jantsch J, Proff P, Renner K, Paddenberg-Schubert E. The Mechanosensitive PIEZO1 Channel Contributes to the Reaction of RAW264.7 Macrophages to Mechanical Strain. Mediators Inflamm 2025; 2025:9998838. [PMID: 40420945 PMCID: PMC12103965 DOI: 10.1155/mi/9998838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 04/29/2025] [Indexed: 05/28/2025] Open
Abstract
The mechanosensitive channel 'piezo type mechanosensitive ion channel component 1' (PIEZO1) plays a regulatory role in the response of periodontal ligament fibroblasts (PDLFs) to the mechanical strain that occurs during orthodontic tooth movement. In addition to PDLFs, immune cells such as macrophages are also exposed to mechanical stimuli. Macrophages respond to mechanical strain with increased expression of inflammatory mediators. The role of PIEZO1 in this response remains elusive. To investigate the effect of PIEZO1 activation, RAW264.7 macrophages were stimulated with the PIEZO1 activator YODA1 without concurrent application of pressure. To further examine the specific role of PIEZO1 during mechanical strain, RAW264.7 macrophages were exposed to mechanical strain without and with simultaneous inhibition of PIEZO1 either by chemical inhibition (GsMTx4) or siRNA silencing. The expression of genes and proteins involved in orthodontic tooth movement was examined by quantitative PCR, western blot, and enzyme-linked immunosorbent assay (ELISA). Activation of PIEZO1 by YODA1 or mechanical strain increased the expression of inflammatory cytokines and osteoprotegerin (Opg), which is critically involved in bone remodeling processes. Conversely, inhibition of the PIEZO1 channel attenuates the effects of mechanical stress. In conclusion, our data demonstrate that the PIEZO1 channel is a major contributor to the response of macrophages to mechanical strain encountered during orthodontic tooth movement and affects the expression of inflammatory and bone remodeling factors.
Collapse
Affiliation(s)
- Agnes Schröder
- Department of Orthodontics, University Hospital Regensburg, Regensburg, Germany
- Institute for Medical Microbiology and Hygiene, University Regensburg, Regensburg, Germany
| | - Hanna Engelhardt
- Department of Orthodontics, University Hospital Regensburg, Regensburg, Germany
| | - Andressa Nogueira
- Department of Periodontology and Operative Dentistry, University Medical Center Mainz, Mainz, Germany
| | - Björn Clausen
- Institute for Molecular Medicine, University Medical Center Mainz, Mainz, Germany
| | | | - Jonathan Jantsch
- Institute for Medical Microbiology and Hygiene, University Regensburg, Regensburg, Germany
- Institute for Medical Microbiology, Immunology and Hygiene, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Peter Proff
- Department of Orthodontics, University Hospital Regensburg, Regensburg, Germany
| | - Kathrin Renner
- Department of Otorhinolaryngology, University Hospital Regensburg, Regensburg, Germany
| | | |
Collapse
|
25
|
Sharma S, Kishen A. Dysfunctional crosstalk between macrophages and fibroblasts under LPS-infected and hyperglycemic environment in diabetic wounds. Sci Rep 2025; 15:17233. [PMID: 40383800 PMCID: PMC12086240 DOI: 10.1038/s41598-025-00673-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 04/29/2025] [Indexed: 05/20/2025] Open
Abstract
Diabetic wounds, especially diabetic foot ulcers, present a major clinical challenge due to delayed healing and prolonged inflammation. Macrophage-fibroblast interactions are essential for wound repair, yet this crosstalk is disrupted in diabetic wounds due to hyperglycemia and bacterial infection. This study investigates the dysfunctional communication between macrophages and fibroblasts, focusing on autocrine, paracrine, and juxtacrine signaling in simulated diabetic environments. Using monoculture and co-culture models of THP-1-derived macrophages and primary human dermal fibroblasts, we simulated conditions of normal glucose, LPS-induced infection, high glucose (with AGEs), and combined high glucose (with AGEs) and LPS. Macrophages in hyperglycemic and LPS-infected environments exhibited a pro-inflammatory M1 phenotype with elevated expression of CD80, and STAT1 and increased production of IL-1β, TNF-α, and MMP9. Fibroblast migration was significantly impaired under high glucose conditions, particularly in paracrine model. Secretome profiling showed heightened pro-inflammatory cytokines and proteases, with reduced anti-inflammatory markers (IL-10 and VEGF-A) under hyperglycemic conditions. Paracrine signaling exacerbated the inflammatory response, while juxtacrine signaling showed more moderate effects, conducive to healing. These findings highlight the pathological macrophage-fibroblast crosstalk in diabetic wounds, particularly under hyperglycemic and LPS-infected conditions, offering insights for potential immunomodulatory therapies aimed at restoring effective signaling and improving wound healing outcomes.
Collapse
Affiliation(s)
- Shivam Sharma
- The Kishen Lab, Dental Research Institute, University of Toronto, Toronto, Canada
- Faculty of Dentistry, University of Toronto, 124 Edward Street, Toronto, ON, M5G 1G6, Canada
| | - Anil Kishen
- The Kishen Lab, Dental Research Institute, University of Toronto, Toronto, Canada.
- Faculty of Dentistry, University of Toronto, 124 Edward Street, Toronto, ON, M5G 1G6, Canada.
- Department of Dentistry, Mount Sinai Hospital, Toronto, Canada.
| |
Collapse
|
26
|
Wang J, Chen B, Meng Q, Qu F, Ma Z. Using eQTL Mendelian randomization and transcriptomic analysis to identify the relationship between ion channel genes and intracranial aneurysmal subarachnoid hemorrhage. Medicine (Baltimore) 2025; 104:e42457. [PMID: 40388745 PMCID: PMC12091597 DOI: 10.1097/md.0000000000042457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 04/28/2025] [Indexed: 05/21/2025] Open
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) is a complex condition associated with high disability and mortality rates, leading to poor clinical outcomes. Previous observational studies have suggested a link between ion channel genes and aSAH, but the causal relationship remains uncertain. This study utilized Mendelian randomization (MR) to explore the causal association between ion channel genes and aSAH, employing 5 MR methods: inverse variance weighted (IVW), MR-Egger, maximum likelihood, weighted median, and weighted mode. If results from these methods are inconclusive, IVW will be prioritized as the primary outcome. Additionally, MR-Egger, MR-PRESSO, and Cochrane Q tests were conducted to assess heterogeneity and pleiotropy. The stability of MR findings was evaluated using the leave-one-out approach; Bonferroni correction tested the strength of the causal relationship between exposure and outcome. The MR analysis revealed that CACNA2D3 was positively correlated with aSAH (OR 1.245; 95% confidence intervals [CI] 1.008-1.537; P = .042), while ANO6 showed a negative correlation (OR 0.728; 95% CI 0.533-0.993; P = .045). Our findings indicate that increased expression of CACNA2D3 promotes aSAH whereas elevated levels of ANO6 inhibit it. Transcriptome data from intracranial aneurysm samples confirmed significant differential expression of CACNA2D3 and ANO6 between ruptured and unruptured groups. CACNA2D3 being higher in ruptured cases while ANO6 was more expressed in unruptured ones. Furthermore, GeneMANIA analysis along with functional enrichment provided insights into risk factors for aSAH. Through MR analysis, we established a causal link between ion channel genes and aSAH, which helps to better understand the pathogenesis of aSAH and provide new therapeutic targets.
Collapse
Affiliation(s)
- Jing Wang
- Department of Intensive Care Unit, Jining No. 1 People’s Hospital, Shandong, China
| | - Bowang Chen
- Department of Intensive Care Unit, Jining No. 1 People’s Hospital, Shandong, China
| | - Qiang Meng
- Department of Intensive Care Unit, Jining No. 1 People’s Hospital, Shandong, China
| | - Feng Qu
- Department of Intensive Care Unit, Jining No. 1 People’s Hospital, Shandong, China
| | - Zhen Ma
- Department of Intensive Care Unit, Jining No. 1 People’s Hospital, Shandong, China
| |
Collapse
|
27
|
Aktay-Cetin Ö, Pullamsetti SS, Herold S, Savai R. Lung tumor immunity: redirecting macrophages through infection-induced inflammation. Trends Immunol 2025:S1471-4906(25)00096-1. [PMID: 40382244 DOI: 10.1016/j.it.2025.04.004] [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: 01/05/2025] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 05/20/2025]
Abstract
Macrophages play a central role in maintaining tissue homeostasis and in surveillance against pathogens and disease. In the lung, they can adopt either proinflammatory or anti-inflammatory states depending on the nature of the stimulus. As the predominant immune cells in both the lung tumor microenvironment and in sites of lung infection, the functional plasticity of macrophages makes them key players in determining disease outcome. Accurately defining their inflammatory profiles offers an opportunity to reprogram infection-associated macrophages towards enhanced tumor-killing phenotypes. This review explores how acute inflammation can drive macrophage-mediated antitumor immunity and highlights key molecules and signaling pathways that may be leveraged to therapeutically modulate macrophage function.
Collapse
Affiliation(s)
- Öznur Aktay-Cetin
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany; Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Soni Savai Pullamsetti
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany; Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany; Department of Internal Medicine II, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Susanne Herold
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany; Department of Internal Medicine V, German Center for Lung Research (DZL), German Center for Infection Research (DZIF), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Rajkumar Savai
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany; Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany; Department of Internal Medicine II, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany.
| |
Collapse
|
28
|
Ren C, Zi Y, Zhang X, Liao X, Chen H. Basal and AT2 cells promote IPF-lung cancer co-occurrence via EMT: Single-cell analysis. Exp Cell Res 2025; 448:114578. [PMID: 40294812 DOI: 10.1016/j.yexcr.2025.114578] [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: 01/16/2025] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, fibrotic interstitial lung disease. With IPF, the probability of complication with lung cancer (LCA) increases considerably, and the prognosis is worse than that of simple IPF. To understand the pathological mechanisms and molecular pathways shared by these two diseases, we used the single-cell analysis from the Gene Expression Omnibus (GEO) database, and find that basal cells (BCs) and alveolar type 2 cells (AT2 cells) are important components of lung epithelial cells. Changes in molecular pathways in BCs and AT2 cells may be involved in the common pathogenesis of IPF and LCA. KRT17 and S100A14 in BCs may promote the IPF co-occurrence with LCA by mediating the EMT. WFDC2 and KRT19 may be the elements in AT2 cells that activate the EMT process to promote IPF co-occurrence with LCA. In both IPF and LCA, FN1-WNT axis may be involved in the interaction between BCs and AT2 cells. Importantly, the results of immunofluorescence colocalization experiments on tissue samples from patients with IPF and LCA were consistent with these conclusions. Basal-macrophage interactions may have also induced the IPF co-occurrence with LCA via the CYBA-ERK1/2 axis. The regulation of M2 macrophage polarization by JUN/SOD2-glycolysis axis may therefore be involved in the co-morbidity mechanism of IPF and LCA. Therefore, our results suggest that molecular changes in BCs, AT2 cells and macrophages may play important roles in the pathogenesis of IPF co-occurrence with LCA, and the cellular interactions between these cells may be critical for the progression of both diseases.
Collapse
Affiliation(s)
- Cheng Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, Chongqing, 400016, China; Department of Respiratory and Critical Care Medicine, Chongqing University Fuling Hospital, Fuling, Chongqing, 408000, China
| | - Yawan Zi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, Chongqing, 400016, China
| | - Xiaobin Zhang
- Department of Respiratory and Critical Care Medicine, Chongqing University Fuling Hospital, Fuling, Chongqing, 408000, China
| | - Xiuqing Liao
- Department of Respiratory and Critical Care Medicine, Chongqing University Fuling Hospital, Fuling, Chongqing, 408000, China
| | - Hong Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, Chongqing, 400016, China.
| |
Collapse
|
29
|
Xu PJ, Gu YX, Xue Y, Sun J, Liao WQ, Yang QQ, Zhou YL. Advanced Biomimetic Materials in the Prevention of Tendon Adhesions: Design, Preparation, and Application of Hydrogel and Electrospun fiber Membranes. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2411913. [PMID: 40370189 DOI: 10.1002/smll.202411913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 04/05/2025] [Indexed: 05/16/2025]
Abstract
Tendon adhesion formation results from a fibrotic process between the tendon and surrounding tissues, typically occurring after tendon injury or surgery. This condition significantly impacts the quality of life and motor function. Currently, treating adhesions following the repair of injured tendons remains challenging and is a prominent clinical issue that needs to be addressed. This review compiles the existing pathophysiological mechanisms underlying tendon adhesion formation, with a particular focus on the critical roles of inflammation and inflammatory pathways, growth factors and their associated pathways, as well as peritendinous cellular behaviors in promoting adhesion formation. Furthermore, this paper is dedicated to summarizing the evaluation of hydrogels and electrospun fiber membranes as anti-adhesion materials, emphasizing their design, preparation, and application. Additionally, the success of composite patches created by combining these two materials in preventing tendon adhesions is reviewed, which demonstrates the broad applicability of the hydrogel and electrospun film combination. Finally, the review provides insights into future directions for preventing tendon adhesion formation, focusing on material structure and functional design.
Collapse
Affiliation(s)
- Peng Jun Xu
- The Nanomedicine Research Laboratory, Hand Surgery Research Center, Research Central of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Ya Xin Gu
- The Nanomedicine Research Laboratory, Hand Surgery Research Center, Research Central of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Yan Xue
- The Nanomedicine Research Laboratory, Hand Surgery Research Center, Research Central of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Jie Sun
- The Nanomedicine Research Laboratory, Hand Surgery Research Center, Research Central of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Wei Quan Liao
- The Nanomedicine Research Laboratory, Hand Surgery Research Center, Research Central of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Qian Qian Yang
- The Nanomedicine Research Laboratory, Hand Surgery Research Center, Research Central of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - You Lang Zhou
- The Nanomedicine Research Laboratory, Hand Surgery Research Center, Research Central of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| |
Collapse
|
30
|
Ai Z, Li H, Xu S, Cai C, Wang X, Guan Y, Guo R, Wang Y. Overexpression of TAFA4 in the dorsal root ganglion ameliorates neuropathic pain in male rats through promoting macrophage M2-Skewing. Neurochem Int 2025; 187:105993. [PMID: 40381955 DOI: 10.1016/j.neuint.2025.105993] [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: 02/23/2025] [Revised: 05/03/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Neuro-immune interactions between macrophages and primary sensory neurons have been implicated in nerve injury and associated pain. This study aims to explore the function of the TAFA4 as a crucial neuroimmune regulator in modulating macrophage states within the context of neuropathic pain. To elucidate the role of TAFA4 in dorsal root ganglia (DRG) following a chronic constriction injury (CCI) model in male rats, immunofluorescent staining, western blot, flow cytometry analysis and enzyme-linked immunosorbent assay were performed. Microinjection of self-complementary adeno-associated virus expressing TAFA4 mRNA into the L4 and L5 DRGs was conducted to overexpress TAFA4 in the DRGs. Following peripheral nerve injury, we observed a downregulation of TAFA4 in ipsilateral DRG neurons. Restoring this downregulation effectively alleviated the mechanical and thermal nociceptive hypersensitivity by inhibiting pro-inflammatory mediators while promoting the secretion of anti-inflammatory cytokines on day 14 post-CCI. Notably, scAAV-TAFA4 microinjection also facilitated the polarization of macrophages in the DRGs towards the M2 phenotype. Mechanistically, TAFA4 modulates the functions of macrophages in a lipoprotein receptor-related protein 1-dependent manner. Our findings revealed the role of TAFA4 in shifting macrophages in favor of an anti-inflammatory phenotype and enhancing interleukin 10 concentrations in the DRG, suggesting it is a potential analgesic target for alleviating neuropathic pain.
Collapse
Affiliation(s)
- Zhangran Ai
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Huili Li
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Songchao Xu
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Chenghui Cai
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Xuejuan Wang
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Ruijuan Guo
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| | - Yun Wang
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
31
|
Wan HQ, Xie LF, Li HL, Ma Y, Li QH, Dai MQ, Fu YD, Li WJ, Zhou JP, Qian MY, Shen X. GPR40 activation alleviates pulmonary fibrosis by repressing M2 macrophage polarization through the PKD1/CD36/TGF-β1 pathway. Acta Pharmacol Sin 2025:10.1038/s41401-025-01558-y. [PMID: 40369224 DOI: 10.1038/s41401-025-01558-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 04/01/2025] [Indexed: 05/16/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive interstitial lung disease characterized by complex aetiologies involving the accumulation of inflammatory cells, such as macrophages, in the alveoli. This process is driven by uncontrolled extracellular matrix (ECM) deposition and the development of fibrous connective tissues. Here, we observed that the mRNA expression of Ffar1, the gene encoding G protein-coupled receptor 40 (GPR40), is repressed, while Cd36 is increased in the bronchoalveolar lavage fluid (BALF), which is predominantly composed of alveolar macrophages, of IPF patients. Furthermore, the GPR40 protein was found to be largely adhered to macrophages and was pathologically downregulated in the lungs of bleomycin (BLM)-induced PF model mice (PF mice) compared with those of control mice. Specific knockdown of GPR40 in pulmonary macrophages by adeno-associated virus 9-F4/80-shGPR40 (AAV9-shGPR40) exacerbated the fibrotic phenotype in the PF mice, and activation of GPR40 by its determined agonist compound SC (1,3-dihydroxy-8-methoxy-9H-xanthen-9-one) effectively protected the PF mice from pathological exacerbation. Moreover, Ffar1 or Cd36 gene knockout mouse-based assays were performed to explore the mechanism underlying the regulation of GPR40 activation in pulmonary macrophages with compound SC as a probe. We found that compound SC mitigated pulmonary fibrosis progression by preventing M2 macrophage polarization from exerting profibrotic effects through the GPR40/PKD1/CD36 axis. Our results strongly support the therapeutic potential of targeting intrinsic GPR40 activation in pulmonary macrophages for IPF and highlight the potential of compound SC in treating this disease.
Collapse
Affiliation(s)
- Hui-Qi Wan
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ling-Feng Xie
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, China
| | - Hong-Lin Li
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yan Ma
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qiu-Hui Li
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Meng-Qing Dai
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yuan-Dong Fu
- Pulmonary Disease Department, Nanjing Pukou District Central Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wen-Jun Li
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jin-Pei Zhou
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, China.
| | - Min-Yi Qian
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xu Shen
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing, China.
| |
Collapse
|
32
|
Yeung ST, Damani-Yokota P, Thannickal SA, Bartnicki E, Bernier ED, Barnett CR, Khairallah C, Duerr R, Noval MG, Segal LN, Stapleford KA, Khanna KM. Nerve- and airway-associated interstitial macrophages mitigate SARS-CoV-2 pathogenesis via type I interferon signaling. Immunity 2025; 58:1327-1342.e5. [PMID: 40286790 DOI: 10.1016/j.immuni.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 12/27/2024] [Accepted: 04/02/2025] [Indexed: 04/29/2025]
Abstract
Despite vaccines, rapidly mutating viruses such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continue to threaten human health due to an impaired immunoregulatory pathway and a hyperactive immune response. Our understanding of the local immune mechanisms used by tissue-resident macrophages to safeguard the host from excessive inflammation during SARS-CoV-2 infection remains limited. Here, we found that nerve- and airway-associated interstitial macrophages (NAMs) are required to control mouse-adapted SARS-CoV-2 (MA-10) infection. Control mice restricted lung viral distribution and survived infection, whereas NAM depletion enhanced viral spread and inflammation and led to 100% mortality. Mechanistically, type I interferon receptor (IFNAR) signaling by NAMs was critical for limiting inflammation and viral spread, and IFNAR deficiency in CD169+ macrophages mirrored NAM-depleted outcomes and abrogated their expansion. These findings highlight the essential protective role of NAMs in regulating viral spread and inflammation, offering insights into SARS-CoV-2 pathogenesis and underscoring the importance of NAMs in mediating host immunity and disease tolerance.
Collapse
Affiliation(s)
- Stephen T Yeung
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Payal Damani-Yokota
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sara A Thannickal
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Eric Bartnicki
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Eduardo D Bernier
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Clea R Barnett
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Camille Khairallah
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ralf Duerr
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA; Department of Medicine, Vaccine Center, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Maria G Noval
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Leopoldo N Segal
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA; Department of Medicine, Division of Pulmonary and Critical Care Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Kenneth A Stapleford
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Kamal M Khanna
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA; Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA.
| |
Collapse
|
33
|
Villa de la Torre F, Tec Caamal EA, Rizo AM, Argáez RB, Pintor Romero VG, Yáñez-Barrientos E, Morales-Tirado DJ, Alonso-Castro AJ, Arana Argáez VE. In vivo and in vitro anti-inflammatory activity of the methanolic leaves extract of Gymnopodium floribundum Rolfe. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119743. [PMID: 40204249 DOI: 10.1016/j.jep.2025.119743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/11/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gymnopodium floribundum Rolfe, known locally as "Dzidzilche" or "Ts'its'ilche," is a native species from Mexico and Central America. In Mayan communities, this plant is used to relieve inflammation and diverse respiratory diseases such as colds, catarrh, bronchitis, and asthma. Usually, a decoction of leaves or flowers is prepared and administered orally. AIM This research explores the anti-inflammatory effects of the methanol extract of Gymnopodium floribundum Rolfe leaves (MGF) using in vitro and in vivo animal models of inflammation. METHODS MGF was characterized by GC-MS, and cytotoxicity was assessed using hemolysis and MTT assays. The antiphlogistic effect in vitro was measuring the release of cytokines, hydrogen peroxide, and nitric oxide in macrophages stimulated with LPS. Additionally, anti- and pro-inflammatory cytokines, prostaglandins, and leukotrienes in serum were quantified in carrageenan-induced mouse paw edema. Finally, 1-fluoro-2,4-dinitrobenzene (DNFB)-induced delayed-type hypersensitivity and TPA-induced ear edema models were analyzed. RESULTS Compounds found in MGF, such as D-pinitol and protocatechuic (3,4-dihydroxybenzoic) acid, are reported to exert anti-inflammatory effects. MGF showed no hemolytic or cytotoxic effects. Nevertheless, it displayed in vitro anti-inflammatory activity by decreasing the release of IL-6, IL-1β, TNF-α, hydrogen peroxide, and nitric oxide levels; on the other hand, it increased IL-10 production. Furthermore, the MGF significantly reduced inflammation in mouse models and reduced the release of leukotrienes, prostaglandins, and pro-inflammatory cytokines. CONCLUSION Gymnopodium floribundum Rolfe exhibits anti-inflammatory activity by suppressing pro-inflammatory mediators, altering cell migration mechanisms, and raising IL-10 production.
Collapse
Affiliation(s)
- Fabiola Villa de la Torre
- Faculty of Chemistry, Laboratory of Pharmacology, Autonomous University of Yucatan, Yucatan, Mexico.
| | | | - Abril Martínez Rizo
- Faculty of Chemistry, Laboratory of Pharmacology, Autonomous University of Yucatan, Yucatan, Mexico.
| | - Rocío Borges Argáez
- Biotechnology Unit, Scientific Research Center of Yucatan (CICY), Yucatan, Mexico.
| | | | - Eunice Yáñez-Barrientos
- Division of Natural and Exact Sciences, Department of Chemistry, University of Guanajuato, Guanajuato, Mexico.
| | | | - Angel Josabad Alonso-Castro
- Division of Natural and Exact Sciences, Department of Chemistry, University of Guanajuato, Guanajuato, Mexico.
| | - Víctor E Arana Argáez
- Faculty of Chemistry, Laboratory of Pharmacology, Autonomous University of Yucatan, Yucatan, Mexico.
| |
Collapse
|
34
|
Shi Q, Tian T, Li Y. Identification of key genes and immune infiltration mechanisms in limb ischemia-reperfusion injury: a bioinformatics and experimental study. Front Immunol 2025; 16:1491928. [PMID: 40416982 PMCID: PMC12098047 DOI: 10.3389/fimmu.2025.1491928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 04/14/2025] [Indexed: 05/27/2025] Open
Abstract
Aim of the study To establish a cross-tissue bioinformatics model for identifying conserved key genes and immune infiltration mechanisms in ischemia-reperfusion injury (IRI) with experimental validation in limb IRI, including pharmacological targeting of the WNT5A/PLC pathway. Materials and methods Transcriptomic data from CTGF-stimulated cardiac myocytes (GSE36073) were analyzed as a surrogate for limb IRI due to shared pathological mechanisms. Random forest, LASSO regression, algorithms identified feature genes, validated in a rat limb IRI model using RT-qPCR, and histology. Pharmacological inhibition (WNT5A inhibitor Box5, PLC inhibitor U-73122) was performed to assess pathway involvement. Immune cell infiltration patterns were analyzed via CIBERSORT. Results From 169 differentially expressed genes (116 upregulated, 53 downregulated), machine learning identified four key genes (WNT5A, PLCG, ITPR1, CAMK2A), significantly upregulated in experimental limb IRI (P<0.01). Pharmacological inhibition confirmed their functional roles: Box5 and U-73122 treatment reduced expression of WNT5A and PLC versus IRI controls (P<0.05), showing IRI-induced muscle fiber disruption, edema, and inflammation. Immune analysis revealed myeloid polarization shifts (increased M1, decreased M2 macrophages; P<0.05). WNT5A correlated negatively with memory immune cells, while PLCG, ITPR1, and CAMK2A correlated with lymphocyte subpopulations. Conclusion We identified a conserved molecular signature across cardiac and skeletal muscle IRI, with WNT5A/PLC pathway components as mechanistically validated therapeutic targets. Our cross-tissue bioinformatic approach, reinforced by pharmacological and histological evidence, provides a novel framework for IRI analysis when direct patient data are unavailable. Combined targeting of macrophage polarization and cellular activation the WNT5A/PLC axis may offer synergistic therapeutic potential.
Collapse
Affiliation(s)
- Qiyun Shi
- Medical Center of Cervical and Lumbar Pain (Tui Na Therapy), Luoyang Orthopedic-Traumatological Hospital Of Henan Province (Henan Provincial Orthopedic Hospital), Luoyang, China
| | - Taotao Tian
- Medical Center of Cervical and Lumbar Pain (Tui Na Therapy), Luoyang Orthopedic-Traumatological Hospital Of Henan Province (Henan Provincial Orthopedic Hospital), Luoyang, China
| | - Yanfeng Li
- Medical Center of Microsurgery, Luoyang Orthopedic-Traumatological Hospital Of Henan Province (Henan Provincial Orthopedic Hospital), Zhengzhou, China
| |
Collapse
|
35
|
Xiong B, Wang H, Song YX, Lan WY, Li J, Wang F. Natural saponins and macrophage polarization: Mechanistic insights and therapeutic perspectives in disease management. Front Pharmacol 2025; 16:1584035. [PMID: 40417220 PMCID: PMC12098594 DOI: 10.3389/fphar.2025.1584035] [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: 02/26/2025] [Accepted: 03/24/2025] [Indexed: 05/27/2025] Open
Abstract
Macrophage polarization plays a pivotal role in immune homeostasis and disease progression across inflammatory, neoplastic, and metabolic disorders. Saponins, which are natural compounds with steroidal/triterpenoid structures, demonstrate therapeutic potential through immunomodulatory, anti-inflammatory, and anti-tumor activities. This study aims to highlight the potential of key saponins-such as ginsenosides, astragaloside IV, dioscin, platycodin D, pulsatilla saponins, and panax notoginseng saponins-in modulating macrophage polarization and enhancing conventional therapies, particularly in oncology. We conducted structured searches in PubMed, Google Scholar, and SciFinder (2013-2024) using controlled vocabulary, including "saponins," "macrophage polarization," and "therapeutic effects." Our findings demonstrate that saponins significantly modulate immune responses and improve treatment efficacy. However, clinical translation is hindered by challenges such as poor bioavailability and safety concerns, which limit systemic exposure and therapeutic utility. To overcome these barriers, innovative delivery strategies, including nanoemulsions and engineered exosomes, are essential for enhancing pharmacokinetics and therapeutic index. Future research should prioritize elucidating the molecular mechanisms underlying saponin-mediated macrophage polarization, identifying novel therapeutic targets, and optimizing drug formulations. Addressing these challenges will enable the restoration of immune balance and more effective management of diverse diseases.
Collapse
Affiliation(s)
- Beibei Xiong
- Department of Oncology, The First People’s Hospital of Shuangliu District, Chengdu, China
| | - Huan Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi-Xuan Song
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wen-Ying Lan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | | | - Fang Wang
- Chengdu First People’s Hospital, Chengdu, China
| |
Collapse
|
36
|
Mao B, Tian M, Yin Y, Li L, Li J, Wei D, Fu W. A Novel Injectable Cell-Loaded Hydrogel System for Cartilage Repair: In Vivo and In Vitro Study. Tissue Eng Part A 2025. [PMID: 40340534 DOI: 10.1089/ten.tea.2025.0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025] Open
Abstract
Polyhydroxyalkanoates are promising biomaterials, but their application in cartilage repair is still limited. In this study, an injectable thermosensitive hydrogel poly(3-hydroxybutyrate-co-3-hydroxyvalerate-co-3-hydroxyhexanoate)-Polyethylene Glycol (PEG)/hyaluronic acid/kartogenin was prepared from 3-hydroxybutyrate, 3-hydroxyvalerate, 3-hydroxyhexanoate, hyaluronic acid, and kartogenin. The hydrogels are porous, temperature-sensitive, and hydrophilic and have good compressive modulus. Mesenchymal stem cells derived from peripheral blood can proliferate on the hydrogels under two- and three-dimensional cultures. In addition, the hydrogel has the ability to induce chondrogenic differentiation of stem cells and induce M2 differentiation of macrophages. The hydrogel loaded with peripheral blood mesenchymal stem cells can repair cartilage defects in the knee joints of New Zealand rabbits and the newly formed cartilage was identified as type II collagen. Overall, this newly developed system could provide a new treatment option for repairing cartilage defects. Impact Statement In this study, poly(3-hydroxybutyrate-co-3-hydroxyvalerate-co-3-hydroxyhexanoate) was modified with hyaluronic acid and kartogenin to synthesize a thermosensitive injectable hydrogel scaffold. The scaffold has anti-inflammatory and cartilage-promoting effects. This study used the scaffold to carry peripheral blood mesenchymal stem cells to repair cartilage defects in rabbit knee joints, providing a new idea for the treatment of cartilage defects.
Collapse
Affiliation(s)
- Beini Mao
- Department of Rehabilitation Medicine, Shenzhen Hospital, Southern Medical University, Shenzhen City, People's Republic of China
- International Sports Medicine and Rehabilitation Center, Shenzhen Hospital, Southern Medical University, Shenzhen City, People's Republic of China
- Department of Occupational Therapy, School of Rehabilitation Sciences, Southern Medical University, Shenzhen City, People's Republic of China
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Ming Tian
- Civil Aviation General Hospital, Chaoyang District, Beijing, People's Republic of China
| | - Yuling Yin
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Lang Li
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jian Li
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Daixu Wei
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, People's Republic of China
| | - Weili Fu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
37
|
Sprenger-Svačina A, Svačina MKR, Otlu HG, Gao T, Sheikh KA, Zhang G. Endoneurial immune interplay in peripheral nerve repair: insights and implications for future therapeutic interventions. Front Neurosci 2025; 19:1602112. [PMID: 40415889 PMCID: PMC12098419 DOI: 10.3389/fnins.2025.1602112] [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: 03/28/2025] [Accepted: 04/16/2025] [Indexed: 05/27/2025] Open
Abstract
The mechanisms underlying axonal injury and repair in peripheral nerves, whether due to traumatic damage or autoimmune neuropathies, are complex and not yet fully understood. Recent research indicates that an orchestrated interplay between damaged neurons, Schwann cells, and especially endoneurial immune cells such as macrophages or T cells is crucial to achieve satisfactory nerve recovery. Following axonal injury, degenerating axons and reactive Schwann cells release chemoattractants and cytokines that recruit immune cells into the endoneurium. Among them, macrophages play a pivotal role by clearing axonal and myelin debris and subsequently creating a pro-regenerative microenvironment that supports axonal outgrowth. There is evidence that the timely switch of a pro-inflammatory M1 toward a pro-regenerative M2 macrophage polarization state is crucial for satisfactory nerve recovery, and supportive cellular and humoral factors that influence the endoneurial microenvironment, such as T cells and their cytokines, can substantially impact this fragile recovery process. The latter explains the limited nerve recovery in immune neuropathies, where a pathologic pro-inflammatory shift within the endoneurial immune cell signature hampers axonal outgrowth. This review aims to provide insights into cellular and humoral determinants of the endoneurial microenvironment during nerve damage and repair, which are assumed to hold substantial potential for future therapeutic interventions, especially since current strategies to enhance peripheral nerve recovery are limited to either surgical interventions in traumatic neuropathies or immunomodulatory drugs in immune neuropathies that often fail to achieve satisfactory functional results.
Collapse
Affiliation(s)
- Alina Sprenger-Svačina
- Neuromuscular Research Laboratory, Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Department of Neurology, Faculty of Medicine, University Hospital of Cologne, Cologne, Germany
| | - Martin K. R. Svačina
- Neuromuscular Research Laboratory, Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Philipps University Marburg and Department of Neurology, University Hospital Gießen and Marburg, Marburg, Germany
| | - Husniye G. Otlu
- Neuromuscular Research Laboratory, Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Vocational Health Sciences, Laboratory Techniques Program, Malatya Turgut Ozal University, Malatya, Türkiye
| | - Tong Gao
- Neuromuscular Research Laboratory, Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Kazim A. Sheikh
- Neuromuscular Research Laboratory, Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Gang Zhang
- Neuromuscular Research Laboratory, Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
38
|
Ivy A, Bess SN, Agrawal S, Kochar V, Stokes AL, Muldoon TJ, Nelson CE. A dual-fluorescence assay for gene delivery vehicle screening in macrophages with an inflammation-inducible reporter construct. BMC METHODS 2025; 2:8. [PMID: 40352095 PMCID: PMC12062070 DOI: 10.1186/s44330-025-00030-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 05/01/2025] [Indexed: 05/14/2025]
Abstract
Background Macrophages are a promising target for therapeutics in various applications such as regenerative medicine and immunotherapy for cancer. Due to their plastic nature, macrophages can switch from a non-activated state to activated with the smallest environmental change. For macrophages to be effective in their respective applications, screening for phenotypic changes is necessary to elucidate the cell response to different delivery vehicles, vaccines, small molecules, and other stimuli. Methods We created a sensitive and dynamic high-throughput screening method for macrophages based on the activation of NF-κB. For this reporter, we placed an mRFP1 fluorescence gene under the control of an inflammatory promoter, which recruits NF-κB response elements to promote expression during the inflammatory response in macrophages. We characterized the inflammatory reporter based on key markers of an inflammatory response in macrophages including TNF-α cytokine release and immunostaining for inflammatory and non-inflammatory cell surface markers. We compared gene delivery and inflammation of several clinically relevant viral vehicles and commercially available non-viral vehicles. Statistical analysis between groups was performed with a one-way ANOVA with post-hoc Tukey's test. Results The reporter macrophages demonstrated a dynamic range after LPS stimulation with an EC50 of 0.61 ng/mL that was highly predictive of TNF-α release. Flow cytometry revealed heterogeneity between groups but confirmed population level shifts in pro-inflammatory markers. Finally, we demonstrated utility of the reporter by showing divergent effects with various leading gene delivery vehicles. Discussion This screening technique developed here provides a dynamic, high-throughput screening technique for determining inflammatory response by mouse macrophages to specific stimuli. The method presented here provides insight into the inflammatory response in mouse macrophages to different viral and non-viral gene delivery methods and provides a tool for high-throughput screening of novel vehicles. Supplementary Information The online version contains supplementary material available at 10.1186/s44330-025-00030-x.
Collapse
Affiliation(s)
- Allie Ivy
- Department of Biomedical Engineering, University of Arkansas, 120 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
| | - Shelby N. Bess
- Department of Biomedical Engineering, University of Arkansas, 120 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
| | - Shilpi Agrawal
- Department of Biomedical Engineering, University of Arkansas, 120 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
| | - Varun Kochar
- Department of Biomedical Engineering, University of Arkansas, 120 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
| | - Abbey L. Stokes
- Department of Biomedical Engineering, University of Arkansas, 120 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
| | - Timothy J. Muldoon
- Department of Biomedical Engineering, University of Arkansas, 120 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR USA
| | - Christopher E. Nelson
- Department of Biomedical Engineering, University of Arkansas, 120 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR USA
| |
Collapse
|
39
|
He T, Tan Q, Huang Y, Chen J, Tan J, Zhou C, Xu L, Nie R, Zhang Q, Liang P, Lv Q, Xie HQ. Extracellular Adipose Matrix Hydrogel Laden with Adipose-Derived Stem Cell Modulates Macrophage Polarization for Enhanced Full-Thickness Skin Wound Repair. Biomacromolecules 2025. [PMID: 40340431 DOI: 10.1021/acs.biomac.5c00194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Adipose-derived stem cells (ADSC) represent a promising approach for wound healing, while the limited survival rate has restricted their application. To address this, we equipped a hydrogel from acellular porcine adipose tissue (HAPA) with ADSC to fabricate the HAPA + ADSC composite hydrogel. In addition to serving as a carrier for stem cell delivery, the bioactive components of the HAPA hydrogel support immune regulation and tissue repair. In this study, we demonstrated that the HAPA + ADSC composite could effectively modulate macrophage polarization, promote angiogenesis, and regulate extracellular matrix (ECM) deposition and remodeling, thereby substantially accelerating wound healing. Additionally, transcriptomic sequencing analysis indicated that the HAPA + ADSC composite upregulated Nfkbia and Nfkbie to inhibit the nuclear transcription of RelA-p50 heterodimer so that macrophages polarization toward an M1 phenotype is suppressed. The combined effects of ADSC and HAPA hydrogel make it a promising candidate for functional skin wound healing.
Collapse
Affiliation(s)
- Tao He
- Division of Breast Center and Breast Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Day Surgery Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qiuwen Tan
- Division of Breast Center and Breast Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yizhou Huang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jun Chen
- Department of Orthopedics and Laboratory of Orthopaedics, Wuhan Fourth Hospital, Wuhan, Hubei 430000, China
| | - Jie Tan
- Department of Orthopedics and Laboratory of Orthopaedics, Wuhan Fourth Hospital, Wuhan, Hubei 430000, China
| | - Chen Zhou
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Li Xu
- Division of Breast Center and Breast Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Rong Nie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qingyi Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Peng Liang
- Day Surgery Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qing Lv
- Division of Breast Center and Breast Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hui-Qi Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
40
|
Lyu Z, Wu Y, Hu F, Zheng X, Ma D, Xu Z, Ding Y, Liu X, Huo S. Controlled release of ionic carrier hydrogels for sequential immunomodulation to facilitate stage-specific treatment of infectious wound. Biomaterials 2025; 322:123376. [PMID: 40349534 DOI: 10.1016/j.biomaterials.2025.123376] [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: 03/28/2025] [Accepted: 04/24/2025] [Indexed: 05/14/2025]
Abstract
Infected wounds present a significant clinical challenge, exacerbated by antibiotic resistance, which complicates effective treatment. This study introduces a hydrogel (CC/AP@CM) embedded with core-shell bioactive glass nanoparticles designed for the controlled, sequential release of copper (Cu2+) and magnesium (Mg2+) ions. The hydrogel is crosslinked via a Schiff base reaction, endowing it with injectable, self-healing, and adhesive properties. Notably, the bilayer structure of the bioactive glass within the hydrogel allows an initial release of Cu2+ ions to trigger an early-stage pro-inflammatory and antimicrobial response, followed by Mg2+ ions that support tissue repair and an anti-inflammatory environment. This design aligns with natural wound healing stages, promoting a shift in macrophage polarization from the M1 to M2 phenotype, effectively balancing antibacterial defense with tissue regeneration. The hydrogel demonstrated robust antibacterial efficacy against MRSA, increased angiogenesis, and enhanced fibroblast proliferation and migration in vitro. In a murine wound model, it significantly accelerated wound closure and immune activation, including responses from dendritic cells and T cells. These findings suggest that this hydrogel, through its stage-specific immunomodulatory properties and temporally controlled ion release, offers a promising strategy for treating complex wound infections, supporting both immune defense and tissue healing.
Collapse
Affiliation(s)
- Zhuocheng Lyu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yuezhou Wu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Fei Hu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xu Zheng
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Dajun Ma
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zhenjiang Xu
- Department of Orthopedic Surgery Spine Center Changzheng Hospital Navy Medical University, Shanghai, China
| | - Yurun Ding
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Xuesong Liu
- Department of Ultrasound, Renji Hospital, School of Medicine, Shanghai Jiaotong University, China.
| | - Shicheng Huo
- Department of Orthopedic Surgery Spine Center Changzheng Hospital Navy Medical University, Shanghai, China.
| |
Collapse
|
41
|
Zheng C, Wu Y, Luan F, Wei C, Zhang C, Liu W, Wang W, Chen J. Advances in biomimetic hydrogel for articular cartilage defect repair: Enabling immunomodulation and chondrogenesis. Colloids Surf B Biointerfaces 2025; 253:114760. [PMID: 40359898 DOI: 10.1016/j.colsurfb.2025.114760] [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: 02/19/2025] [Revised: 04/22/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025]
Abstract
Articular cartilage defects, as a core pathologic feature in the progression of osteoarthritis, and their irreversible degenerative changes lead to functional impairment and socioeconomic burden for tens of millions of patients worldwide. Hydrogels have become key biomaterials in cartilage regeneration with the three-dimensional network structure, programmable mechanical properties, and cell-adaptive microenvironment of biomimetic extracellular matrix. In recent years, several hydrogel systems with in vitro/in vivo repair potential have been developed by modulating the material topology, dynamic mechanical response, and delivery of bioactive factors, and some of them have entered the clinical translation stage. This review systematically explains the biomimetic design principles of hydrogels. It analyzes the immunomodulation and chondrogenic mechanisms mediated by hydrogels, providing a theoretical framework for the development of next-generation smart cartilage repair materials.
Collapse
Affiliation(s)
- Chenxiao Zheng
- Department ofOrthopaedics and Traumatology, Zhongshan Hospital of Traditional ChineseMedicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, Guangdong 528401, China
| | - Yurui Wu
- Department ofOrthopaedics and Traumatology, Zhongshan Hospital of Traditional ChineseMedicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, Guangdong 528401, China
| | - Feifan Luan
- Department ofOrthopaedics and Traumatology, Zhongshan Hospital of Traditional ChineseMedicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, Guangdong 528401, China
| | - Chunwei Wei
- Department ofOrthopaedics and Traumatology, Zhongshan Hospital of Traditional ChineseMedicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, Guangdong 528401, China
| | - Chunye Zhang
- Biomedical and HealthTechnology Innovation Platform, National University of Singapore (Suzhou)Research Institute, Suzhou, Jiangsu 215123, China
| | - Wenjun Liu
- Zhejiang ShangyueBiotechnology Research Center, Hangzhou, Zhejiang 310018, China
| | - Wenjun Wang
- Biomedical and HealthTechnology Innovation Platform, National University of Singapore (Suzhou)Research Institute, Suzhou, Jiangsu 215123, China.
| | - Jiayi Chen
- Department ofOrthopaedics and Traumatology, Zhongshan Hospital of Traditional ChineseMedicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, Guangdong 528401, China.
| |
Collapse
|
42
|
Wang Y, Gao J, Wu T, Wang Z. M2 Macrophages Mitigate Ocular Surface Inflammation and Promote Recovery in a Mouse Model of Dry Eye. Ocul Immunol Inflamm 2025:1-10. [PMID: 40327794 DOI: 10.1080/09273948.2025.2497484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 03/27/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025]
Abstract
PURPOSE Dry eye disease (DED) is a chronic, progressive, multifactorial condition characterized by tear film instability and ocular surface damage. Ocular surface inflammation is one of the main mechanisms of DED. This study aims to investigate the therapeutic effects of anti-inflammatory M2 macrophages on ocular surface inflammation and their potential mechanisms in improving dry eye symptoms in a mouse model. METHODS Mouse macrophages (RAW264.7) were polarized into M2 macrophages by IL-4 under different osmolarities, and M2 macrophage conditioned medium (M2-CM) was collected. Flow cytometry and ELISA were applied to measure the cytokine expression of the M2 macrophages. Primary mouse corneal epithelial cells (CECs) were co-cultured with RAW264.7 and M2 macrophages using a Transwell system. The viability and migration of CECs were assessed using CCK-8 and scratch assays. Mouse DED was established by subcutaneous injection of scopolamine, and the therapeutic effects of M2-CM were evaluated by phenol red thread test, fluorescein staining, and tear film breakup time (BUT). PCR and immunofluorescence staining were applied to observe inflammatory factors and cells on the ocular surface. RESULTS M2 macrophages enhanced CEC viability, proliferation, and migration, but hyperosmolarity inhibited M2 macrophage polarization. In the DED model, M2-CM improved ocular surface conditions, reduced pro-inflammatory cytokine expression, and increased anti-inflammatory factors. Immunofluorescence revealed reduced pro-inflammatory cells (M1 macrophages, Th1, and Th17) and increased M2 macrophages in the ocular tissues after M2-CM treatment. CONCLUSION These results suggest that M2-CM ameliorates ocular surface inflammation and promotes recovery in DED, offering a potential therapeutic strategy for DED.
Collapse
Affiliation(s)
- Yingming Wang
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jing Gao
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Tianhong Wu
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhenyu Wang
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
43
|
Froom ZSCS, Callaghan NI, Davenport Huyer L. Cellular crosstalk in fibrosis: insights into macrophage and fibroblast dynamics. J Biol Chem 2025:110203. [PMID: 40334985 DOI: 10.1016/j.jbc.2025.110203] [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: 03/13/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
Pathological fibrosis, the excessive deposition of extracellular matrix and tissue stiffening that causes progressive organ dysfunction, underlies diverse chronic diseases. The fibrotic microenvironment is driven by the dynamic microenvironmental interaction between various cell types; macrophages and fibroblasts play central roles in fibrotic disease initiation, maintenance, and progression. Macrophage functional plasticity to microenvironmental stimuli modulates fibroblast functionality by releasing pro-inflammatory cytokines, growth factors, and matrix remodeling enzymes that promote fibroblast proliferation, activation, and differentiation into myofibroblasts. Activated fibroblasts and myofibroblasts serve as the fibrotic effector cells, secreting extracellular matrix components and initiating microenvironmental contracture. Fibroblasts also modulate macrophage function through the release of their own pro-inflammatory cytokines and growth factors, creating bidirectional crosstalk that reinforces the chronic fibrotic cycle. The intricate interplay between macrophages and fibroblasts, including their secretomes and signaling interactions, leads to tissue damage and pathological loss of tissue function. In this review, we examine macrophage-fibroblast reciprocal dynamic interactions in pathological fibrotic conditions. We discuss the specific lineages and functionality of macrophages and fibroblasts implicated in fibrotic progression, with focus on their signal transduction pathways and secretory signalling that enables their pro-fibrotic behaviour. We then finish with a set of recommendations for future experimentation with the goal of developing a set of potential targets for anti-fibrotic therapeutic candidates. Understanding the cellular interactions between macrophages and fibroblasts provides valuable insights into potential therapeutic strategies to mitigate fibrotic disease progression.
Collapse
Affiliation(s)
- Zachary S C S Froom
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Neal I Callaghan
- Department of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Locke Davenport Huyer
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada; Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; Department of Biomaterials & Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, NS B3H 4R2, Canada; Nova Scotia Health, Halifax, NS B3S 0H6, Canada.
| |
Collapse
|
44
|
Bai X, Guo YR, Zhao ZM, Li XY, Dai DQ, Zhang JK, Li YS, Zhang CD. Macrophage polarization in cancer and beyond: from inflammatory signaling pathways to potential therapeutic strategies. Cancer Lett 2025; 625:217772. [PMID: 40324582 DOI: 10.1016/j.canlet.2025.217772] [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: 02/26/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
Macrophages are innate immune cells distributed throughout the body that play vital roles in organ development, tissue homeostasis, and immune surveillance. Macrophages acquire a binary M1/M2 polarized phenotype through signaling cascades upon sensing different signaling molecules in the environment, thereby playing a core role in a series of immune tasks, rendering precise regulation essential. M1/M2 macrophage phenotypes regulate inflammatory responses, while controlled activation of inflammatory signaling pathways is involved in regulating macrophage polarization. Among the relevant signaling pathways, we focus on the six well-characterized NF-κB, MAPK, JAK-STAT, PI3K/AKT, inflammasome, and cGAS-STING inflammatory pathways, and elucidate their roles and crosstalk in macrophage polarization. Furthermore, the effects of many environmental signals that influence macrophage polarization are investigated by modulating these pathways in vivo and in vitro. We thus detail the physiological and pathophysiological status of these six inflammatory signaling pathways and involvement in regulating macrophage polarization in cancer and beyond, as well as describe potential therapeutic approaches targeting these signaling pathways. In this review, the latest research advances in inflammatory signaling pathways regulating macrophage polarization are reviewed, as targeting these inflammatory signaling pathways provides suitable strategies to intervene in macrophage polarization and various tumor and non-tumor diseases.
Collapse
Affiliation(s)
- Xiao Bai
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Yun-Ran Guo
- Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Zhe-Ming Zhao
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Xin-Yun Li
- Clinical Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Dong-Qiu Dai
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Cancer Center, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Jia-Kui Zhang
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Yong-Shuang Li
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Chun-Dong Zhang
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Central Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| |
Collapse
|
45
|
Ma S, Ni J, Ye D, Kuang Y, Wang Z, Yang L. Human umbilical cord mesenchymal stem cells improve the survial of flaps by promoting angiogenesis in mice. Eur J Med Res 2025; 30:356. [PMID: 40312717 PMCID: PMC12046903 DOI: 10.1186/s40001-025-02602-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 04/15/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Flap necrosis post-operation disturbs surgeons during plastic and reconstructive surgery. This is caused by hypoperfusion and subsequent ischemia-reperfusion injury, where restricted blood flow followed by restored circulation paradoxically exacerbates tissue damage. Mesenchymal stem cells, which show multidirectional differentiation, provide hematopoietic support and are involved in immune regulation and anti-fibrosis, have inspired research on improving the blood supply of flaps. METHODS Primary human umbilical cord mesenchymal stem cells (HuMSCs), were obtained and subcultured for expansion. The cells of the third generation were incubated in a gelatin sponge. Thirty Kunming mice were randomly divided into three groups, and saline, HuMSCs, and HuMSCs-CM were injected preoperatively into the skin of the back. The vessel density was assessed on the tenth day. Forty-eight Kunming mice were divided into two groups. Group A was subdivided into the saline group, HuMSCs, and HuMSCs-CM groups and pretreated as described above. In Group B, the intervention was changed from injection to subcutaneous embedding. Random flaps were made on the back in both groups on the tenth day after pretreatment. The survival rate of the flap was calculated on the seventh day. RESULTS HuMSCs-CM significantly increased the microvessel density on the tenth day after pretreatment. The flap survival rate was higher in the cell and CM groups, rising from approximately 13% to 60% in Group A, and to about 75% in Group B. Moreover, subcutaneous embedding of cell-carrying gelatin sponges improved flap survival compared to other interventions. CONCLUSION Improved cell incubation conditions can enhance its utility. The application of HuMSCs and their conditioned medium promoted the survival of the flap by inducing neovasculogenesis.
Collapse
Affiliation(s)
- Siyi Ma
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jintao Ni
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Danyan Ye
- Research Center for Translational Medicine, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yuping Kuang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zhixia Wang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Lujun Yang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China.
- Research Center for Translational Medicine, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China.
| |
Collapse
|
46
|
Wang X, Liu C, Wang M, Yin B, Ge Y, Shu L, Sun H, Zhang W. Multi-modal microcarriers reprogram mitochondrial metabolism and activate efferocytosis in macrophages for osteoporotic bone repair. Biomaterials 2025; 322:123384. [PMID: 40319678 DOI: 10.1016/j.biomaterials.2025.123384] [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: 11/25/2024] [Revised: 04/15/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Osteoporotic bone repair remains challenging due to the ineffectiveness of traditional bone repair materials in adapting to the complex immune microenvironment of aging bone tissue. Exploiting the key role of macrophages in regulating this immune environment through the rational design of osteoimmunomodulatory biomaterials has emerged as a promising approach. However, current designs inadequately address the complexity of macrophage functions in aging environments, resulting in suboptimal regulatory effects. Hence, we explored multi-modal microcarriers for enhancing macrophage functionality. In this work, we developed a VGX-1027-loaded mesoporous silica nanosphere composite PLLA microcarrier. The dual-carrier system, featuring a micro-nano hybrid design by spatially separating the mesoporous silica nanoparticles and PLLA microspheres, enables sustained intracellular release of VGX-1027, addressing the chronic nature of osteoporotic fractures. Our studies demonstrate this VGX-1027 microcarrier (PMVGX) promotes M2 macrophage polarization by reprogramming mitochondrial metabolism. Simultaneously, it enhances efferocytosis, facilitating the clearance of dead or senescent cells and reducing inflammatory responses, thus reshaping the aging osteoimmunomodulatory. Furthermore, PMVGX induces macrophages to release osteogenic exosomes containing miR-5106 through paracrine signaling, significantly enhancing osteogenic function. In a postmenopausal osteoporosis animal model, PMVGX exhibited remarkable efficacy in repairing osteoporotic bone defects. This proof-of-concept study demonstrates that our multi-modal microcarrier effectively regulates macrophage functions via mitochondrial homeostasis, efferocytosis, and exosome content, offering great potential for osteoporotic bone repair.
Collapse
Affiliation(s)
- Xin Wang
- National Center for Orthopaedics, Shanghai Sixth People's Hospital, Shanghai, 200233, China; Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Chenjun Liu
- National Center for Orthopaedics, Shanghai Sixth People's Hospital, Shanghai, 200233, China; Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Mingyue Wang
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Bohao Yin
- National Center for Orthopaedics, Shanghai Sixth People's Hospital, Shanghai, 200233, China; Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yuwei Ge
- National Center for Orthopaedics, Shanghai Sixth People's Hospital, Shanghai, 200233, China; Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Linyuan Shu
- Department of Emergency Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Hui Sun
- National Center for Orthopaedics, Shanghai Sixth People's Hospital, Shanghai, 200233, China; Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Wei Zhang
- National Center for Orthopaedics, Shanghai Sixth People's Hospital, Shanghai, 200233, China; Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
47
|
Chen Z, Yang J, Zhang Q, Zeng W, Liu Y, Hu W, Chen L, Shen J, Miao Y, Xiao Z, Wu Z, Wang H, Shen H, Ding C, Chen Q, Zhao J, Yang Y. Inhalable myofibroblast targeting nanoparticles for synergistic treatment of pulmonary fibrosis. SCIENCE ADVANCES 2025; 11:eadv9571. [PMID: 40305619 PMCID: PMC12042884 DOI: 10.1126/sciadv.adv9571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/25/2025] [Indexed: 05/02/2025]
Abstract
Pulmonary fibrosis (PF) is a life-threatening interstitial lung disease, characterized by excessive fibroblast activation and collagen deposition, leading to progressive pulmonary function decline and limited therapeutic efficacy. Here, the inhalable, myofibroblast-targeted, and pH-responsive liposomes (FL-NI) were developed for effective codelivery of nintedanib, a mainstream antifibrotic drug in clinic, and siIL11, a small interfering RNA that silences the key profibrosis cytokine IL-11. Notably, FL-NI achieved a 117.8% increase in pulmonary drug delivery by noninvasive inhalation and a 71.5% increase in delivery specifically to fibroblast activation protein-positive myofibroblasts while reducing nonspecific immune cell and epithelial uptake by 29.8 and 55.8%, respectively. The accurate inhalation codelivery of nintedanib and siIL11 into myofibroblasts achieved synergistic effects, effectively enhanced myofibroblast deactivation, reduced pathological collagen deposition by 50.8%, and promoted epithelial tissue repair. FL-NI remodeled the aberrant immune microenvironment without inducing systemic toxicities. Therefore, this work demonstrated the notable potential for this pluripotent strategy for improving PF outcomes and its promising clinical translation.
Collapse
Affiliation(s)
- Zhike Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices Soochow University, Suzhou 215123, China
| | - Jian Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qiang Zhang
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices Soochow University, Suzhou 215123, China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- School of Materials Science and Engineering, Tongji University, Shanghai 201804, China
| | - Weibiao Zeng
- Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yi Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices Soochow University, Suzhou 215123, China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Wenxuan Hu
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Linfu Chen
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices Soochow University, Suzhou 215123, China
| | - Jingjing Shen
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices Soochow University, Suzhou 215123, China
| | - Yu Miao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices Soochow University, Suzhou 215123, China
| | - Zhisheng Xiao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices Soochow University, Suzhou 215123, China
| | - Zhiqiang Wu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices Soochow University, Suzhou 215123, China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - He Wang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hui Shen
- Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Cheng Ding
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qian Chen
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices Soochow University, Suzhou 215123, China
| | - Jun Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- School of Materials Science and Engineering, Tongji University, Shanghai 201804, China
| |
Collapse
|
48
|
Zhan X, Bai Y, Zhu Q, Gao Y, Li F, Bu Q, Zhu Z, Rao Z, Zhou H. Macrophage ATG16L1 promotes liver regeneration after partial hepatectomy. JHEP Rep 2025; 7:101330. [PMID: 40290519 PMCID: PMC12023798 DOI: 10.1016/j.jhepr.2025.101330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 04/30/2025] Open
Abstract
Background & Aims Autophagy plays an important role in liver regeneration. However, most studies are limited to hepatocytes, and the function and mechanism of macrophage autophagy in liver regeneration remain unclear. This study investigated the role of the essential autophagy gene encoding autophagy-related 16-like 1 (ATG16L1), which regulates the macrophage phenotype in liver regeneration. Methods We generated FloxP-Atg16l1 (Atg16l1 FL/FL ), Lyz2-Cre Atg16l1 knockout (KO) (Atg16l1 M-KO ), and myeloid-specific Atg16l1-overexpression-knock-in (Atg16l1 OE ) mice. These mice were subjected to 70% partial hepatectomy to demonstrate the role of ATG16L1 in macrophages during liver regeneration. Results ATG16L1 expression was significantly upregulated in macrophages during the early stages of liver regeneration. ATG16L1 deletion in macrophages substantially delayed liver regeneration in mice and caused a marked imbalance in Ly6Chi and Ly6Clo macrophage proportions in the liver. RNA-sequencing analysis revealed that, compared with macrophages isolated from Atg16l1 FL/FL mice, those from Atg16l1 M-KO mice exhibited significant downregulation of genes associated with oxidative phosphorylation and upregulation of proinflammatory gene expression. Mechanistically, ATG16L1 loss impaired mitophagy in macrophages, leading to the accumulation of mitochondrial damage and a metabolic shift that promoted proinflammatory macrophage polarization. ATG16L1 deficiency not only promoted macrophage mitochondrial (mt)DNA release and cyclic GMP-AMP synthase-stimulator of interferon genes (STING) activation, but also suppressed STING degradation. Sustained STING hyperactivation and subsequent increased release of downstream interferons further contributed to the inhibition of liver regeneration. Notably, pharmacological activation or genetic overexpression of ATG16L1 significantly enhanced liver regeneration in mice. Conclusions ATG16L1 has a pivotal role in liver regeneration by affecting the phenotype and function of macrophages. Thus, targeting ATG16L1 in macrophages could present a novel strategy for promoting liver regeneration. Impact and implications The autophagy-related gene ATG16L1 mediates mitophagy, facilitating the clearance of damaged mitochondria in macrophages following partial hepatectomy and maintaining a reparative macrophage phenotype. ATG16L1 deficiency triggers excessive STING activation and inhibits its degradation, thereby suppressing liver regeneration. Thus, targeting ATG16L1 in macrophages could represent a novel strategy to promote liver regeneration.
Collapse
Affiliation(s)
- Xinyu Zhan
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Yan Bai
- Department of Anesthesiology, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, 210029, China
| | - Qing Zhu
- Department of Anesthesiology, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, 210029, China
| | - Yiyun Gao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Fan Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Qingfa Bu
- Department of General Surgery, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Zeyu Zhu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Zhuqing Rao
- Department of Anesthesiology, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, 210029, China
| | - Haoming Zhou
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| |
Collapse
|
49
|
Mezouar S, Mege J. Monitoring Macrophage Polarization in Infectious Disease, Lesson From SARS-CoV-2 Infection. Rev Med Virol 2025; 35:e70034. [PMID: 40148134 PMCID: PMC11976041 DOI: 10.1002/ird3.70006] [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: 03/11/2025] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
The concept of macrophage polarization has been largely used in human diseases to define a typology of activation of myeloid cells reminiscent of lymphocyte functional subsets. In COVID-19, several studies have investigated myeloid compartment dysregulation and macrophage polarization as an indicator of disease prognosis and monitoring. SARS-CoV-2 induces an in vitro activation state in monocytes and macrophages that does not match the polarization categories in most studies. In COVID-19 patients, monocytes and macrophages are activated but they do not show a polarization profile. Therefore, the investigation of polarization under basic conditions was not relevant to assess monocyte and macrophage activation. The analysis of monocytes and macrophages with high-throughput methods has allowed the identification of new functional subsets in the context of COVID-19. This approach proposes an innovative stratification of myeloid cell activation. These new functional subsets of myeloid cells would be better biomarkers to assess the risk of complications in COVID-19, reserving the concept of polarization for pharmacological programme evaluation. This review reappraises the polarization of monocytes and macrophages in viral infections, particularly in COVID-19.
Collapse
Affiliation(s)
- Soraya Mezouar
- Centre National de la Recherche ScientifiqueÉtablissement Français du SangAnthropologie Bio‐Culturelle, Droit, Éthique et SantéAix‐Marseille UniversityMarseilleFrance
- Faculty of Medical and Paramedical SciencesAix‐Marseille UniversityHIPE Human LabMarseilleFrance
| | - Jean‐Louis Mege
- Centre National de la Recherche ScientifiqueÉtablissement Français du SangAnthropologie Bio‐Culturelle, Droit, Éthique et SantéAix‐Marseille UniversityMarseilleFrance
- Department of ImmunologyLa Timone HospitalMarseilleFrance
| |
Collapse
|
50
|
Wang T, Wang X, Ren W, Sun Z, Zhang Y, Wu N, Diao H. Cardiomyocyte proliferation: Advances and insights in macrophage-targeted therapy for myocardial injury. Genes Dis 2025; 12:101332. [PMID: 39935606 PMCID: PMC11810708 DOI: 10.1016/j.gendis.2024.101332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 02/18/2024] [Accepted: 03/20/2024] [Indexed: 02/13/2025] Open
Abstract
In the mammalian heart, cardiomyocytes undergo a transient window of proliferation that leads to regenerative impairment, limiting cardiomyocyte proliferation and myocardial repair capacity. Cardiac developmental patterns exacerbate the progression of heart disease characterized by myocardial cell loss, ultimately leading to cardiac dysfunction and heart failure. Myocardial infarction causes the death of partial cardiomyocytes, which triggers an immune response to remove debris and restore tissue integrity. Interestingly, when transient myocardial injury triggers irreversible loss of cardiomyocytes, the subsequent macrophages responsible for proliferation and regeneration have a unique immune phenotype that promotes the formation of pre-existing new cardiomyocytes. During mammalian regeneration, mononuclear-derived macrophages and self-renewing resident cardiac macrophages provide multiple cytokines and molecular signals that create a regenerative environment and cellular plasticity capacity in postnatal cardiomyocytes, a pivotal strategy for achieving myocardial repair. Consistent with other human tissues, cardiac macrophages originating from the embryonic endothelium produce a hierarchy of contributions to monocyte recruitment and fate specification. In this review, we discuss the novel functions of macrophages in triggering cardiac regeneration and repair after myocardial infarction and provide recent advances and prospective insights into the phenotypic transformation and heterogeneous features involving cardiac macrophages. In conclusion, macrophages contribute critically to regeneration, repair, and remodeling, and are challenging targets for cardiovascular therapeutic interventions.
Collapse
Affiliation(s)
- Tao Wang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Xueyao Wang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Weibin Ren
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Zeyu Sun
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Yanhui Zhang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Nanping Wu
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Hongyan Diao
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| |
Collapse
|