1
|
Yang P, Chen X, Qin Y, Yu L, Ge G, Yin W, Zhang W, Li W, Li W, Xia W, Wu Z, Ding F, Bai J, Meng F, Geng D. Regulation of osteoimmune microenvironment via functional dynamic hydrogel for diabetic bone regeneration. Biomaterials 2025; 320:123273. [PMID: 40121832 DOI: 10.1016/j.biomaterials.2025.123273] [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: 11/23/2024] [Revised: 02/20/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Bone regeneration and repair face formidable challenges under diabetic conditions, primarily due to the disruption of macrophage polarization induced by diabetes and the inflammatory imbalance within the bone microenvironment. We have developed a novel dynamic hydrogel system (AG-CD@LINA), constructed through the coordination crosslinking of thiolated gelatin (SH-Gelatin) and gold ions (Au3+), followed by grafting with cyclodextrin to load the ligand linagliptin. This hydrogel effectively inhibits the formation of M1 macrophages and the expression of pro-inflammatory cytokines by gradually releasing linagliptin. Simultaneously, it promotes the formation of M2 macrophages and the expression of anti-inflammatory cytokines, thus improving the inflammatory microenvironment of diabetic bone defects. Consequently, it facilitates the migration of mesenchymal stem cells and angiogenic cells, augments osteogenic activity, and promotes vascularization, collectively accelerating the regeneration of diabetic bone tissue. Mechanistically, polarization occurs through the TLR3-NF-κB signaling pathway. In vivo experiments demonstrate that the in-situ injection of the hydrogel enhances the regeneration of bone tissue and the restoration of bone structure in diabetic bone defects, effectively modulating local inflammation and promoting vascular formation. This study suggests that functionalized dynamic hydrogels can improve the inflammatory microenvironment by regulating in situ macrophage polarization, thereby facilitating the reconstruction of bone microstructure. This approach represents a promising novel therapeutic strategy for diabetic bone defects.
Collapse
Affiliation(s)
- Peng Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China; Department of Orthopedics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215006, Jiangsu, China; Suzhou Key Laboratory of Orthopedic Medical Engineering, Suzhou, 215006, Jiangsu, China
| | - Xu Chen
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Yi Qin
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Lei Yu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Gaoran Ge
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Weiling Yin
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Wei Zhang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Wenming Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Wenhao Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Wenyu Xia
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Zebin Wu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Fan Ding
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Jiaxiang Bai
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230022, China.
| | - Fanwen Meng
- Department of Implant Dentistry, Suzhou Stomatological Hospital, Suzhou, 215005, China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
2
|
xin D, Junwen Z, Jiacheng Z, Qiongyu X, Huanzong H, peng L, juan L. In vitro and in vivo study of concentrated growth factor (CGF) mediating macrophage polarization in bone defect repair. Regen Ther 2025; 29:474-483. [PMID: 40337617 PMCID: PMC12056952 DOI: 10.1016/j.reth.2025.04.013] [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: 01/08/2025] [Revised: 04/07/2025] [Accepted: 04/17/2025] [Indexed: 05/09/2025] Open
Abstract
Concentrated growth factor (CGF) is widely applied in clinical practice, but whether it has bone promoting effects and its mechanism of action are still the focus of discussion. In this study, in vitro experiments demonstrate that CGF can promote the expression of Arg-1 in BMDM cells, facilitating their polarization towards the M2 macrophages and encouraging the secretion of IL-10 and VEGF-A. CGF modulates M1 macrophages by reducing the expression of iNOS, while enhancing Arg-1 expression, thereby converting them to M2 macrophages. This is accompanied by a decrease in the secretion of TNF- α and IL-1β, and an increase in the secretion of IL-10 and VEGF-A. Mechanistically, CGF promotes the phosphorylation of STAT3, which in turn induces M2 macrophage polarization, suggesting that the function of CGF-mediated macrophages may be associated with the STAT3 signaling pathway. Moreover, CGF-mediated macrophages were found to enhance osteoblast activity, increasing the expression of ALP, RUNX2, and BMP-2, and improving cell migration capabilities. In vivo experiments showed that CGF could early recruit M2 macrophages to the bone defect site, promoting the expression of bone formation-related proteins such as ALP and BMP-2, and accelerating bone tissue regeneration. In summary, our study demonstrates that CGF can induce bone repair and regeneration by promoting immune modulation and macrophage polarization.
Collapse
Affiliation(s)
- Deng xin
- Department of Stomatology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Zheng Junwen
- Department of Stomatology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Zou Jiacheng
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xun Qiongyu
- Pingshan District Central Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - He Huanzong
- Department of Stomatology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Li peng
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Liao juan
- Department of Stomatology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Department of Stomatology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
3
|
Shen F, Chen Y, Xu Z, Wang W, Chen G, Ye F. Inhibition of M2 macrophage-mediated mesenchymal stem cell migration: Boldine attenuates elbow heterotopic ossification. Int J Biochem Cell Biol 2025; 185:106787. [PMID: 40287052 DOI: 10.1016/j.biocel.2025.106787] [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: 01/22/2025] [Revised: 03/26/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Heterotopic ossification (HO) is characterized by abnormal bone formation in soft tissues, often following trauma or surgery. Transforming growth factor-beta (TGF-β) signaling and M2 macrophage polarization play critical roles in the recruitment and differentiation of mesenchymal stromal/progenitor cells (MSPCs), promoting HO. METHODS An elbow joint trauma-induced HO mouse model was established, where model mice were treated with dichloromethylene-bisphosphonate (Cl2MBP) liposomes or PBS liposomes to deplete macrophages. In addition, boldine was administered to evaluate its therapeutic effect on HO formation. Bone marrow mesenchymal stem cells (BMSCs) were also extracted for in vitro experiments. Quantitative real-time PCR (qRT-PCR) and Western blot were conducted to assess gene and protein expression. In vivo methods included Micro-Computed Tomography (Micro-CT) to assess bone formation, histological staining to evaluate tissue changes, immunohistochemistry (IHC) and immunofluorescence to analyze macrophage, CD73+ and CD105+ cells infiltration. In vitro, BMSCs were identified by flow cytometry and treated with interleukin-10 (IL-10) and/or boldine, and assays such as cell viability (Cell Counting Kit 8 (CCK8)), migration (Transwell), immunofluorescence, ALP staining, and Alizarin Red S staining, were conducted to assess osteogenic differentiation. RESULTS Boldine treatment significantly reduced HO formation, decreased collagen deposition, and inhibited M2 macrophage infiltration (P < 0.05). In vitro, boldine reduced IL-10-induced cell activity, migration, and osteogenic differentiation of BMSCs and inhibited TGF-β and pSmad2/3/Smad2/3 protein (P < 0.05). CONCLUSION Boldine attenuates HO by inhibiting M2 macrophage-mediated MSPC migration and might involve the TGF-β signaling, suggesting its potential as a therapeutic approach for managing HO.
Collapse
Affiliation(s)
- Fengteng Shen
- Department of Orthopedics, Tongxiang First People's Hospital, China
| | - Yansong Chen
- Department of Urology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou City, 311200, China
| | - Zhikun Xu
- Department of Urology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou City, 311200, China
| | - Wei Wang
- Department of Urology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou City, 311200, China
| | - Guofang Chen
- Department of Urology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou City, 311200, China
| | - Fusheng Ye
- Department of Urology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou City, 311200, China.
| |
Collapse
|
4
|
Zhou W, Liu Y, Nie X, Zhu C, Xiong L, Zhou J, Huang W. Peptide-based inflammation-responsive implant coating sequentially regulates bone regeneration to enhance interfacial osseointegration. Nat Commun 2025; 16:3283. [PMID: 40189598 PMCID: PMC11973180 DOI: 10.1038/s41467-025-58444-8] [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: 09/22/2024] [Accepted: 03/20/2025] [Indexed: 04/09/2025] Open
Abstract
Aseptic loosening is the primary cause of bone prosthesis failure, commonly attributed to inadequate osseointegration due to coatings misaligned with bone regeneration. Here, we modify the titanium surface with a mussel-inspired peptide to form a 3,4-dihydroxyphenylalanine (DOPA)-rich coating, then graft N3-K15-PVGLIG-K23 (P1) and N3-Y5-PVGLIG-K23 (P2), which are composed of anti-inflammatory (K23), angiogenic (K15), osteogenic (Y5), and inflammation-responsive (PVGLIG) sequences, onto the surface via click chemistry, forming the DOPA-P1@P2 coating. DOPA-P1@P2 promotes bone regeneration through sequential regulation. In the initial stage, the outermost K23 induces M2 macrophage polarization, establishing a pro-regenerative immune microenvironment. Subsequently, K15 and Y5, exposed by the release of K23, enhance angiogenesis and osteogenesis. In the final stage, DOPA-P1@P2 outperforms the TiO₂ control, showing a 161% increase in maximal push-out force, a 207% increase in bone volume fraction, and a 1409% increase in bone-to-implant contact. These findings show that DOPA-P1@P2 efficiently enhances interfacial osseointegration by sequentially regulating bone regeneration, providing viable insights into coating design.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Liu
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xuan Nie
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, University of Science and Technology of China, Hefei, China
| | - Chen Zhu
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Liming Xiong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jing Zhou
- Department of Urology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Wei Huang
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
5
|
Zhang JW. Role of 1,25-dihydroxyvitamin D 3 in alleviating hepatic steatosis: Targeting M1 macrophage polarization in non-alcoholic fatty liver disease. World J Gastrointest Oncol 2025; 17:102424. [PMID: 40092934 PMCID: PMC11866219 DOI: 10.4251/wjgo.v17.i3.102424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/30/2024] [Accepted: 12/17/2024] [Indexed: 02/14/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), a critical global health concern, continues to challenge medical researchers with limited treatment options. This letter examines on the study by Luo et al, demonstrating that vitamin D 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] improves hepatic steatosis in NAFLD by inhibiting M1 macrophage polarization via the vitamin D receptor-peroxisome proliferator-activated receptor gamma signaling pathway. This letter critically appraises these findings, comparing them to similar studies, and discusses their potential implications for treating NAFLD. Furthermore, we highlight future directions, including dose optimization and mechanistic studies.
Collapse
Affiliation(s)
- Jin-Wei Zhang
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| |
Collapse
|
6
|
Burgan J, Rahmati M, Lee M, Saiz AM. Innate immune response to bone fracture healing. Bone 2025; 190:117327. [PMID: 39522707 DOI: 10.1016/j.bone.2024.117327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
The field of osteoimmunology has primarily focused on fracture healing in isolated musculoskeletal injuries. The innate immune system is the initial response to fracture, with inflammatory macrophages, cytokines, and neutrophils arriving first at the fracture hematoma, followed by an anti-inflammatory phase to begin the process of new bone formation. This review aims to first discuss the current literature and knowledge gaps on the immune responses governing single fracture healing by encompassing the individual role of macrophages, neutrophils, cytokines, mesenchymal stem cells, bone cells, and other immune cells. This paper discusses the interactive effects of these cellular responses underscoring the field of osteoimmunology. The critical role of the metabolic environment in guiding the immune system properties will be highlighted along with some effective therapeutics for fracture healing in the context of osteoimmunology. However, compared to isolated fractures, which frequently heal well, long bone fractures in over 30 % of polytrauma patients exhibit impaired healing. Clinical evidence suggests there may be distinct physiologic and inflammatory pathways altered in polytrauma resulting in nonunion. Nonunion is associated with worse patient outcomes and increased societal healthcare costs. The dysregulated immunomodulatory/inflammatory response seen in polytrauma may lead to this increased nonunion rate. This paper will investigate the differences in immune response between isolated and polytrauma fractures. Finally, future directions for fracture studies are explored with consideration of the emerging roles of newly discovered immune cell functions in fracture healing, the existing challenges and conflicting results in the field, the translational potential of these studies in clinic, and the more complex nature of polytrauma fractures that can alter cell functions in different tissues.
Collapse
Affiliation(s)
- Jane Burgan
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA; Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Maryam Rahmati
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA; Department of Biomaterials, Institute for Clinical Dentistry, University of Oslo, PO Box 1109, Blindern, NO-0317 Oslo, Norway
| | - Mark Lee
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA
| | - Augustine Mark Saiz
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA.
| |
Collapse
|
7
|
Zhao Z, Yuwen W, Duan Z, Zhu C, Fan D. Novel Collagen Analogs with Multicopy Mucin-Type Sequences for Multifunctional Enhancement Properties Using SUMO Fusion Tags. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:22173-22185. [PMID: 39318025 DOI: 10.1021/acs.jafc.4c07179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Multifunctional enhanced collagen materials in green biomanufacturing are highly desired yet challenging due to the poor comprehensive performance caused by the adoption of targeting monofunctional peptides. Herein, novel collagen analog design strategy using multicopy tandem of mucin-type sequence (GAPGAPGSQGAPGLQ) derived from human COL1α1 to construct basic building blocks is reported, in which SUMO tag is added to the N-terminal of the protein as a stabilizing core. In particular, novel collagen analogs (named S1506, S1511, S1523, and S1552) with multicopy mucin-type sequences (repeated 6, 11, 23, and 52 times), which were constructed in Escherichia coli, have distinct orientation preferences of functional enhancement (including cell proliferation, differentiation, migration, antioxidant activity, and anti-inflammatory property) compared to COL1α1 in HaCaT and THP-1 cell experiments due to variant three-dimensional structures (the different-length mucin-type polypeptide chains wind around central SUMO tag). Our findings suggest that the innovative protein design and synthesis approaches employed in the construction of these novel S15 proteins have the potential to advance the development of new types of recombinant collagen analogs.
Collapse
Affiliation(s)
- Zilong Zhao
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710127, China
- Shaanxi Key Laboratory of Biomaterials and Synthetic Biology, Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710127, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710127, China
| | - Weigang Yuwen
- Shaanxi Key Laboratory of Biomaterials and Synthetic Biology, Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710127, China
| | - Zhiguang Duan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710127, China
- Shaanxi Key Laboratory of Biomaterials and Synthetic Biology, Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710127, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710127, China
| | - Chenhui Zhu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710127, China
- Shaanxi Key Laboratory of Biomaterials and Synthetic Biology, Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710127, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710127, China
| | - Daidi Fan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710127, China
- Shaanxi Key Laboratory of Biomaterials and Synthetic Biology, Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710127, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710127, China
| |
Collapse
|
8
|
Stachowicz-Suhs M, Łabędź N, Milczarek M, Kłopotowska D, Filip-Psurska B, Maciejczyk A, Matkowski R, Wietrzyk J. Vitamin D 3 reduces the expression of M1 and M2 macrophage markers in breast cancer patients. Sci Rep 2024; 14:22126. [PMID: 39333342 PMCID: PMC11437092 DOI: 10.1038/s41598-024-73152-x] [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: 04/10/2024] [Accepted: 09/13/2024] [Indexed: 09/29/2024] Open
Abstract
Vitamin D3 (VD) is known for its immunomodulatory and anticancer effects. This study aimed to characterize tumor-associated macrophages (TAMs) in breast cancer (BC) and assess the influence of VD and its active metabolite, calcitriol, on their polarization. TAMs were isolated from BC patients and characterized. Monocytes were differentiated into macrophage classes (M0, M1, M2a, M2c) and treated ex vivo with calcitriol. The expression of VD-related proteins in tumor tissue was correlated with TAMs and monocyte-derived macrophages (MDMs) characteristics. TAM expression of CD200R, CD204, CD80, HLA-DR, and CD44 was negatively correlated with CYP27B1 in selected patient groups. Patients with high CYP27B1 tumor expression showed significantly lower CD200R, CD204, and CD44 expression. In patients with normal VD levels and premenopausal, CD80 expression in M2a and M2c MDMs (control, untreated ex vivo with calcitriol) was negatively correlated with plasma VD. Calcitriol reduced HLA-DR during MDM differentiation in all patients; CD80 decrease significantly except in patients with normal VD levels or metastasis. Calcitriol also decreased CD163 expression. The decrease in both M1 and M2 macrophage markers by calcitriol or their negative correlation with CYP27B1 indicate the modulatory, but rather anticancer activity of VD. The intensity of these effects was the strongest in postmenopausal patients and those without metastases.
Collapse
Affiliation(s)
- Martyna Stachowicz-Suhs
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| | - Natalia Łabędź
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| | - Magdalena Milczarek
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| | - Dagmara Kłopotowska
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| | - Beata Filip-Psurska
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| | - Adam Maciejczyk
- Department of Oncology, Wroclaw Medical University, Wrocław, Poland
- Lower Silesian Oncology, Pulmonology and Hematology Center, Wrocław, Poland
| | - Rafał Matkowski
- Department of Oncology, Wroclaw Medical University, Wrocław, Poland
- Lower Silesian Oncology, Pulmonology and Hematology Center, Wrocław, Poland
| | - Joanna Wietrzyk
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland.
| |
Collapse
|
9
|
Lun H, Li P, Li J, Liu F. The effect of intestinal flora metabolites on macrophage polarization. Heliyon 2024; 10:e35755. [PMID: 39170251 PMCID: PMC11337042 DOI: 10.1016/j.heliyon.2024.e35755] [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: 04/17/2024] [Revised: 07/28/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024] Open
Abstract
Intestinal flora metabolites played a crucial role in immunomodulation by influencing host immune responses through various pathways. Macrophages, as a type of innate immune cell, were essential in chemotaxis, phagocytosis, inflammatory responses, and microbial elimination. Different macrophage phenotypes had distinct biological functions, regulated by diverse factors and mechanisms. Advances in intestinal flora sequencing and metabolomics have enhanced understanding of how intestinal flora metabolites affect macrophage phenotypes and functions. These metabolites had varying effects on macrophage polarization and different mechanisms of influence. This study summarized the impact of gut microbiota metabolites on macrophage phenotype and function, along with the underlying mechanisms associated with different metabolites produced by intestinal flora.
Collapse
Affiliation(s)
- Hengzhong Lun
- Department of Clinical Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| | - Peilong Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Juan Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Fenfen Liu
- Department of Nephrology, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| |
Collapse
|
10
|
Stachowicz-Suhs M, Łabędź N, Anisiewicz A, Banach J, Kłopotowska D, Milczarek M, Piotrowska A, Dzięgiel P, Maciejczyk A, Matkowski R, Wietrzyk J. Calcitriol promotes M2 polarization of tumor-associated macrophages in 4T1 mouse mammary gland cancer via the induction of proinflammatory cytokines. Sci Rep 2024; 14:3778. [PMID: 38355711 PMCID: PMC10866890 DOI: 10.1038/s41598-024-54433-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/13/2024] [Indexed: 02/16/2024] Open
Abstract
Our research found that vitamin D3 (VD3) treatment increased lung metastasis in mice with 4T1 murine breast cancer (BC). This study aims to investigate the impact of VD3 on the activation of tumor-associated macrophages (TAMs) in BC. Mice bearing 4T1, E0771, 67NR BC cells, and healthy mice, were fed diets with varying VD3 contents (100-deficient, 1000-normal, and 5000 IU/kg-elevated). Some mice in the 1000 and 100 IU/kg groups received calcitriol. We studied bone metastasis and characterized TAMs and bone marrow-derived macrophages (BMDMs). 4T1 cells had higher bone metastasis potential in the 5000 IU/kg and calcitriol groups. In the same mice, an elevated tumor osteopontin level and M2 polarization of TAMs (MHCIIlow CD44high phenotype) were observed. Gene expression analysis confirmed M2 polarization of 4T1 (but not 67NR) TAMs and BMDMs, particularly in the 100 IU + cal group (increased Mrc1, Il23, and Il6). This polarization was likely due to COX-2/PGE2 induction in 4T1 calcitriol-treated cells, leading to increased proinflammatory cytokines like IL-6 and IL-23. Future studies will explore COX-2/PGE2 as a primary mediator of calcitriol-stimulated inflammation in the BC microenvironment, especially relevant for BC patients with VD3 deficiency and supplementation.
Collapse
Affiliation(s)
- Martyna Stachowicz-Suhs
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Weigla 12, 53-114, Wroclaw, Poland
| | - Natalia Łabędź
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Weigla 12, 53-114, Wroclaw, Poland
| | - Artur Anisiewicz
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Weigla 12, 53-114, Wroclaw, Poland
| | - Joanna Banach
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Weigla 12, 53-114, Wroclaw, Poland
| | - Dagmara Kłopotowska
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Weigla 12, 53-114, Wroclaw, Poland
| | - Magdalena Milczarek
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Weigla 12, 53-114, Wroclaw, Poland
| | - Aleksandra Piotrowska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Chałubińskiego 6a, 50-368, Wroclaw, Poland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Chałubińskiego 6a, 50-368, Wroclaw, Poland
| | - Adam Maciejczyk
- Department of Oncology, Wroclaw Medical University, Pl. Ludwika Hirszfelda 12, 53-413, Wrocław, Poland
- Lower Silesian Oncology, Pulmonology and Hematology Center, Pl. Ludwika Hirszfelda 12, 53-413, Wrocław, Poland
| | - Rafał Matkowski
- Department of Oncology, Wroclaw Medical University, Pl. Ludwika Hirszfelda 12, 53-413, Wrocław, Poland
- Lower Silesian Oncology, Pulmonology and Hematology Center, Pl. Ludwika Hirszfelda 12, 53-413, Wrocław, Poland
| | - Joanna Wietrzyk
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Weigla 12, 53-114, Wroclaw, Poland.
| |
Collapse
|
11
|
Xiao B, Liu Y, Chandrasiri I, Adjei-Sowah E, Mereness J, Yan M, Benoit DSW. Bone-Targeted Nanoparticle Drug Delivery System-Mediated Macrophage Modulation for Enhanced Fracture Healing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305336. [PMID: 37797180 PMCID: PMC10922143 DOI: 10.1002/smll.202305336] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/03/2023] [Indexed: 10/07/2023]
Abstract
Despite decades of progress, developing minimally invasive bone-specific drug delivery systems (DDS) to improve fracture healing remains a significant clinical challenge. To address this critical therapeutic need, nanoparticle (NP) DDS comprised of poly(styrene-alt-maleic anhydride)-b-poly(styrene) (PSMA-b-PS) functionalized with a peptide that targets tartrate-resistant acid phosphatase (TRAP) and achieves preferential fracture accumulation has been developed. The delivery of AR28, a glycogen synthase kinase-3 beta (GSK3β) inhibitor, via the TRAP binding peptide-NP (TBP-NP) expedites fracture healing. Interestingly, however, NPs are predominantly taken up by fracture-associated macrophages rather than cells typically associated with fracture healing. Therefore, the underlying mechanism of healing via TBP-NP is comprehensively investigated herein. TBP-NPAR28 promotes M2 macrophage polarization and enhances osteogenesis in preosteoblast-macrophage co-cultures in vitro. Longitudinal analysis of TBP-NPAR28 -mediated fracture healing reveals distinct spatial distributions of M2 macrophages, an increased M2/M1 ratio, and upregulation of anti-inflammatory and downregulated pro-inflammatory genes compared to controls. This work demonstrates the underlying therapeutic mechanism of bone-targeted NP DDS, which leverages macrophages as druggable targets and modulates M2 macrophage polarization to enhance fracture healing, highlighting the therapeutic benefit of this approach for fractures and bone-associated diseases.
Collapse
Affiliation(s)
- Baixue Xiao
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14623, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14623, USA
| | - Yuxuan Liu
- Materials Science Program, University of Rochester, Rochester, NY, 14623, USA
| | - Indika Chandrasiri
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14623, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14623, USA
| | - Emmanuela Adjei-Sowah
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14623, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14623, USA
| | - Jared Mereness
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14623, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14623, USA
| | - Ming Yan
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14623, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14623, USA
| | - Danielle S W Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14623, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14623, USA
- Materials Science Program, University of Rochester, Rochester, NY, 14623, USA
- Department of Chemical Engineering, University of Rochester, Rochester, NY, 14623, USA
- Department of Bioengineering, Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, 97403, USA
| |
Collapse
|
12
|
Tan J, Li S, Sun C, Bao G, Liu M, Jing Z, Fu H, Sun Y, Yang Q, Zheng Y, Wang X, Yang H. A Dose-Dependent Spatiotemporal Response of Angiogenesis Elicited by Zn Biodegradation during the Initial Stage of Bone Regeneration. Adv Healthc Mater 2024; 13:e2302305. [PMID: 37843190 DOI: 10.1002/adhm.202302305] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/09/2023] [Indexed: 10/17/2023]
Abstract
Zinc (Zn) plays a crucial role in bone metabolism and imbues biodegradable Zn-based materials with the ability to promote bone regeneration in bone trauma. However, the impact of Zn biodegradation on bone repair, particularly its influence on angiogenesis, remains unexplored. This study reveals that Zn biodegradation induces a consistent dose-dependent spatiotemporal response in angiogenesis,both in vivo and in vitro. In a critical bone defect model, an increase in Zn release intensity from day 3 to 10 post-surgery is observed. By day 10, the CD31-positive area around the Zn implant significantly surpasses that of the Ti implant, indicating enhanced angiogenesis. Furthermore,angiogenesis exhibits a distance-dependent pattern closely mirroring the distribution of Zn signals from the implant. In vitro experiments demonstrate that Zn extraction fosters the proliferation and migration of human umbilical vein endothelial cells and upregulates the key genes associated with tube formation, such as HIF-1α and VEGF-A, peaking at a concentration of 22.5 µM. Additionally, Zn concentrations within the range of 11.25-45 µM promote the polarization of M0-type macrophages toward the M2-type, while inhibiting polarization toward the M1-type. These findings provide essential insights into the biological effects of Zn on bone repair, shedding light on its potential applications.
Collapse
Affiliation(s)
- Junlong Tan
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 37 Xueyuan Rd, Beijing, 100191, China
| | - Shuang Li
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 37 Xueyuan Rd, Beijing, 100191, China
| | - Chaoyang Sun
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 37 Xueyuan Rd, Beijing, 100191, China
| | - Guo Bao
- Department of Reproduction and Physiology, National Research Institute for Family Planning, Beijing, 100081, China
| | - Meijing Liu
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 37 Xueyuan Rd, Beijing, 100191, China
| | - Zehao Jing
- Beijing Key Laboratory of Spinal Disease Research, Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, P. R. China
| | - Hanwei Fu
- School of Materials Science and Engineering, Beihang University, 37 Xueyuan Rd, Beijing, China
| | - Yanhua Sun
- Shandong Provincial Key Laboratory of Microparticles Drug Delivery Technology, Qilu Pharmaceutical Co. Ltd., Jinan, 250100, China
| | - Qingmin Yang
- Shandong Provincial Key Laboratory of Microparticles Drug Delivery Technology, Qilu Pharmaceutical Co. Ltd., Jinan, 250100, China
| | - Yufeng Zheng
- Beijing Advanced Innovation Center for Materials Genome Engineering and School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Xiaogang Wang
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 37 Xueyuan Rd, Beijing, 100191, China
| | - Hongtao Yang
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 37 Xueyuan Rd, Beijing, 100191, China
| |
Collapse
|
13
|
Xiao B, Adjei-Sowah E, Benoit DSW. Integrating osteoimmunology and nanoparticle-based drug delivery systems for enhanced fracture healing. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 56:102727. [PMID: 38056586 PMCID: PMC10872334 DOI: 10.1016/j.nano.2023.102727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/23/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023]
Abstract
Fracture healing is a complex interplay of molecular and cellular mechanisms lasting from days to weeks. The inflammatory phase is the first stage of fracture healing and is critical in setting the stage for successful healing. There has been growing interest in exploring the role of the immune system and novel therapeutic strategies, such as nanoparticle drug delivery systems in enhancing fracture healing. Advancements in nanotechnology have revolutionized drug delivery systems to the extent that they can modulate immune response during fracture healing by leveraging unique physiochemical properties. Therefore, understanding the intricate interactions between nanoparticle-based drug delivery systems and the immune response, specifically macrophages, is essential for therapeutic efficacy. This review provides a comprehensive overview of the relationship between the immune system and nanoparticles during fracture healing. Specifically, we highlight the influence of nanoparticle characteristics, such as size, surface properties, and composition, on macrophage activation, polarization, and subsequent immune responses. IMPACT STATEMENT: This review provides valuable insights into the interplay between fracture healing, the immune system, and nanoparticle-based drug delivery systems. Understanding nanoparticle-macrophage interactions can advance the development of innovative therapeutic approaches to enhance fracture healing, improve patient outcomes, and pave the way for advancements in regenerative medicine.
Collapse
Affiliation(s)
- Baixue Xiao
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14623, USA; Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14623, USA
| | - Emmanuela Adjei-Sowah
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14623, USA; Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14623, USA
| | - Danielle S W Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14623, USA; Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14623, USA; Department of Chemical Engineering, University of Rochester, Rochester, NY 14623, USA; Materials Science Program, University of Rochester, Rochester, NY 14623, USA; Department of Bioengineering, Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR 97403, USA.
| |
Collapse
|
14
|
Wu Y, Huo S, Liu S, Hong Q, Wang Y, Lyu Z. Cu-Sr Bilayer Bioactive Glass Nanoparticles/Polydopamine Functionalized Polyetheretherketone Enhances Osteogenic Activity and Prevents Implant-Associated Infections through Spatiotemporal Immunomodulation. Adv Healthc Mater 2023; 12:e2301772. [PMID: 37723927 DOI: 10.1002/adhm.202301772] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/03/2023] [Indexed: 09/20/2023]
Abstract
Key factors contributing to implantation failures include implant-associated infections (IAIs) and insufficient osseointegration of the implants. Polyetheretherketone (PEEK) is widely used in orthopedics, yet its clinical applications are restricted due to its poor osteogenic and antibacterial properties as well as inadequate immune responses. To overcome these drawbacks, a novel spatiotemporal immunomodulation approach is proposed, chelating Cu-Sr bilayer bioactive glass nanoparticles (CS-BGNs) onto the PEEK surface via polydopamine (PDA). The CS-BGNs possess a bilayer core-shell structure where copper is distributed in the outer layer and strontium is clustered in the inner layer. The results show that CS-BGNs/PDA functionalized PEEK demonstrates a controlled and sequential release of Cu2+ and Sr2+ . In the early stage, Cu2+ from the outer layer releases rapidly, while Sr2+ from the inner layer releases in the late stage. This well-ordered release pattern modulates the phenotypic transition of macrophages, which induces M1 polarization in the early stage and M2 polarization in the late stage. Combined with the direct effects of Cu2+ and Sr2+ , the spatiotemporal immunomodulation not only benefits the early antibacterial and tissue-healing process, but also promotes the long-term process of osseointegration, providing new perspectives on the design of novel immunomodulatory biomaterials.
Collapse
Affiliation(s)
- Yuezhou Wu
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China
| | - Shicheng Huo
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China
| | - Shu Liu
- Department of Orthopedic Surgery, Changhai Hospital, The Second Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Qimin Hong
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China
| | - You Wang
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China
| | - Zhuocheng Lyu
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China
| |
Collapse
|
15
|
Kozutsumi R, Kuroshima S, Al-Omari FA, Hayano H, Nakajima K, Kakehashi H, Sawase T. Depletion of macrophages deteriorates bisphosphonate-related osteonecrosis of the jaw-like lesions in mice. Bone 2023; 177:116899. [PMID: 37708951 DOI: 10.1016/j.bone.2023.116899] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/09/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023]
Abstract
Bisphosphonate-related osteonecrosis of the jaw (BRONJ) is a potentially intractable disease with no definitive pathophysiology and no treatment and prevention strategies. This study aimed to investigate whether time-selective depletion of macrophages worsens BRONJ-like lesions in mice. A murine model of high-prevalence BRONJ-like lesions in combination with zoledronate/chemotherapeutic drug administration and tooth extraction was created according to the methods of our previous studies. Daily intra-oral submucosal administration of clodronate-loaded liposomes, which temporarily depletes systemic macrophages, was performed immediately after tooth extraction. Spleens, femora, tibiae, and maxillae were dissected 2 weeks after extraction to evaluate BRONJ-like lesions and systemic conditions by micro-computed tomography analysis, histomorphometric and immunofluorescent analyses, and serum chemistry with ELISA. Depletion of macrophages significantly decreased the numbers of local and systemic macrophages and osteoclasts on the bone surface, which markedly worsened osseous healing, with increased necrotic bone and empty lacunae in the existing alveolar bone and newly formed bone in the extraction sockets, and soft tissue healing, with decreased collagen production and increased infiltration of polymorphonuclear cells. Interestingly, the depletion of macrophages significantly shifted macrophage polarization to M1 macrophages through an increase in F4/80+CD38+ M1 macrophages and a decrease in F4/80+CD163+ M2 macrophages, with decreases in the total number of F4/80+ macrophages. These data demonstrated that severe inhibition of osteoclasts in bone tissue and polarization shifting of macrophages in soft tissue are essential factors associated with BRONJ.
Collapse
Affiliation(s)
- Ryohei Kozutsumi
- Department of Applied Prosthodontics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan
| | - Shinichiro Kuroshima
- Department of Applied Prosthodontics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan.
| | - Farah A Al-Omari
- Department of Applied Prosthodontics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan
| | - Hiroki Hayano
- Department of Applied Prosthodontics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan
| | - Kazunori Nakajima
- Department of Applied Prosthodontics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan
| | - Hiroe Kakehashi
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnosis and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Takashi Sawase
- Department of Applied Prosthodontics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan
| |
Collapse
|
16
|
Maroquenne M, Bourguignon M, Larochette N, El-Hafci H, Margottin M, Potier E, Logeart-Avramoglou D. The Lower in Vivo Osteogenicity of Adipose Tissue-Derived Stem Cells Correlates with a Higher Innate Immune Response. Stem Cell Rev Rep 2023; 19:2869-2885. [PMID: 37642900 DOI: 10.1007/s12015-023-10614-1] [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] [Accepted: 08/21/2023] [Indexed: 08/31/2023]
Abstract
Adipose tissue-derived mesenchymal stem cells (ATSCs) have been used as an alternative to bone marrow-derived mesenchymal stem cells (BMSCs) for bone tissue engineering applications. The ability of ATSCs to promote new bone formation remains lower than that of BMSCs. This study aimed to investigate the mechanisms underlying osteogenicity differences between human ATSCs and BMSCs in ceramic constructs, focusing on the effects of inflammation on this process. In contrast to ATSC-containing constructs, which did not induce bone formation in an ectopic mouse model, BMSC constructs consistently did so. Gene expression analysis revealed that human BMSCs, concomitantly with host murine progenitors, differentiated into the osteogenic lineage early post-implantation. In contrast, ATSCs differentiated later, when few implanted viable cells remained post-implantation, while the host murine cells did not differentiate. Comparison of the inflammatory profile in the cell constructs indicated concomitant upregulation of some human and murine inflammatory genes in the ATSC-constructs compared to the BMSC-constructs during the first-week post-implantation. The high level of chemokine production by the ATSCs was confirmed at the gene and protein levels before implantation. The immune cell recruitment within the constructs was then explored post-implantation. Higher numbers of TRAP-/ MRC1 (CD206) + multinucleated giant cells, NOS2 + M1, and ARG1 + M2 macrophages were present in the ATSC constructs than in the BMSC constructs. These results proved that ATSCs are a transient source of inflammatory cytokines promoting a transient immune response post-implantation; this milieu correlates with impaired osteogenic differentiation of both the implanted ATSCs and the host osteoprogenitor cells.
Collapse
Affiliation(s)
- Manon Maroquenne
- Université Paris Cité, CNRS, INSERM, ENVA, Paris, B3OA, F-75010, France
| | | | | | - Hanane El-Hafci
- Université Paris Cité, CNRS, INSERM, ENVA, Paris, B3OA, F-75010, France
| | - Morgane Margottin
- Université Paris Cité, CNRS, INSERM, ENVA, Paris, B3OA, F-75010, France
| | - Esther Potier
- Université Paris Cité, CNRS, INSERM, ENVA, Paris, B3OA, F-75010, France
| | - Delphine Logeart-Avramoglou
- Université Paris Cité, CNRS, INSERM, ENVA, Paris, B3OA, F-75010, France.
- Laboratoire de Biologie, Bioingénierie et Bioimagerie Ostéo-articulaires, Université Paris Cité, 10 Avenue de Verdun, Paris, F-75010, France.
| |
Collapse
|
17
|
Fan S, Sun X, Su C, Xue Y, Song X, Deng R. Macrophages-bone marrow mesenchymal stem cells crosstalk in bone healing. Front Cell Dev Biol 2023; 11:1193765. [PMID: 37427382 PMCID: PMC10327485 DOI: 10.3389/fcell.2023.1193765] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/14/2023] [Indexed: 07/11/2023] Open
Abstract
Bone healing is associated with many orthopedic conditions, including fractures and osteonecrosis, arthritis, metabolic bone disease, tumors and periprosthetic particle-associated osteolysis. How to effectively promote bone healing has become a keen topic for researchers. The role of macrophages and bone marrow mesenchymal stem cells (BMSCs) in bone healing has gradually come to light with the development of the concept of osteoimmunity. Their interaction regulates the balance between inflammation and regeneration, and when the inflammatory response is over-excited, attenuated, or disturbed, it results in the failure of bone healing. Therefore, an in-depth understanding of the function of macrophages and bone marrow mesenchymal stem cells in bone regeneration and the relationship between the two could provide new directions to promote bone healing. This paper reviews the role of macrophages and bone marrow mesenchymal stem cells in bone healing and the mechanism and significance of their interaction. Several new therapeutic ideas for regulating the inflammatory response in bone healing by targeting macrophages and bone marrow mesenchymal stem cells crosstalk are also discussed.
Collapse
Affiliation(s)
- Siyu Fan
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Xin Sun
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Chuanchao Su
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Yiwen Xue
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Xiao Song
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Runzhi Deng
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
18
|
Xiao B, Liu Y, Chandrasiri I, Overby C, Benoit DSW. Impact of Nanoparticle Physicochemical Properties on Protein Corona and Macrophage Polarization. ACS APPLIED MATERIALS & INTERFACES 2023:10.1021/acsami.2c22471. [PMID: 36916683 PMCID: PMC11955209 DOI: 10.1021/acsami.2c22471] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Macrophages, the major component of the mononuclear phagocyte system, uptake and clear systemically administered nanoparticles (NPs). Therefore, leveraging macrophages as a druggable target may be advantageous to enhance NP-mediated drug delivery. Despite many studies focused on NP-cell interactions, NP-mediated macrophage polarization mechanisms are still poorly understood. This work aimed to explore the effect of NP physicochemical parameters (size and charge) on macrophage polarization. Upon exposure to biological fluids, proteins rapidly adsorb to NPs and form protein coronas. To this end, we hypothesized that NP protein coronas govern NP-macrophage interactions, uptake, and subsequent macrophage polarization. To test this hypothesis, model polystyrene NPs with various charges and sizes, as well as NPs relevant to drug delivery, were utilized. Data suggest that cationic NPs potentiate both M1 and M2 macrophage markers, while anionic NPs promote M1-to-M2 polarization. Additionally, anionic polystyrene nanoparticles (APNs) of 50 nm exhibit the greatest influence on M2 polarization. Proteomics was pursued to further understand the effect of NPs physicochemical parameters on protein corona, which revealed unique protein patterns based on NP charge and size. Several proteins impacting M1 and M2 macrophage polarization were identified within cationic polystyrene nanoparticles (CPNs) corona, while APNs corona included fewer M1 but more M2-promoting proteins. Nevertheless, size impacts protein corona abundance but not identities. Altogether, protein corona identities varied based on NP surface charge and correlated to dramatic differences in macrophage polarization. In contrast, NP size differentially impacts macrophage polarization, which is dominated by NP uptake level rather than protein corona. In this work, specific corona proteins were identified as a function of NP physicochemical properties. These proteins are correlated to specific macrophage polarization programs and may provide design principles for developing macrophage-mediated NP drug delivery systems.
Collapse
Affiliation(s)
- Baixue Xiao
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14623, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14623, USA
| | - Yuxuan Liu
- Materials Science Program, University of Rochester, Rochester, NY 14623, USA
| | - Indika Chandrasiri
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14623, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14623, USA
| | - Clyde Overby
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14623, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14623, USA
| | - Danielle S. W. Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14623, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14623, USA
- Department of Chemical Engineering, University of Rochester, Rochester, NY 14623, USA
- Materials Science Program, University of Rochester, Rochester, NY 14623, USA
- Knight Campus, Department of Bioengineering, University of Oregon, Eugene, OR 97403, USA
| |
Collapse
|
19
|
Orvalho JM, Fernandes JCH, Moraes Castilho R, Fernandes GVO. The Macrophage’s Role on Bone Remodeling and Osteogenesis: a Systematic Review. Clin Rev Bone Miner Metab 2023. [DOI: 10.1007/s12018-023-09286-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
|
20
|
McColl LF, Chen X, Solga MD, Schlegel K, Haughey SP, Lobo PI, Fread K, Zunder E, Cha R, Park S, Christophel JJ, Cui Q, Dighe AS. BMP-6 promotes type 2 immune response during enhancement of rat mandibular bone defect healing. Front Immunol 2023; 14:1064238. [PMID: 36845161 PMCID: PMC9950738 DOI: 10.3389/fimmu.2023.1064238] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 01/31/2023] [Indexed: 02/12/2023] Open
Abstract
Introduction Bone morphogenetic proteins (BMPs) are used as key therapeutic agents for the treatment of difficult fractures. While their effects on osteoprogenitors are known, little is known about their effects on the immune system. Methods We used permutations of BMP-6 (B), vascular endothelial growth factor (V), and Hedgehog signaling pathway activator smoothened agonist (S), to treat a rat mandibular defect and investigated healing outcomes at week 8, in correlation with the cellular landscape of the immune cells in the fracture callus at week 2. Results Maximum recruitment of immune cells to the fracture callus is known to occur at week 2. While the control, S, V, and VS groups remained as nonunions at week 8; all BMP-6 containing groups - B, BV, BS and BVS, showed near-complete to complete healing. This healing pattern was strongly associated with significantly higher ratios of CD4 T (CD45+CD3+CD4+) to putative CD8 T cells (CD45+CD3+CD4-), in groups treated with any permutation of BMP-6. Although, the numbers of putative M1 macrophages (CD45+CD3-CD11b/c+CD38high) were significantly lower in BMP-6 containing groups in comparison with S and VS groups, percentages of putative - Th1 cells or M1 macrophages (CD45+CD4+IFN-γ+) and putative - NK, NKT or cytotoxic CD8T cells (CD45+CD4-IFN-γ+) were similar in control and all treatment groups. Further interrogation revealed that the BMP-6 treatment promoted type 2 immune response by significantly increasing the numbers of CD45+CD3-CD11b/c+CD38low putative M2 macrophages, putative - Th2 cells or M2 macrophages (CD45+CD4+IL-4+) cells and putative - mast cells, eosinophils or basophils (CD45+CD4-IL-4+ cells). CD45- non-haematopoietic fractions of cells which encompass all known osteoprogenitor stem cells populations, were similar in control and treatment groups. Discussion This study uncovers previously unidentified regulatory functions of BMP-6 and shows that BMP-6 enhances fracture healing by not only acting on osteoprogenitor stem cells but also by promoting type 2 immune response.
Collapse
Affiliation(s)
- Logan F. McColl
- Department of Orthopedic Surgery, University of Virginia Health System, Charlottesville, VA, United States
| | - Xizhao Chen
- Department of Orthopedic Surgery, University of Virginia Health System, Charlottesville, VA, United States
| | - Michael D. Solga
- Flow Cytometry Core Facility, University of Virginia, Charlottesville, VA, United States
| | - Kailo Schlegel
- Department of Nephrology, University of Virginia Health System, Charlottesville, VA, United States
| | - Sean P. Haughey
- Department of Orthopedic Surgery, University of Virginia Health System, Charlottesville, VA, United States
| | - Peter I. Lobo
- Department of Nephrology, University of Virginia Health System, Charlottesville, VA, United States
| | - Kristen Fread
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Eli Zunder
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Ryan Cha
- Department of Orthopedic Surgery, University of Virginia Health System, Charlottesville, VA, United States
| | - Stephen Park
- Department of Otolaryngology–Head and Neck Surgery, University of Virginia Health System, Charlottesville, VA, United States
| | - J. Jared Christophel
- Department of Otolaryngology–Head and Neck Surgery, University of Virginia Health System, Charlottesville, VA, United States
| | - Quanjun Cui
- Department of Orthopedic Surgery, University of Virginia Health System, Charlottesville, VA, United States
| | - Abhijit S. Dighe
- Department of Orthopedic Surgery, University of Virginia Health System, Charlottesville, VA, United States,*Correspondence: Abhijit S. Dighe,
| |
Collapse
|
21
|
Frade BB, Dias RB, Gemini Piperni S, Bonfim DC. The role of macrophages in fracture healing: a narrative review of the recent updates and therapeutic perspectives. Stem Cell Investig 2023; 10:4. [PMID: 36817259 PMCID: PMC9936163 DOI: 10.21037/sci-2022-038] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 01/10/2023] [Indexed: 02/10/2023]
Abstract
Objective This review addresses the latest advances in research on the role of macrophages in fracture healing, exploring their relationship with failures in bone consolidation and the perspectives for the development of advanced and innovative therapies to promote bone regeneration. Background The bone can fully restore its form and function after a fracture. However, the regenerative process of fracture healing is complex and is influenced by several factors, including macrophage activity. These cells have been found in the fracture site at all stages of bone regeneration, and their general depletion or the knockdown of receptors that mediate their differentiation, polarization, and/or function result in impaired fracture healing. Methods The literature search was carried out in the PubMed database, using combinations of the keywords "macrophage", "fracture healing, "bone regeneration", and "bone repair". Articles published within the last years (2017-2022) reporting evidence from in vivo long bone fracture healing experiments were included. Conclusions Studies published in the last five years on the role of macrophages in fracture healing strengthened the idea that what appears to be essential when it comes to a successful consolidation is the right balance between the M1/M2 populations, which have different but complementary roles in the process. These findings opened promising new avenues for the development of several macrophage-targeted therapies, including the administration of molecules and/or biomaterials intended to regulate macrophage differentiation and polarization, the local transplantation of macrophage precursors, and the use of exosomes to deliver signaling molecules that influence macrophage activities. However, more research is still warranted to better understand the diversity of macrophage phenotypes and their specific roles in each step of fracture healing and to decipher the key molecular mechanisms involved in the in vivo crosstalk between macrophages and other microenvironmental cell types, such as endothelial and skeletal stem/progenitor cells.
Collapse
Affiliation(s)
- Bianca Braga Frade
- Laboratory of Stem Cells and Bone Regeneration, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil;,Postgraduation Program in Biological Sciences-Biophysics, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rhayra Braga Dias
- Laboratory of Stem Cells and Bone Regeneration, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil;,Postgraduation Program in Morphological Sciences, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sara Gemini Piperni
- Laboratory of Biotechnology, Bioengineering and Nanostructured Biomaterials, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danielle Cabral Bonfim
- Laboratory of Stem Cells and Bone Regeneration, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
22
|
Zhang D, Dang Y, Deng R, Ma Y, Wang J, Ao J, Wang X. Research Progress of Macrophages in Bone Regeneration. J Tissue Eng Regen Med 2023; 2023:1512966. [PMID: 40226416 PMCID: PMC11919137 DOI: 10.1155/2023/1512966] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/07/2022] [Accepted: 12/17/2022] [Indexed: 04/15/2025]
Abstract
Bone tissue regeneration plays an increasingly important role in contemporary clinical treatment. The reconstruction of bone defects remains a huge challenge for clinicians. Bone regeneration is regulated by the immune system, in which inflammation is an important regulating factor in bone formation and remodeling. As the main cells involved in inflammation, macrophages play a key role in osteogenesis by polarizing into different phenotypes during different stages of bone regeneration. Considering this, this review mainly summarizes the function of macrophage in bone regeneration based on mesenchymal stem cells (MSCs), osteoblasts, osteoclasts, and vascular cells. In conclusion, anti-inflammatory macrophages (M2) have a greater potentiality to promote bone regeneration than M0 and classically activated proinflammatory macrophages (M1). In the fracture and bone defect models, tissue engineering materials can induce the transition from M1 to M2, alter the bone microenvironment, and promote bone regeneration through interactions with bone-related cells and blood vessels. The review provides a further understanding of macrophage polarization behavior in the evolving field of bone immunology.
Collapse
Affiliation(s)
- Dingmei Zhang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Yi Dang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Renli Deng
- Nurse Department, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Yaping Ma
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Jing Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Jun Ao
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Xin Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| |
Collapse
|
23
|
Liu F, Sun T, An Y, Ming L, Li Y, Zhou Z, Shang F. The potential therapeutic role of extracellular vesicles in critical-size bone defects: Spring of cell-free regenerative medicine is coming. Front Bioeng Biotechnol 2023; 11:1050916. [PMID: 36733961 PMCID: PMC9887316 DOI: 10.3389/fbioe.2023.1050916] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/02/2023] [Indexed: 01/19/2023] Open
Abstract
In recent years, the incidence of critical-size bone defects has significantly increased. Critical-size bone defects seriously affect patients' motor functions and quality of life and increase the need for additional clinical treatments. Bone tissue engineering (BTE) has made great progress in repairing critical-size bone defects. As one of the main components of bone tissue engineering, stem cell-based therapy is considered a potential effective strategy to regenerate bone tissues. However, there are some disadvantages including phenotypic changes, immune rejection, potential tumorigenicity, low homing efficiency and cell survival rate that restrict its wider clinical applications. Evidence has shown that the positive biological effects of stem cells on tissue repair are largely mediated through paracrine action by nanostructured extracellular vesicles (EVs), which may overcome the limitations of traditional stem cell-based treatments. In addition to stem cell-derived extracellular vesicles, the potential therapeutic roles of nonstem cell-derived extracellular vesicles in critical-size bone defect repair have also attracted attention from scholars in recent years. Currently, the development of extracellular vesicles-mediated cell-free regenerative medicine is still in the preliminary stage, and the specific mechanisms remain elusive. Herein, the authors first review the research progress and possible mechanisms of extracellular vesicles combined with bone tissue engineering scaffolds to promote bone regeneration via bioactive molecules. Engineering modified extracellular vesicles is an emerging component of bone tissue engineering and its main progression and clinical applications will be discussed. Finally, future perspectives and challenges of developing extracellular vesicle-based regenerative medicine will be given. This review may provide a theoretical basis for the future development of extracellular vesicle-based biomedicine and provide clinical references for promoting the repair of critical-size bone defects.
Collapse
Affiliation(s)
- Fen Liu
- Department of Periodontology, Shenzhen Stomatological Hospital (Pingshan), Southern Medical University, Shenzhen, Guangdong, China
| | - Tianyu Sun
- Department of Periodontology, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying An
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture and Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Leiguo Ming
- Department of Research and Development, Shaanxi Zhonghong Institute of Regenerative Medicine, Xi’an, Shaanxi, China
| | - Yinghui Li
- Department of Orthodontics, Stomatological Hospital, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhifei Zhou
- Department of Stomatology, General Hospital of Tibetan Military Command, Lhasa, Tibet, China,*Correspondence: Fengqing Shang, ; Zhifei Zhou,
| | - Fengqing Shang
- Department of Stomatology, Air Force Medical Center, Fourth Military Medical University, Beijing, China,*Correspondence: Fengqing Shang, ; Zhifei Zhou,
| |
Collapse
|
24
|
Khosrowpour Z, Hashemi SM, Mohammadi-Yeganeh S, Moghtadaei M, Brouki Milan P, Moroni L, Kundu SC, Gholipourmalekabadi M. Coculture of adipose-derived mesenchymal stem cells/macrophages on decellularized placental sponge promotes differentiation into the osteogenic lineage. Artif Organs 2023; 47:47-61. [PMID: 36029128 DOI: 10.1111/aor.14394] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/07/2022] [Accepted: 02/10/2022] [Indexed: 01/04/2023]
Abstract
BACKGROUND Several factors like three-dimensional microstructure, growth factors, cytokines, cell-cell communication, and coculture with functional cells can affect the stem cells behavior and differentiation. The purpose of this study was to investigate the potential of decellularized placental sponge as adipose-derived mesenchymal stem cells (AD-MSCs) and macrophage coculture systems, and guiding the osteogenic differentiation of stem cells. METHODS The decellularized placental sponge (DPS) was fabricated, and its mechanical characteristics were evaluated using degradation assay, swelling rate, and pore size determination. Its structure was also investigated using hematoxylin and eosin staining and scanning electron microscopy. Mouse peritoneal macrophages and AD-MSCs were isolated and characterized. The differentiation potential of AD-MSCs co-cultured with macrophages was evaluated by RT-qPCR of osteogenic genes on the surface of DPS. The in vivo biocompatibility of DPS was determined by subcutaneous implantation of scaffold and histological evaluations of the implanted site. RESULTS The DPS had 67% porosity with an average pore size of 238 μm. The in vitro degradation assay showed around 25% weight loss during 30 days in PBS. The swelling rate was around 50% during 72 h. The coculture of AD-MSCs/macrophages on the DPS showed a significant upregulation of four differentiation osteogenic lineage genes in AD-MSCs on days 14 and 21 and a significantly higher mineralization rate than the groups without DPS. Subcutaneous implantation of DPS showed in vivo biocompatibility of scaffold during 28 days follow-up. CONCLUSIONS Our findings suggest the decellularized placental sponge as an excellent bone substitute providing a naturally derived matrix substrate with biostructure close to the natural bone that guided differentiation of stem cells toward bone cells and a promising coculture substrate for crosstalk of macrophage and mesenchymal stem cells in vitro.
Collapse
Affiliation(s)
- Zahra Khosrowpour
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Mohammadi-Yeganeh
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Moghtadaei
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.,Orthopaedic Department, Hazrat-Rasul Hospital, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Peiman Brouki Milan
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Lorenzo Moroni
- Complex Tissue Regeneration Department, Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht, The Netherlands
| | - Subhas C Kundu
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradable and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimaraes, Portugal
| | - Mazaher Gholipourmalekabadi
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.,Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Hu Y, Huang J, Chen C, Wang Y, Hao Z, Chen T, Wang J, Li J. Strategies of Macrophages to Maintain Bone Homeostasis and Promote Bone Repair: A Narrative Review. J Funct Biomater 2022; 14:18. [PMID: 36662065 PMCID: PMC9864083 DOI: 10.3390/jfb14010018] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/17/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
Bone homeostasis (a healthy bone mass) is regulated by maintaining a delicate balance between bone resorption and bone formation. The regulation of physiological bone remodeling by a complex system that involves multiple cells in the skeleton is closely related to bone homeostasis. Loss of bone mass or repair of bone is always accompanied by changes in bone homeostasis. However, due to the complexity of bone homeostasis, we are currently unable to identify all the mechanisms that affect bone homeostasis. To date, bone macrophages have been considered a third cellular component in addition to osteogenic spectrum cells and osteoclasts. As confirmed by co-culture models or in vivo experiments, polarized or unpolarized macrophages interact with multiple components within the bone to ensure bone homeostasis. Different macrophage phenotypes are prone to resorption and formation of bone differently. This review comprehensively summarizes the mechanisms by which macrophages regulate bone homeostasis and concludes that macrophages can control bone homeostasis from osteoclasts, mesenchymal cells, osteoblasts, osteocytes, and the blood/vasculature system. The elaboration of these mechanisms in this narrative review facilitates the development of macrophage-based strategies for the treatment of bone metabolic diseases and bone defects.
Collapse
Affiliation(s)
- Yingkun Hu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Jinghuan Huang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200000, China
| | - Chunying Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Yi Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Zhuowen Hao
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Tianhong Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Junwu Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Jingfeng Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| |
Collapse
|
26
|
Wang YH, Zhao CZ, Wang RY, Du QX, Liu JY, Pan J. The crosstalk between macrophages and bone marrow mesenchymal stem cells in bone healing. Stem Cell Res Ther 2022; 13:511. [PMID: 36333820 PMCID: PMC9636722 DOI: 10.1186/s13287-022-03199-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
Bone injury plagues millions of patients worldwide every year, and it demands a heavy portion of expense from the public medical insurance system. At present, orthopedists think that autologous bone transplantation is the gold standard for treating large-scale bone defects. However, this method has significant limitations, which means that parts of patients cannot obtain a satisfactory prognosis. Therefore, a basic study on new therapeutic methods is urgently needed. The in-depth research on crosstalk between macrophages (Mϕs) and bone marrow mesenchymal stem cells (BMSCs) suggests that there is a close relationship between inflammation and regeneration. The in-depth understanding of the crosstalk between Mϕs and BMSCs is helpful to amplify the efficacy of stem cell-based treatment for bone injury. Only in the suitable inflammatory microenvironment can the damaged tissues containing stem cells obtain satisfactory healing outcomes. The excessive tissue inflammation and lack of stem cells make the transplantation of biomaterials necessary. We can expect that the crosstalk between Mϕs and BMSCs and biomaterials will become the mainstream to explore new methods for bone injury in the future. This review mainly summarizes the research on the crosstalk between Mϕs and BMSCs and also briefly describes the effects of biomaterials and aging on cell transplantation therapy.
Collapse
Affiliation(s)
- Yu-Hao Wang
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, #14 Third Section, Renmin Road South, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581Chengdu Advanced Medical Science Center, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan Province People’s Republic of China
| | - Cheng-Zhi Zhao
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, #14 Third Section, Renmin Road South, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581Chengdu Advanced Medical Science Center, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan Province People’s Republic of China
| | - Ren-Yi Wang
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, #14 Third Section, Renmin Road South, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581Chengdu Advanced Medical Science Center, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan Province People’s Republic of China
| | - Qian-Xin Du
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, #14 Third Section, Renmin Road South, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581Chengdu Advanced Medical Science Center, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan Province People’s Republic of China
| | - Ji-Yuan Liu
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, #14 Third Section, Renmin Road South, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 People’s Republic of China
| | - Jian Pan
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, #14 Third Section, Renmin Road South, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581Chengdu Advanced Medical Science Center, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan Province People’s Republic of China
| |
Collapse
|
27
|
Effects of Exercise or Mechanical Stimulation on Bone Development and Bone Repair. Stem Cells Int 2022; 2022:5372229. [PMID: 36213684 PMCID: PMC9534715 DOI: 10.1155/2022/5372229] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/07/2022] [Indexed: 11/25/2022] Open
Abstract
The development and regeneration of the bone are tightly regulated by mechanical cues. Multiple cell types, including osteoblasts, osteocytes, osteoclasts, mesenchymal stem cells (MSCs), and recently found skeletal stem cells (SSCs), are responsible for efficient bone development and injury repair. The immune cells in the environment interact with bone cells to maintain homeostasis and facilitate bone regeneration. Investigation of the mechanism by which these cells sense and respond to mechanical signals in bone is fundamental for optimal clinical intervention in bone injury healing. We discuss the effects of exercise programs on fracture healing in animal models and human patients, which encouragingly suggest that carefully designed exercise prescriptions can improve the result of fracture healing during the remodeling phase. However, additional clinical tracing and date accumulation are still required for the pervasive application of exercise prescriptions to improve fracture healing.
Collapse
|
28
|
Chow SKH, Wong CHW, Cui C, Li MMC, Wong RMY, Cheung WH. Modulating macrophage polarization for the enhancement of fracture healing, a systematic review. J Orthop Translat 2022; 36:83-90. [PMID: 35979176 PMCID: PMC9364046 DOI: 10.1016/j.jot.2022.05.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 12/04/2022] Open
Abstract
Background All fracture repairs start with the innate immune system with the inflammatory response known as the inflammatory stage guided and driven by the secretion of chemokine by the ruptured tissue, followed by the sequential recruitment of neutrophils, monocytes and macrophages. These innate immune cells would infiltrate the fracture site and secrete inflammatory cytokines to stimulate recruitment of more immune cells to arrive at the fracture site coordinating subsequent stages of the repair process. In which, subsidence of pro-inflammatory M1 macrophage and transformation to anti-inflammatory M2 macrophages promotes osteogenesis that marks the start of the anabolic endochondral stage. Methods Literature search was performed on Pubmed, Embase, and Web of Science databases (last accessed 15th April 2021) using “macrophage AND fracture”. Review was performed in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guideline. Results Eleven pre-clinical animal studies out of 429 articles were included in this systematic review according to our inclusion and exclusion criteria. All of which investigated interventions targeting to modulate the acute inflammatory response and macrophage polarization as evident by various markers in association with fracture healing outcomes. Conclusion This systematic review summarizes attempts to modulate the innate immune response with focuses on promoting macrophage polarization from M1 to M2 phenotype targeting the enhancement of fracture injury repair. Methods used to achieve the goal may include applications of damage-associated molecular pattern (DAMP), pathogen-associated molecular pattern (PAMP) or mechanical stimulation that hold high translational potentials for clinical application in the near future.
Collapse
Affiliation(s)
- Simon Kwoon-Ho Chow
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Carissa Hing-Wai Wong
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Can Cui
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Michelle Meng-Chen Li
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ronald Man Yeung Wong
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wing-Hoi Cheung
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
29
|
Wang D, Cao H, Hua W, Gao L, Yuan Y, Zhou X, Zeng Z. Mesenchymal Stem Cell-Derived Extracellular Vesicles for Bone Defect Repair. MEMBRANES 2022; 12:716. [PMID: 35877919 PMCID: PMC9315966 DOI: 10.3390/membranes12070716] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/17/2022] [Accepted: 07/18/2022] [Indexed: 12/12/2022]
Abstract
The repair of critical bone defects is a hotspot of orthopedic research. With the development of bone tissue engineering (BTE), there is increasing evidence showing that the combined application of extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) (MSC-EVs), especially exosomes, with hydrogels, scaffolds, and other bioactive materials has made great progress, exhibiting a good potential for bone regeneration. Recent studies have found that miRNAs, proteins, and other cargo loaded in EVs are key factors in promoting osteogenesis and angiogenesis. In BTE, the expression profile of the intrinsic cargo of EVs can be changed by modifying the gene expression of MSCs to obtain EVs with enhanced osteogenic activity and ultimately enhance the osteoinductive ability of bone graft materials. However, the current research on MSC-EVs for repairing bone defects is still in its infancy, and the underlying mechanism remains unclear. Therefore, in this review, the effect of bioactive materials such as hydrogels and scaffolds combined with MSC-EVs in repairing bone defects is summarized, and the mechanism of MSC-EVs promoting bone defect repair by delivering active molecules such as internal miRNAs is further elucidated, which provides a theoretical basis and reference for the clinical application of MSC-EVs in repairing bone defects.
Collapse
Affiliation(s)
- Dongxue Wang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China; (D.W.); (W.H.); (L.G.)
| | - Hong Cao
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China; (H.C.); (Y.Y.)
| | - Weizhong Hua
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China; (D.W.); (W.H.); (L.G.)
| | - Lu Gao
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China; (D.W.); (W.H.); (L.G.)
| | - Yu Yuan
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China; (H.C.); (Y.Y.)
| | - Xuchang Zhou
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China; (D.W.); (W.H.); (L.G.)
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China; (H.C.); (Y.Y.)
| | - Zhipeng Zeng
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China; (D.W.); (W.H.); (L.G.)
| |
Collapse
|
30
|
Du C, Xiao P, Gao S, Chen S, Chen B, Huang W, Zhao C. High Fluoride Ingestion Impairs Bone Fracture Healing by Attenuating M2 Macrophage Differentiation. Front Bioeng Biotechnol 2022; 10:791433. [PMID: 35669059 PMCID: PMC9164140 DOI: 10.3389/fbioe.2022.791433] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 04/20/2022] [Indexed: 11/23/2022] Open
Abstract
Fluorosis is still endemic in at least 25 countries around the world. In this study, we investigated the effect of high fluoride intake on fracture healing. Our in vitro experiments found that fluoride inhibited the osteogenic and angiogenic differentiation of MSCs in a dose-dependent manner. By constructing a bone fracture model, we found that high fluoride intake influences bone fracture by attenuating endochondral ossification and angiogenesis. In the mechanism, we clarified that high fluoride inhibits M2 differentiation rather than M1 differentiation in the fracture area, which may contribute to the delayed healing of the fracture. These findings provide an essential reference for the clinical treatment of bone fracture patients with a history of high fluoride intake or skeletal fluorosis patients.
Collapse
Affiliation(s)
| | | | | | | | | | - Wei Huang
- *Correspondence: Wei Huang, ; Chen Zhao,
| | - Chen Zhao
- *Correspondence: Wei Huang, ; Chen Zhao,
| |
Collapse
|
31
|
Li R, Li D, Wang H, Chen K, Wang S, Xu J, Ji P. Exosomes from adipose-derived stem cells regulate M1/M2 macrophage phenotypic polarization to promote bone healing via miR-451a/MIF. Stem Cell Res Ther 2022; 13:149. [PMID: 35395782 PMCID: PMC8994256 DOI: 10.1186/s13287-022-02823-1] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/11/2021] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES Bone defects caused by diseases and trauma are usually accompanied by inflammation, and the implantation of biomaterials as a common repair method has also been found to cause inflammatory reactions, which affect bone metabolism and new bone formation. This study investigated whether exosomes from adipose-derived stem cells (ADSC-Exos) plays an immunomodulatory role in traumatic bone defects and elucidated the underlying mechanisms. METHODS ADSC-Exos were loaded by a biomaterial named gelatine nanoparticles (GNPs), physical and chemical properties were analysed by zeta potential, surface topography and rheology. A rat model of skull defect was used for our in vivo studies, and micro-CT and histological staining were used to analyse histological changes in the bone defect area. RT-qPCR and western blotting were performed to verify that ADSC-Exos could regulate M1/M2 macrophage polarization. MicroRNA (miRNA) array analysis was conducted to determine the miRNA expression profiles of ADSC-Exos. After macrophages were treated with a miR-451a mimic, miR-451a inhibitor and ISO-1, the relative expression of genes and proteins was measured by RT-qPCR and western blotting. RESULTS In vivo, micro-CT and histological staining showed that exosome-loaded GNPs (GNP-Exos) hydrogel, with good biocompatibility and strong mechanical adaptability, exhibited immunomodulatory effect mainly by regulating macrophage immunity and promoting bone tissue healing. Immunofluorescence further indicated that ADSC-Exos reduced M1 marker (iNOS) expression and increased M2 marker (CD206) expression. Moreover, in vitro studies, western blotting and RT-qPCR showed that ADSC-Exos inhibited M1 macrophage marker expression and upregulated M2 macrophage marker expression. MiR-451a was enriched in ADSC-Exos and targeted macrophage migration inhibitory factor (MIF). Macrophages treated with the miR-451a mimic showed lower expression of M1 markers. In contrast, miR-451a inhibitor treatment upregulated the expression of M1 markers and downregulated the expression of M2 markers, while ISO-1 (a MIF inhibitor) treatment upregulated miR-451a expression and downregulated M1 macrophage marker expression. CONCLUSION GNP-Exos can effectively regulate bone immune metabolism and further promote bone healing partly through immune regulation of miR-451a, which may provide a therapeutic direction for bone repair.
Collapse
Affiliation(s)
- Rui Li
- Department of Pediatric Dentistry, The College of Stomatology, Chongqing Medical University, No. 426, North Songshi Road, Yubei District, Chongqing, 401147, People's Republic of China
- Department of Oral and Maxillofacial Surgery, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, People's Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China
| | - Dize Li
- Department of Pediatric Dentistry, The College of Stomatology, Chongqing Medical University, No. 426, North Songshi Road, Yubei District, Chongqing, 401147, People's Republic of China
- Department of Oral and Maxillofacial Surgery, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, People's Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China
| | - Huanan Wang
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, Dalian, 116023, People's Republic of China
| | - Kaiwen Chen
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, Dalian, 116023, People's Republic of China
| | - Si Wang
- Department of Pediatric Dentistry, The College of Stomatology, Chongqing Medical University, No. 426, North Songshi Road, Yubei District, Chongqing, 401147, People's Republic of China.
- Department of Oral and Maxillofacial Surgery, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, People's Republic of China.
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China.
| | - Jie Xu
- Department of Pediatric Dentistry, The College of Stomatology, Chongqing Medical University, No. 426, North Songshi Road, Yubei District, Chongqing, 401147, People's Republic of China.
- Department of Oral and Maxillofacial Surgery, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, People's Republic of China.
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China.
| | - Ping Ji
- Department of Pediatric Dentistry, The College of Stomatology, Chongqing Medical University, No. 426, North Songshi Road, Yubei District, Chongqing, 401147, People's Republic of China.
- Department of Oral and Maxillofacial Surgery, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, People's Republic of China.
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China.
| |
Collapse
|
32
|
Li X, Xue S, Zhan Q, Sun X, Chen N, Li S, Zhao J, Hou X, Yuan X. Sequential Delivery of Different MicroRNA Nanocarriers Facilitates the M1-to-M2 Transition of Macrophages. ACS OMEGA 2022; 7:8174-8183. [PMID: 35284756 PMCID: PMC8908531 DOI: 10.1021/acsomega.2c00297] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 02/14/2022] [Indexed: 06/01/2023]
Abstract
The early-stage repair of bone injuries dominated by the inflammatory phase is significant for successful bone healing, and the phenotypic transition of macrophages in the inflammatory phase plays indispensable roles during the bone healing process. The goal of this paper is to design a microRNA delivery nanocarrier for strictly temporal guidance of the polarization of macrophages by the sequential delivery of different microRNAs. The results showed that microRNA nanocarriers, synthesized through free radical polymerization, could be internalized by macrophages with about a cellular uptake efficiency of 80%, and the sequential delivery of microRNA-155 nanocarriers and microRNA-21 nanocarriers proved, for the first time, that it could promote an efficient and timely switch from the M1 to the M2 phenotype along the time point of bone tissue repair. The strategy proposed in this paper holds potential for controlling sequential M1-to-M2 polarization of macrophages, which provides another perspective for the treatment of bone tissue regeneration.
Collapse
Affiliation(s)
- Xueping Li
- Tianjin
Key Laboratory of Composite and Functional Materials, School of Materials
Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Suling Xue
- Tianjin
Key Laboratory of Composite and Functional Materials, School of Materials
Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Qi Zhan
- Tianjin
Key Laboratory of Composite and Functional Materials, School of Materials
Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Xiaolei Sun
- Tianjin
Key Laboratory of Composite and Functional Materials, School of Materials
Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Ning Chen
- Tianjin
Key Laboratory of Composite and Functional Materials, School of Materials
Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Sidi Li
- College
of Chemistry and Chemical Engineering, Yantai
University, Yantai 264005, Shandong Province, China
| | - Jin Zhao
- Tianjin
Key Laboratory of Composite and Functional Materials, School of Materials
Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Xin Hou
- Tianjin
Key Laboratory of Composite and Functional Materials, School of Materials
Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Xubo Yuan
- Tianjin
Key Laboratory of Composite and Functional Materials, School of Materials
Science and Engineering, Tianjin University, Tianjin 300072, China
| |
Collapse
|
33
|
Zhao Q, Liu X, Yu C, Xiao Y. Macrophages and Bone Marrow-Derived Mesenchymal Stem Cells Work in Concert to Promote Fracture Healing: A Brief Review. DNA Cell Biol 2022; 41:276-284. [PMID: 35196145 DOI: 10.1089/dna.2021.0869] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cell (BMSC)-based and macrophage-based cell therapy are regarded as promising strategies to promote fracture healing because of incredible osteogenic potential of BMSCs and typical immunomodulatory function of macrophages. Apart from their respective key roles, accumulative evidence has also demonstrated the importance of cross talk between these two cell types in fracture healing process. This review takes a deep insight into the recent research progress of the synergic performance of BMSCs and macrophages by discussing not only the cells own functions but also the relevant impact factors and mechanisms (ambient microenvironment stimulus, miRNAs, etc). The aim of this review is to provide some valuable cues and technique support for the macrophage- and BMSC-related research, which will be helpful to propel BMSC/macrophage-based combined cell therapy for bone fracture treatment.
Collapse
Affiliation(s)
- Qing Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu, China
| | - Xinran Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu, China
| | - Chuanying Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu, China
| | - Yu Xiao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu, China
| |
Collapse
|
34
|
Guo Y, Jiang F, Yang W, Shi W, Wan J, Li J, Pan J, Wang P, Qiu J, Zhang Z, Li B. Effect of 1α,25(OH) 2D 3-Treated M1 and M2 Macrophages on Cell Proliferation and Migration Ability in Ovarian Cancer. Nutr Cancer 2021; 74:2632-2643. [PMID: 34894920 DOI: 10.1080/01635581.2021.2014903] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The biological active form of vitamin D3, 1α,25-dehydroxyvitamin D3 [1α,25(OH)2D3], exerts pleiotropic effects including bone mineralization, anti-tumor, as well as immunomodulator. This study aimed to explore the potential impact of 1α,25(OH)2D3 on tumor-associated macrophages (TAMs) infiltration in ovarian cancer. Firstly, human monocytic THP-1 cells were differentiated into macrophages (M0) in the presence of phorbol 12-myristate 13-acetate (PMA). In Vivo, 1α,25(OH)2D3 not only reversed the polarization of M2 macrophages, but also decreased the proliferation and migration abilities of ovarian cancer cells induced by M2 macrophages supernatant. Furthermore, 1α,25(OH)2D3 dramatically decreased the secretion of TGF-β1 and MMP-9 in M2 macrophages. However, no significant effect was observed in 1α,25(OH)2D3 treated M1 macrophages. In Vivo, vitamin D3 had an inhibitive effect of 1α,25(OH)2D3-treated M2 macrophages on tumorigenesis. In addition, we conducted the association of TAMs with the poor prognosis of patients with ovarian cancer by meta-analysis, which suggested the higher proportion of M2 macrophages was related to the poorer prognosis in ovarian cancer. Collectively, these results identified distinct roles of 1α,25(OH)2D3 treated M1 and M2 macrophages on cell proliferation and migration abilities in ovarian cancer.
Collapse
Affiliation(s)
- Yi Guo
- Medical College of Soochow University, Suzhou, China.,Jiangpu Community Healthcare Service, Suzhou, Kunshan, China
| | - Fei Jiang
- Medical College of Soochow University, Suzhou, China
| | - Wenqing Yang
- Medical College of Soochow University, Suzhou, China
| | - Weiqiang Shi
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianmei Wan
- Medical College of Soochow University, Suzhou, China
| | - Jie Li
- Medical College of Soochow University, Suzhou, China
| | - Jinjing Pan
- Medical College of Soochow University, Suzhou, China
| | - Ping Wang
- Medical College of Soochow University, Suzhou, China
| | - Junlan Qiu
- Medical College of Soochow University, Suzhou, China.,Department of Oncology and Hematology, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, China
| | - Zengli Zhang
- Medical College of Soochow University, Suzhou, China
| | - Bingyan Li
- Medical College of Soochow University, Suzhou, China
| |
Collapse
|
35
|
Bone formation recovery with gold nanoparticle-induced M2 macrophage polarization in mice. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2021; 38:102457. [PMID: 34400295 DOI: 10.1016/j.nano.2021.102457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 07/09/2021] [Accepted: 07/28/2021] [Indexed: 11/20/2022]
Abstract
The prevention of fractures induced by inflammatory bone disease remains a clinical challenge. This is because of a lack of bone formation to fill in the bone defects, which are believed to be due in part to persistent inflammation caused by the imbalance of M1 over M2 macrophages. In this study, gold nanoparticles (AuNPs) were synthesized to shift the balance of macrophages at the site of bone damage to improve osteanagenesis in a mouse model of LPS-induced inflammatory bone erosion. Specifically, the AuNPs treatment improved bone structure and increased bone mineral density (BMD) by ~14% compared with model group. Macrophages recruited by LPS treatment were reduced by ~11% after AuNPs injection. Compared to LPS treatment only, the percentage of M2 macrophages increased threefold by AuNPs, while the proportion of M1 macrophages decreased by 59%. This promoted the regeneration of bone matrix proteins in the bone defect site, which finally leads to increased bone mass and improved bone structure in model mice. These data suggest that AuNPs could be a novel candidate therapeutic for inflammatory bone disease rather than a drug carrier.
Collapse
|
36
|
Liao X, Lan Y, Shao R, Liu J, Liang S, Yin Z, Gudmundsson GH, Bergman P, Wan M. Vitamin D Enhances Neutrophil Generation and Function in Zebrafish (Danio rerio). J Innate Immun 2021; 14:229-242. [PMID: 34564076 DOI: 10.1159/000519183] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 08/23/2021] [Indexed: 11/19/2022] Open
Abstract
Vitamin D (VD) is a major regulator of calcium metabolism in many living organisms. In addition, VD plays a key role in regulating innate and adaptive immunity in vertebrates. Neutrophils constitute an important part of the first line of defense against invading microbes; however, the potential effect of VD on neutrophils remains elusive. Thus, in this study zebrafish in different developmental stages were utilized to identify the potential role of VD in the basal homeostasis and functions of neutrophils. Our results showed that addition of exogenous VD3 promoted granulopoiesis in zebrafish larvae. Reciprocally, neutrophil abundance in the intestine of adult zebrafish with a cyp2r1 mutant, lacking the capacity to 25-hydroxylate VD, was reduced. Moreover, VD-mediated granulopoiesis was still observed in gnotobiotic zebrafish larvae, indicating that VD regulates neutrophil generation independent of the microbiota during early development. In contrast, VD was incapable to influence granulopoiesis in adult zebrafish when the commensal bacteria were depleted by antibiotic treatment, suggesting that VD might modulate neutrophil activity via different mechanisms depending on the developmental stage. In addition, we found that VD3 augmented the expression of il-8 and neutrophil recruitment to the site of caudal fin amputation. Finally, VD3 treatment significantly decreased bacterial counts and mortality in zebrafish infected with Edwardsiella tarda (E. tarda) in a neutrophil-dependent manner. Combined, these findings demonstrate that VD regulates granulopoiesis and neutrophil function in zebrafish immunity.
Collapse
Affiliation(s)
- Xinmeng Liao
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Yawen Lan
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Rui Shao
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Jiayu Liu
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Shufei Liang
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Zhan Yin
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | | | - Peter Bergman
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,The Immunodeficiency Unit, Infectious Disease Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Min Wan
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China.,Pilot National Laboratory of Marine Science and Technology, Qingdao, China
| |
Collapse
|
37
|
Yu OB, Webb DA, Di Milo ES, Mutchie TR, Teske KA, Chen T, Lin W, Peluso-Iltis C, Rochel N, Helmstädter M, Merk D, Arnold LA. Biological evaluation and synthesis of calcitroic acid. Bioorg Chem 2021; 116:105310. [PMID: 34482171 DOI: 10.1016/j.bioorg.2021.105310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 11/26/2022]
Abstract
We describe the synthesis and broad profiling of calcitroic acid (CTA) as vitamin D receptor (VDR) ligand. The x-ray co-crystal structure of the Danio Rerio VDR ligand binding domain in complex with CTA and peptide MED1 confirmed an agonistic conformation of the receptor. CTA adopted a similar conformation as 1,25(OH)2D3 in the binding pocket. A hydrogen bond with His333 and a water molecule were observed in the binding pocket, which was accommodated due to the shorter CTA side chain. In contrast, 1,25(OH)2D3 interacted with His423 and His333 due to its longer side chain. In vitro, the EC50 values of CTA and CTA-ME for VDR-mediated transcription were 2.89 µM and 0.66 µM, respectively, confirming both compounds as VDR agonists. CTA was further evaluated for interaction with fourteen nuclear receptors demonstrating selective activation of VDR. VDR mediated gene regulation by CTA in intestinal cells was observed for the VDR target gene CYP24A1. CTA at 10 µM upregulated CYP24A1 with similar efficacy as 1,25(OH)2D3 at 20 nM and 100-fold stronger compared to lithocholic acid at 10 µM. CTA reduced the transcription of iNOS and IL-1β in interferon γ and lipopolysaccharide stimulated mouse macrophages resulting in a reduction of nitric oxide production and secretion of IL-1β. These observed anti-inflammatory properties of 20 µM CTA were similar to 20 nM 1,25(OH)2D3.
Collapse
Affiliation(s)
- Olivia B Yu
- Department of Chemistry and Biochemistry and Milwaukee Institute for Drug Discovery (MIDD), University of Wisconsin, 3210 N Cramer Street, Milwaukee, WI 53211, USA
| | - Daniel A Webb
- Department of Chemistry and Biochemistry and Milwaukee Institute for Drug Discovery (MIDD), University of Wisconsin, 3210 N Cramer Street, Milwaukee, WI 53211, USA
| | - Elliot S Di Milo
- Department of Chemistry and Biochemistry and Milwaukee Institute for Drug Discovery (MIDD), University of Wisconsin, 3210 N Cramer Street, Milwaukee, WI 53211, USA
| | - Tania R Mutchie
- Department of Chemistry and Biochemistry and Milwaukee Institute for Drug Discovery (MIDD), University of Wisconsin, 3210 N Cramer Street, Milwaukee, WI 53211, USA
| | - Kelly A Teske
- Department of Chemistry and Biochemistry and Milwaukee Institute for Drug Discovery (MIDD), University of Wisconsin, 3210 N Cramer Street, Milwaukee, WI 53211, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, 262 Danny Thomas Place, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Wenwei Lin
- Department of Chemical Biology and Therapeutics, 262 Danny Thomas Place, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Carole Peluso-Iltis
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM, U1258/CNRS, UMR 7104, University of Strasbourg, 67404 Illkirch, France
| | - Natacha Rochel
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM, U1258/CNRS, UMR 7104, University of Strasbourg, 67404 Illkirch, France
| | - Moritz Helmstädter
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, D-60438 Frankfurt, Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, D-60438 Frankfurt, Germany
| | - Leggy A Arnold
- Department of Chemistry and Biochemistry and Milwaukee Institute for Drug Discovery (MIDD), University of Wisconsin, 3210 N Cramer Street, Milwaukee, WI 53211, USA.
| |
Collapse
|
38
|
Adipose Tissue Immunometabolism and Apoptotic Cell Clearance. Cells 2021; 10:cells10092288. [PMID: 34571937 PMCID: PMC8470283 DOI: 10.3390/cells10092288] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 08/26/2021] [Accepted: 08/30/2021] [Indexed: 12/19/2022] Open
Abstract
The safe removal of apoptotic debris by macrophages—often referred to as efferocytosis—is crucial for maintaining tissue integrity and preventing self-immunity or tissue damaging inflammation. Macrophages clear tissues of hazardous materials from dying cells and ultimately adopt a pro-resolving activation state. However, adipocyte apoptosis is an inflammation-generating process, and the removal of apoptotic adipocytes by so-called adipose tissue macrophages triggers a sequence of events that lead to meta-inflammation and obesity-associated metabolic diseases. Signals that allow apoptotic cells to control macrophage immune functions are complex and involve metabolites released by the apoptotic cells and also metabolites produced by the macrophages during the digestion of apoptotic cell contents. This review provides a concise summary of the adipocyte-derived metabolites that potentially control adipose tissue macrophage immune functions and, hence, may induce or alleviate adipose tissue inflammation.
Collapse
|
39
|
Zarobkiewicz MK, Morawska I, Michalski A, Roliński J, Bojarska-Junak A. NKT and NKT-like Cells in Autoimmune Neuroinflammatory Diseases-Multiple Sclerosis, Myasthenia Gravis and Guillain-Barre Syndrome. Int J Mol Sci 2021; 22:9520. [PMID: 34502425 PMCID: PMC8431671 DOI: 10.3390/ijms22179520] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 12/14/2022] Open
Abstract
NKT cells comprise three subsets-type I (invariant, iNKT), type II, and NKT-like cells, of which iNKT cells are the most studied subset. They are capable of rapid cytokine production after the initial stimulus, thus they may be important for polarisation of Th cells. Due to this, they may be an important cell subset in autoimmune diseases. In the current review, we are summarising results of NKT-oriented studies in major neurological autoimmune diseases-multiple sclerosis, myasthenia gravis, and Guillain-Barre syndrome and their corresponding animal models.
Collapse
Affiliation(s)
- Michał K. Zarobkiewicz
- Department of Clinical Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (I.M.); (A.M.); (J.R.)
| | | | | | | | - Agnieszka Bojarska-Junak
- Department of Clinical Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (I.M.); (A.M.); (J.R.)
| |
Collapse
|
40
|
The Crosstalk between Mesenchymal Stem Cells and Macrophages in Bone Regeneration: A Systematic Review. Stem Cells Int 2021; 2021:8835156. [PMID: 34221025 PMCID: PMC8219422 DOI: 10.1155/2021/8835156] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 02/28/2021] [Accepted: 05/13/2021] [Indexed: 12/24/2022] Open
Abstract
Bone regeneration is a complex and well-coordinated process that involves crosstalk between immune cells and resident cells in the injury site. Transplantation of mesenchymal stem cells (MSCs) is a promising strategy to enhance bone regeneration. Growing evidence suggests that macrophages have a significant impact on osteogenesis during bone regeneration. However, the precise mechanisms by which macrophage subtypes influence bone regeneration and how MSCs communicate with macrophages have not yet been fully elucidated. In this systematic literature review, we gathered evidence regarding the crosstalk between MSCs and macrophages during bone regeneration. According to the PRISMA protocol, we extracted literature from PubMed and Embase databases by using "mesenchymal stem cells" and "macrophages" and "bone regeneration" as keywords. Thirty-three studies were selected for this review. MSCs isolated from both bone marrow and adipose tissue and both primary macrophages and macrophage cell lines were used in the selected studies. In conclusion, anti-inflammatory macrophages (M2) have significantly more potential to strengthen bone regeneration compared with naïve (M0) and classically activated macrophages (M1). Transplantation of MSCs induced M1-to-M2 transition and transformed the skeletal microenvironment to facilitate bone regeneration in bone fracture and bone defect models. This review highlights the complexity between MSCs and macrophages, providing more insight into the polarized macrophage behavior in this evolving field of osteoimmunology. The results may serve as a useful reference for definite success in MSC-based therapy based on the critical interaction with macrophages.
Collapse
|
41
|
Treatment with a long-acting chimeric CSF1 molecule enhances fracture healing of healthy and osteoporotic bones. Biomaterials 2021; 275:120936. [PMID: 34303178 DOI: 10.1016/j.biomaterials.2021.120936] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/28/2021] [Accepted: 05/25/2021] [Indexed: 12/27/2022]
Abstract
Macrophage-targeted therapies, including macrophage colony-stimulating factor 1 (CSF1), have been shown to have pro-repair impacts post-fracture. Preclinical/clinical applications of CSF1 have been expedited by development of chimeric CSF1-Fc which has extended circulating half-life. Here, we used mouse models to investigate the bone regenerative potential of CSF1-Fc in healthy and osteoporotic fracture. We also explored whether combination of CSF1-Fc with interleukin (IL)-4 provided additional fracture healing benefit in osteopenic bone. Micro-computed tomography, in situ histomorphometry, and bone mechanical parameters were used to assess systemic impacts of CSF1-Fc therapy in naive mice (male and female young, adult and geriatric). An intermittent CSF1-Fc regimen was optimized to mitigate undesirable impacts on bone resorption and hepatosplenomegaly, irrespective of age or gender. The intermittent CSF1-Fc regimen was tested in a mid-diaphyseal femoral fracture model in healthy bones with treatment initiated 1-day post-fracture. Weekly CSF1-Fc did not impact osteoclasts but increased osteal macrophages and improved fracture strength. Importantly, this treatment regimen also improved fracture union and strength in an ovariectomy-model of delayed fracture repair. Combining CSF1-Fc with IL-4 initiated 1-week post-fracture reduced the efficacy of CSF1-Fc. This study describes a novel strategy to specifically achieve bone regenerative actions of CSF1-Fc that has the potential to alleviate fragility fracture morbidity and mortality.
Collapse
|
42
|
Lei H, He M, He X, Li G, Wang Y, Gao Y, Yan G, Wang Q, Li T, Liu G, Du W, Yuan Y, Yang L. METTL3 induces bone marrow mesenchymal stem cells osteogenic differentiation and migration through facilitating M1 macrophage differentiation. Am J Transl Res 2021; 13:4376-4388. [PMID: 34150020 PMCID: PMC8205672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 12/11/2020] [Indexed: 06/12/2023]
Abstract
Despite the crucial role of m6A methyltransferase METTL3 in multiple diseases onset and progression, there are still lacking hard evidence proving that METTL3 could affect macrophage polarization in the stage of bone repair. Here, we aimed to explore the potential involvement of METTL3 in bone repair through modulating macrophage polarization and decipher the underlying cellular/molecular mechanisms. Here we treated RAW 264.7 cells and BM-derived primary macrophages (BMDM) with lipopolysaccharide (LPS) to induce M1 differentiation. METTL3 expression was upregulated in pro-inflammatory macrophages (M1) as compared with macrophages (M0). And overexpression of METTL3 promoted the expression of IL-6 and iNOS secretion by M1 macrophage. In the coculture condition, M1 macrophages with forced expression of METTL3 significantly enhanced migration ability of BMSCs, and also remarkably facilitated osteogenesis ability of BMSCs; the opposite was true when expression of METTL3 was knockdown. In addition, the m6A-RIP microarray suggested that METTL3 silencing significantly reduce the m6A modification of DUSP14, HDAC5 and Nfam1. Furthermore, the findings showed that expression of HADC5 was downregulated in M1 macrophages with METTL3 knockdown, while the DUSP14 expression had slight change and Nfam1 expression was very low. In contrast, METTL3 overexpression promoted HDAC5 expression, indicating that HDAC5 is the critical target gene of METTL3. Under such a theme, we proposed that METTL3 overexpression might be a new approach of replacement therapy for the treatment of bone repair.
Collapse
Affiliation(s)
- Hong Lei
- Department of Pharmacology, College of Pharmacy, Harbin Medical UniversityHarbin 150081, China
| | - Mingyu He
- Department of Pharmacology, College of Pharmacy, Harbin Medical UniversityHarbin 150081, China
| | - Xiaoqi He
- Department of Pharmacology, College of Pharmacy, Harbin Medical UniversityHarbin 150081, China
| | - Guanghui Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical UniversityHarbin 150081, China
| | - Yang Wang
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical UniversityHarbin 150081, China
- Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical UniversityHarbin 150081, China
| | - Yuelin Gao
- Department of Pharmacology, College of Pharmacy, Harbin Medical UniversityHarbin 150081, China
| | - Gege Yan
- Department of Pharmacology, College of Pharmacy, Harbin Medical UniversityHarbin 150081, China
| | - Quan Wang
- Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical UniversityHarbin 150081, China
| | - Tao Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical UniversityHarbin 150081, China
| | - Guoxin Liu
- Department of Pharmacology, College of Pharmacy, Harbin Medical UniversityHarbin 150081, China
| | - Weijie Du
- Department of Pharmacology, College of Pharmacy, Harbin Medical UniversityHarbin 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, 2019RU070Harbin 150081, China
| | - Ye Yuan
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical UniversityHarbin 150081, China
- Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical UniversityHarbin 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, 2019RU070Harbin 150081, China
| | - Lei Yang
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical UniversityHarbin 150081, China
| |
Collapse
|
43
|
Calcitriol in the Presence of Conditioned Media from Metastatic Breast Cancer Cells Enhances Ex Vivo Polarization of M2 Alternative Murine Bone Marrow-Derived Macrophages. Cancers (Basel) 2020; 12:cancers12113485. [PMID: 33238581 PMCID: PMC7700498 DOI: 10.3390/cancers12113485] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 12/31/2022] Open
Abstract
Simple Summary In this study, we stimulated bone marrow-derived macrophages to M0, M1, and M2 subtypes, with or without calcitriol, or with or without 4T1 (metastatic), 67NR (non-metastatic), and Eph4-Ev (normal) cell culture supernatants (CMs) to test their effect on polarization. We showed that calcitriol increased the expression of Cd206 and Spp1 mRNA and CD36, CCL2, and arginase levels for M2 macrophages and decreased Cd80 and Spp1 mRNA and IL-1, IL-6, OPN, and iNOS for M1 macrophages. 4T1 CM influenced the expression of the studied genes and proteins to a greater extent than 67NR and Eph4; the strongest effect was noted for M2 macrophages. We show that calcitriol and 4T1 CM enhance the polarization of M2 macrophages and M2 macrophages differentiated with calcitriol-stimulated migration of 4T1 and 67NR cells. We indicate that the immunosuppressive properties of calcitriol may unfavorably affect the tumor microenvironment, and supplementation with vitamin D in oncological patients may not always bring benefits. Abstract In this study, we differentiated murine bone marrow-derived macrophages (BMDMs) into M0, M1, and M2 in the presence or absence of calcitriol. Real-time PCR analysis of gene expression, FACS analysis of surface markers, and chemokine/cytokine production assays were performed. In addition, the effect of the conditioned media (CM) from murine breast cancer 4T1 (metastatic) and 67NR (non-metastatic) and Eph4-Ev (normal) cells with and without calcitriol on the polarization of M1/M2 cells was determined. We found that calcitriol enhanced the differentiation of M2 macrophages, which was manifested by increased expression of Cd206 and Spp1 mRNA and CD36, Arg, and CCL2 in M2 BMDMs and by decreased expression of Cd80 and Spp1 mRNA and IL-1, IL-6, OPN, and iNOS in M1 BMDMs. 4T1 CM showed a higher effect on the gene and protein expression in macrophages than 67NR and Eph4-Ev, with the greatest effect observed on M2 macrophages which increased their differentiation and properties characteristic of alternative macrophages. Moreover, M2 macrophages differentiated with calcitriol-stimulated migration of 4T1 and 67NR cells through fibronectin and collagen type IV, respectively. Overall, our results indicated that vitamin D supplementation may not always be beneficial, especially in relation to cancers causing excessive, pathological activation of the immune system.
Collapse
|
44
|
Roy S, Spinali K, Schmuck EG, Kink JA, Hematti P, Raval AN. Cardiac fibroblast derived matrix-educated macrophages express VEGF and IL-6, and recruit mesenchymal stromal cells. ACTA ACUST UNITED AC 2020; 10. [PMID: 33564732 DOI: 10.1016/j.regen.2020.100033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The polarization of monocytes into macrophages that possess anti-inflammatory and pro-angiogenic properties could provide a novel therapeutic strategy for patients who are at a high risk for developing heart failure following myocardial infarction (MI). Here in, we describe a novel method of "educating" monocytes into a distinct population of macrophages that exhibit anti-inflammatory and pro-angiogenic features through a 3-day culture on fibronectin-rich cardiac matrix (CX) manufactured using cultured human cardiac fibroblasts. Our data suggest that CX can educate monocytes into a unique macrophage population termed CX educated macrophages (CXMq) that secrete high levels of VEGF and IL-6. In vitro, CXMq also demonstrate the ability to recruit mesenchymal stromal cells (MSC) with known anti-inflammatory properties. Selective inhibition of fibronectin binding to αVβ3 surface integrins on CXMq prevented MSC recruitment. This suggests that insoluble fibronectin within CX is, at least in part, responsible for CXMq conversion.
Collapse
Affiliation(s)
- Sushmita Roy
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Keith Spinali
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Eric G Schmuck
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - John A Kink
- Division of Hematology/Oncology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Peiman Hematti
- Division of Hematology/Oncology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
| | - Amish N Raval
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
45
|
Fleet JC, Bernal-Mizrachi C, Hewison M, Jorde R, White J, Cantorna M, Schoenmakers I, Erben R. Highlights from the 22nd workshop on vitamin D in New York City, May 2019. J Steroid Biochem Mol Biol 2020; 203:105747. [PMID: 32858157 DOI: 10.1016/j.jsbmb.2020.105747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- James C Fleet
- Department of Nutrition Science, College of Health and Human Sciences, Purdue University, 700 West State St., West Lafayette, IN 47906-2059, USA.
| | - Carlos Bernal-Mizrachi
- Division of Endocrinology, Metabolism & Lipid Research, School of Medicine, Washington University, 660 S. Euclid Ave. Campus Box 8127, St. Louis, MO 63110, USA.
| | - Martin Hewison
- Institute of Metabolism and Systems Research, Centre for Endocrinology, Diabetes, and Metabolism, Birmingham Health Partners, Level 2, IBR, Rom 225, The University of Birmingham, Birmingham, B15 2TT, UK.
| | - Rolf Jorde
- Endocrine Research Group, Department of Clinical Medicine, UiT The Arctic University of Norway, 9037 Tromsø, Norway.
| | - John White
- Department of Physiology, Department of Medicine, McGill University, 3655 Drummond St. Room 1112, Montreal, QC H3G 1Y6, Canada.
| | - Margherita Cantorna
- Department of Veterinary and Biomedical Sciences, College of Agricultural Sciences, 125 Henning Building, University Park, PA 16802, USA.
| | - Inez Schoenmakers
- Department of Medicine, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, NR4, 7TJ, UK.
| | - Reinhold Erben
- Institute of Physiology, Pathophysiology, and Biophysics, Unit of Physiology, Pathophysiology, and Experimental Endocrinology, Veterinärplatz 1, 1210 Wien, Vienna, Austria.
| |
Collapse
|
46
|
Zhao SJ, Liu H, Chen J, Qian DF, Kong FQ, Jie J, Yin GY, Li QQ, Fan J. Macrophage GIT1 Contributes to Bone Regeneration by Regulating Inflammatory Responses in an ERK/NRF2-Dependent Way. J Bone Miner Res 2020; 35:2015-2031. [PMID: 32460388 PMCID: PMC7689802 DOI: 10.1002/jbmr.4099] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 12/12/2022]
Abstract
Despite the best treatment, approximately 10% of fractures still face undesirable repair. Recently, many studies have focused on the importance of macrophages in bone repair; however, the cellular mechanisms by which they work are not yet fully understood. In this study, we explored the functions of macrophage G-protein-coupled receptor interacting protein 1 (GIT1) in healing a tibial monocortical defect model. Using GIT1flox/flox Lyz2-Cre (GIT1 CKO) mice, we observed that a GIT1 deficiency in the macrophages led to an exacerbation of interleukin 1β (IL1β) production, more M1-like macrophage infiltration, and impaired intramembranous ossification in vivo. The results of in vitro assays further indicated that the macrophage GIT1 plays a critical role in several cellular processes in response to lipopolysaccharide (LPS), such as anti-oxidation, IL1β production alleviation, and glycolysis control. Although GIT1 has been recognized as a scaffold protein, our data clarified that GIT1-mediated extracellular-signal-regulated kinase (ERK) phosphorylation could activate nuclear factor (erythroid-derived 2)-like 2 (NRF2) in macrophages after LPS treatment. Moreover, we demonstrated that macrophage GIT1-activated ERK/NRF2 negatively regulates the 6-phosphofructo-2-kinase/fructose-2, 6-biphosphatase 3 (PFKFB3), facilitating the decrease of glycolysis. Our findings uncovered a previously unrecognized role of GIT1 in regulating ERK/NRF2 in macrophages to control the inflammatory response, suggesting that macrophage GIT1 could be a potential target to improve bone regeneration. © 2020 The Authors. Journal of Bone and Mineral Research published by American Society for Bone and Mineral Research..
Collapse
Affiliation(s)
- Shu-Jie Zhao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jian Chen
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ding-Fei Qian
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fan-Qi Kong
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jian Jie
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Orthopedics, Pukou Branch of Jiangsu Province Hospital (Nanjing Pukou Central Hospital), Nanjing, China
| | - Guo-Yong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qing-Qing Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jin Fan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
47
|
Paschalidi P, Gkouveris I, Soundia A, Kalfarentzos E, Vardas E, Georgaki M, Kostakis G, Erovic BM, Tetradis S, Perisanidis C, Nikitakis NG. The role of M1 and M2 macrophage polarization in progression of medication-related osteonecrosis of the jaw. Clin Oral Investig 2020; 25:2845-2857. [PMID: 32964311 DOI: 10.1007/s00784-020-03602-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/17/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVES The aim of this study was to investigate the relationship between M1 and M2 macrophage polarization and clinical stage in patients with medication-related osteonecrosis of the jaw (MRONJ) who underwent treatment with bisphosphonates or denosumab. MATERIALS AND METHODS M1 and M2 macrophage density and expression of interleukin (IL)-6 and IL-10 were assessed on biopsies of mucosal tissues surrounding necrotic bone in 30 MRONJ patients with stages 1-3 and controls. For identification of M1 and M2 macrophages, double CD68/iNOS and CD68/CD206 immunofluorescence staining was conducted, respectively. Computer-assisted immunofluorescence quantification of markers was performed. RESULTS Early stage 1 MRONJ patients showed a switch toward the M2 phenotype, as indicated by the higher density of M2 macrophages, the decreased M1/M2 ratio, and the upregulation of IL-10. MRONJ patients with advanced stages 2 and 3 showed a shift toward M1-polarized macrophages, as suggested by the higher density of M1 macrophages, the increased M1/M2 ratio, and the overexpression of IL-6. The macrophage density of both M1 and M2 subsets was significantly enhanced in patients receiving bisphosphonates compared with those receiving denosumab. CONCLUSIONS The M1-M2 macrophage polarization status in mucosal tissues bordering necrotic bone correlates with clinical stage of MRONJ. Patients with early-stage MRONJ show a switch toward M2-polarized macrophages, while MRONJ patients with advanced stage demonstrate a shift toward the M1 phenotype. CLINICAL RELEVANCE Therapeutic molecules targeting the inflammatory microenvironment via the regulation of either M1 or M2 macrophage polarization may represent a novel strategy for treatment of MRONJ.
Collapse
Affiliation(s)
- Polytimi Paschalidi
- Department of Oral and Maxillofacial Surgery, School of Dentistry, National and Kapodistrian University of Athens, Thivon 2, Goudi, Athens, Greece.
| | - Ioannis Gkouveris
- Department of Oral Medicine and Pathology, School of Dentistry, National and Kapodistrian University of Athens, Thivon 2, Goudi, Athens, Greece
| | - Akrivoula Soundia
- Division of Diagnostic and Surgical Sciences, UCLA School of Dentistry, Los Angeles, CA, 90095, USA
| | - Evangelos Kalfarentzos
- Department of Oral and Maxillofacial Surgery, School of Dentistry, National and Kapodistrian University of Athens, Thivon 2, Goudi, Athens, Greece
| | - Emmanouil Vardas
- Clinic of Hospital Dentistry, School of Dentistry, National and Kapodistrian University of Athens, Thivon 2, Goudi, Athens, Greece
| | - Maria Georgaki
- Department of Oral Medicine and Pathology, School of Dentistry, National and Kapodistrian University of Athens, Thivon 2, Goudi, Athens, Greece
| | - Georgios Kostakis
- Department of Oral and Maxillofacial Surgery, School of Dentistry, National and Kapodistrian University of Athens, Thivon 2, Goudi, Athens, Greece
| | - Boban M Erovic
- Institute of Head and Neck Diseases, Evangelical Hospital Vienna, Hans-Sachs Gasse 10-12, 1180, Vienna, Austria
| | - Sotirios Tetradis
- Division of Diagnostic and Surgical Sciences, UCLA School of Dentistry, Los Angeles, CA, 90095, USA
| | - Christos Perisanidis
- Department of Oral and Maxillofacial Surgery, School of Dentistry, National and Kapodistrian University of Athens, Thivon 2, Goudi, Athens, Greece
| | - Nikolaos G Nikitakis
- Department of Oral Medicine and Pathology, School of Dentistry, National and Kapodistrian University of Athens, Thivon 2, Goudi, Athens, Greece
| |
Collapse
|
48
|
Hozain S, Cottrell J. CDllb+ targeted depletion of macrophages negatively affects bone fracture healing. Bone 2020; 138:115479. [PMID: 32535290 DOI: 10.1016/j.bone.2020.115479] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/06/2020] [Accepted: 06/09/2020] [Indexed: 01/13/2023]
Abstract
Inflammation is an important part of the fracture repair process which requires osteogenic cells to interact with innate immune cells such as macrophages. All murine macrophages express the F4/80 cell surface marker but they may be further subdivided into two main phenotypes: M1 (proinflammatory) or M2 (anti-inflammatory) based on surface marker expression and function. Macrophages polarize between these two main classes in response to inflammation while differentially regulating the healing process. Studies have shown that F4/80+ cell ablation impairs fracture healing, however, the distinct phenotypes that participate in the early healing process is unclear. We hypothesized that the M1 subtype is essential for the early steps of fracture healing and their depletion would impair fracture repair. To test this hypothesis, M1 (F4/80+/MHCII+/CD86+/CDllb+) macrophages were depleted using a saporin conjugated Mac-1 antibody (Mac1SAP) in vitro using primary macrophages and in vivo using a mouse femur fracture model. Primary macrophages isolated from mice femoral bone marrow were either left undifferentiated (+PBS), differentiated into M1 macrophages (+LPS), or differentiated to M2 macrophages (+IL-4), and then treated with either vehicle or 10 pM Mac1SAP. Samples were collected at day 2 and 5 post Mac1SAP treatment. Macrophage subtypes were identified by flow cytometry and cytokine secretion profiles were quantified using xMAP. For the in vivo model, mice were treated with Mac1SAP 24 h prior to fracture. Femur bone marrow samples were collected and analyzed by flow cytometry, xMAP, immunohistochemistry, MicroCT, and histology. The results demonstrated that Mac1SAP significantly depleted M1 macrophages both in vivo and in vitro. Mac1SAP treatment altered expression of 75% of cytokines in vitro and 30% of cytokines in vivo including IL-6, TNF-a, and IP-10. In both the in vitro and in vivo models, the M1 subtype correlated highly with cytokines G-CSF, IL-1α, IL-6, IL-10, LIX, KC, MCP-1, IP-10, MIP1α, MIP1β, RANTES, IL-9, IL-2 and TNFα. M1 depletion was also found to reduced callus properties at day 14 via microCT analysis. Overall, the data suggests that depletion of M1 macrophages by Mac1SAP treatment alters the cytokine expression profiles during early bone repair which ultimately impairs bone healing.
Collapse
Affiliation(s)
- Sarah Hozain
- Seton Hall University, South Orange, NJ 07079, United States of America
| | - Jessica Cottrell
- Seton Hall University, South Orange, NJ 07079, United States of America.
| |
Collapse
|
49
|
McCauley J, Bitsaktsis C, Cottrell J. Macrophage subtype and cytokine expression characterization during the acute inflammatory phase of mouse bone fracture repair. J Orthop Res 2020; 38:1693-1702. [PMID: 31989683 DOI: 10.1002/jor.24603] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 01/07/2020] [Accepted: 01/21/2020] [Indexed: 02/04/2023]
Abstract
Fracture repair is a complex process requiring heterotypic interactions between osteogenic cells and immune cells. Recent evidence indicates that macrophages are critically involved in fracture repair. Polarized macrophage populations differentially promote and regulate inflammation in other tissues, but little is known about the various macrophage subtypes and their signaling activities following a bone fracture. The authors hypothesized that classically activated (M1 subtype) and alternatively activated (M2 subtype) macrophages are active during the early repair process to initiate and regulate the inflammatory response. To test our hypothesis, bone marrow was collected from intact femurs (naïve group), contralateral and fractured femurs of mice on days 0, 1, 2, 4, and 7 postfracture. Macrophages were isolated from the bone marrow and macrophage subtypes were identified using flow cytometry with antibodies to F4/80, MHC II, CD86, CD11c, and CD40. Bone marrow cytokine levels were measured using xMAP. Flow cytometry revealed dynamic changes in M1 subtype (F4/80+/MHC II+/CD86+), M2 subtype (F4/80+/MHC II-/CD86-), and dendritic cell (DCs; MHCII+/CD11c+/CD40+) populations following fracture as compared to naïve controls. M1 subtype levels were correlated with IL-1α, IL-1ß, IL-2, IL-17, Eotaxin, and MCP-1, while DCs were correlated with IL-6, G-CSF, LIF, KC, and VEGF-A. The results indicate that M1 and M2 subtypes and DCs are recruited to the fracture site early during the repair process and consequently may work in tandem to regulate the inflammatory response required to recruit osteogenic cells needed for later stages of repair.
Collapse
Affiliation(s)
- James McCauley
- Department of Biological Sciences, Seton Hall University, South Orange, New Jersey
| | | | - Jessica Cottrell
- Department of Biological Sciences, Seton Hall University, South Orange, New Jersey
| |
Collapse
|
50
|
Martineau C, Kaufmann M, Arabian A, Jones G, St-Arnaud R. Preclinical safety and efficacy of 24R,25-dihydroxyvitamin D 3 or lactosylceramide treatment to enhance fracture repair. J Orthop Translat 2020; 23:77-88. [PMID: 32518749 PMCID: PMC7270532 DOI: 10.1016/j.jot.2020.03.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 02/05/2020] [Accepted: 03/25/2020] [Indexed: 01/22/2023] Open
Abstract
Background/Objective Cyp24a1-null mice deficient in 24,25(OH)2D3 display impaired callus formation during the endochondral phase of bone fracture repair. The 24,25(OH)2D3 metabolite acted by binding to the TLC domain containing 3B isoform 2 (TLCD3B2, previously named FAM57B2) effector protein, which then synthesizes lactosylceramide (LacCer). Treatment with 24,25(OH)2D3 or LacCer restored callus size and mechanical properties in Cyp24a1-null mice. Methods To assess the safety of these molecules and test their efficacy for bone healing in wild-type, non-genetically modified mice, we treated 12-week-old, osteotomized C57BL/6 female mice with each compound for up to 21 days post-osteotomy. Control cohorts were injected with vehicle. Results Neither compound was found to exhibit any nephro- nor hepato-toxicity. Calcemia remained stable throughout the experiment and was unaffected by either treatment. Supplementation with 24,25(OH)2D3 increased circulating levels of this metabolite about 8-fold, decreased 1,25(OH)2D3 levels, and significantly increased circulating 1,24,25(OH)3D3 levels, suggesting 1?-hydroxylation of 24,25(OH)2D3. TLCD3B2 was found to be expressed in fracture callus at the surface of unmineralized or pre-mineralized cartilage on day 10 and day 12 post-osteotomy and to progressively recede to become undetectable by day 18. Treatment with 24,25(OH)2D3 or LacCer reduced the number of TLCD3B2-positive cells. Both treatments also significantly increased stiffness and elastic modulus of the healing bone callus. Conclusion Exogenous administration of 24,25(OH)2D3 or LacCer improved the biomechanical properties of repaired bones in wild-type animals without affecting circulating calcium levels or other blood parameters, demonstrating preclinical safety and efficacy. Translational potential Our data suggest the use of 24R,25-dihydroxyvitamin D3 or lactosylceramide for ameliorating fracture healing in clinical practice.
Collapse
Affiliation(s)
- Corine Martineau
- Research Centre, Shriners Hospitals for Children – Canada, Montreal, Quebec, H4A 0A9, Canada
| | - Martin Kaufmann
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, K7L 3N6, Canada
- Department of Surgery, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| | - Alice Arabian
- Research Centre, Shriners Hospitals for Children – Canada, Montreal, Quebec, H4A 0A9, Canada
| | - Glenville Jones
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| | - René St-Arnaud
- Research Centre, Shriners Hospitals for Children – Canada, Montreal, Quebec, H4A 0A9, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, H3A 1A1, Canada
- Department of Surgery, McGill University, Montreal, Quebec, H3A 1A1, Canada
- Department of Medicine, McGill University, Montreal, Quebec, H3A 1A1, Canada
- Research Institute of the McGill University Health Centre, Montreal, Quebec, H3H 2R9, Canada
- Corresponding author. Research Centre, Shriners Hospitals for Children – Canada, 1003 Decarie Boulevard, Montreal, Quebec, H4A 0A9, Canada.
| |
Collapse
|