1
|
Yang Y, Li H, Ma Z, Li Z, Gu J. Lamb1-mediated Wnt/β-catenin signaling pathway drives endothelial angiogenesis for fracture healing. Gene 2025; 959:149481. [PMID: 40221061 DOI: 10.1016/j.gene.2025.149481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/20/2025] [Accepted: 04/06/2025] [Indexed: 04/14/2025]
Abstract
OBJECTIVES Fractures, usually caused by trauma or osteoporosis, are the most common traumatic injuries to large organs in humans. Osteogenesis and angiogenesis are two crucial parts of fracture healing that work together to promote the repair and regeneration of damaged bone. Endothelial cell migration is critical for angiogenesis. Therefore, it is well worth exploring whether endothelial cells (ECs) can enhance fracture healing. METHODS The public datasets were analyzed by scRNA-seq, and the ECs were subjected to subset analysis and pseudotime analysis. Next, ECs_Lamb1+ cells underwent GO and KEGG pathway enrichment analyses, and were subjected to GSVA. Finally, the mechanism was verified and evaluated via qRT-PCR, cellular immunofluorescence staining, and transwell assay. RESULTS After cell annotations, 9 cell types were obtained, and it was found that the proportions of ECs were significantly reduced. EC subset analysis showed that the ratio of ECs_Lamb1+ cells was significantly up-regulated in the Fracture group; pseudotime analysis showed that ECs_Lamb1- cells were gradually reduced over time, whereas ECs_Lamb1+ cells were gradually expanding along the trajectories to reach a maximum at the end of the trajectory; pathway enrichment analyses revealed that ECs_Lamb1+ cells were mainly associated with several signaling pathways regulating cell proliferation, differentiation, repair, angiogenesis, and inflammatory responses, such as PI3K-Akt signaling pathway, Wnt/β-catenin, and MAPK. The results of basic assays demonstrated that successful knockdown of Lamb1 expression via siRNA-LAMB1 was detrimental to HUVEC proliferation, migration, and tube formation, and could suppress the expression of wnt3a, GSK-3β, β-catenin, and VEGFA; whereas, HY-141873 in combination with siRNA-LAMB1 partially reversed the down-regulated wnt3a, GSK-3β, β-catenin, and VEGFA expression, and HUVEC proliferation, migration, and tube formation were partially improved. CONCLUSION Lamb1 promotes fracture repair and healing by up-regulating VEGFA expression via the activation of Wnt signaling pathway to catalyze EC growth and migration and induce endothelial angiopoiesis.
Collapse
Affiliation(s)
- Yajun Yang
- People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, China.
| | - Hangyu Li
- People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, China
| | - Zhirong Ma
- People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, China
| | | | | |
Collapse
|
2
|
Zhu Z, Bai Z, Cui Y, Li X, Zhu X. The potential therapeutic effects of Panax notoginseng in osteoporosis: A comprehensive review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156703. [PMID: 40354676 DOI: 10.1016/j.phymed.2025.156703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Accumulating evidence shows that Panax notoginseng, a well-known medicinal herb, has an ideal effect on prevention and treatment of skeletal diseases. In this study, we reviewed clinical applications of clinical application as well as phytochemistry, pharmacokinetics, pharmacology in improving bone quality and toxicity of Panax notoginseng. PURPOSE Review the phytochemistry, pharmacokinetics, pharmacology involved in the improving bone metabolism and toxicity of Panax notoginseng and evaluate its potential as a traditional Chinese herbal medicine for osteoporosis. METHODS Several databases were consulted, including PubMed, China National Knowledge Infrastructure, National Science and Technology Library and Web of Science. The following words or phrases were used alone or in combinations in the titles and/or abstracts: "","Panax notoginseng", "Sanqi", "osteoporosis", "bone", "osteoblast", "osteoclast", "phytochemistry", "pharmacology" and "pharmacokinetics". Altogether 160 papers were cited. RESULTS 8 clinical trials of Panax notoginseng alone for the treatment of osteoporosis were identified, most of which used traditional Chinese patent medicines to treat osteoporosis fractures. In these clinical trials, Panax notoginseng preparations have achieved relatively good therapeutic effects. However, more rigorous large-scale experiments are expected to prove their efficacy. Phytochemistry study showed that saponins, flavonoids, polysaccharides are the main active ingredients extracted from Panax notoginseng and the transformation of saponins during the processing explains the different effects of raw and cooked Panax notoginseng. The pharmacokinetics data reveals that protopanaxdiol-type (ppd-type) saponins possesses higher bioavailability than protopanaxtriol-type(ppt-type) saponins and ppd-type saponins such as ginsenoside Ra3, Rb1, and Rd can represent suitable pharmacokinetic markers for Panax notoginseng extracts. The data from animal experiments demonstrates that Panax notoginseng can improve bone quality in ovariectomized, diabetic, hyperlipidemia, radiation-induced, and arthritis rats through the regulation of anti-adipogenesis, anti-inflammation, anti-oxidation, angiogenesis and estrogenic effects. In vitro experiments, the activities of improving bone quality of Panax notoginseng and its ingredients may be attributed to the regulation of multiple signaling pathways, including Wnt/β-catenin, BMP/BMP-R, AMPK/mTOR, GPER/PI3K/AKT, etc. Acute and chronic toxicity as well as genotoxicity studies show that Panax notoginseng is well tolerated while long term use may lead to liver and kidney toxicity. CONCLUSIONS Panax notoginseng is a superior medicinal herb that contains multiple active ingredients and could play a potential role in the prevention and treatment of osteoporosis. Further studies should concentrate on developing Panax notoginseng products with higher curative effect and bioavailability.
Collapse
Affiliation(s)
- Zijun Zhu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong 510630, PR China
| | - Zhenyu Bai
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong 510630, PR China
| | - Yan Cui
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong 510630, PR China
| | - Xiaoyun Li
- College of Pharmacy, Jinan University, Guangzhou, Guangdong 510630, PR China
| | - XiaoFeng Zhu
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, PR China; School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong 510630, PR China.
| |
Collapse
|
3
|
Kibritoglu E, Yuksel H. Numerical analysis of coil designs to expedite fracture healing using dielectrophoresis with S method. Comput Biol Med 2025; 192:110213. [PMID: 40279972 DOI: 10.1016/j.compbiomed.2025.110213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/11/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Classical methods for speeding up fracture healing usually rely on direct electrical stimulation and electromagnetic fields to boost the levels of growth factors at the fracture site. However, these techniques often concentrate on bone cells themselves rather than addressing the critical blood flow dynamics necessary for effective healing. This study introduces a mathematical model designed to explore the potential of dielectrophoretic forces (DEPFs) in improving blood flow at the fracture site. By adjusting blood flow, the model seeks to enhance the delivery of vital nutrients, hormones, and growth factors, including endothelial cells (ECs), vascular endothelial growth factor (VEGF) and oxygen, which are essential for accelerating the fracture healing process. METHOD The proposed approach includes a new technique, termed the S method, which assesses the non-uniformity of DEPFs by algebraically analyzing the electric field lines associated with positive and negative dielectrophoresis. We developed analytical equations to simulate various coil configurations, focusing on long bone fractures where blood flow is vertically oriented. The DEPF Factor (χDEPF) was used to measure the ratio of blood flow velocity in the presence of DEPFs compared to the absence of DEPFs, thus indicating the effectiveness of DEPF in enhancing blood flow. RESULTS The simulation results revealed that DEPF reaches its peak efficacy at the gamma dispersion band, with the most significant enhancement occurring at a frequency of 15 MHz. Specifically, the average values of χDEPF were 1.8, 3.2, and 7.9 for the catenary, lintearia, and valeria coils, respectively. Our computational model, which incorporated VEGF, ECs, and oxygen tension, demonstrated that the catenary coil slightly improved healing rates in impaired fractures, the lintearia coil normalized healing times between impaired and normal fractures, and the valeria coil not only accelerated healing in impaired fractures but also enhanced healing in normal fractures. CONCLUSIONS This paper's findings suggest that the valeria coil exhibits the best DEPF functionality, making it the optimal configuration for future experimental studies aimed at evaluating the efficacy of DEPF in promoting fracture healing. The ability of DEPFs to significantly enhance blood flow could represent a substantial advancement in the treatment of both normal and impaired fractures.
Collapse
Affiliation(s)
- Erman Kibritoglu
- Department of Electrical and Electronics Engineering, Bogazici University, Bebek, İstanbul, 34342, Turkey
| | - Heba Yuksel
- Department of Electrical and Electronics Engineering, Bogazici University, Bebek, İstanbul, 34342, Turkey.
| |
Collapse
|
4
|
Lin X, Deng S, Fu T, Lei Y, Wang Y, Yao J, Lu Y, Huang Y, Shang J, Chen J, Zhou X. Hyaluronic acid-based hydrogel microspheres with multi-responsive properties for antibacterial therapy and bone regeneration in Staphylococcus aureus-infected skull defects. Mater Today Bio 2025; 32:101676. [PMID: 40236808 PMCID: PMC11997343 DOI: 10.1016/j.mtbio.2025.101676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/20/2025] [Accepted: 03/16/2025] [Indexed: 04/17/2025] Open
Abstract
This study introduces hyaluronic acid-based (HA) hydrogel microspheres loaded with zinc oxide nanoparticles (ZnO-NPs) for the treatment of infectious bone defects. The microspheres were fabricated using a 3D-printing process, with a formulation consisting of 6 wt% HAD (methacrylated HA), 3 wt% AOHA (AMP-conjugated oxidized HA), 1 % BOHA (phenylboric acid-conjugated HA), 0.5 % photoinitiator, and 0.05 % ZnO-NPs. In vitro, the hydrogel microspheres demonstrated significant antibacterial activity against Staphylococcus aureus, with colony counts and biofilm inhibition assays showing a marked reduction in bacterial growth after 12 and 24 h. The release of antimicrobial peptides (AMPs) was enhanced in acidic conditions and in the presence of hyaluronidase. The microspheres also promoted osteogenic differentiation of bone marrow stromal cells (BMSCs), as evidenced by increased expression of osteogenic markers (ALP, OCN, OPN, and COL-1). In vivo, the hydrogel microspheres were tested in a rat skull defect model, showing significant bone regeneration, improved angiogenesis, and an anti-inflammatory response. These results indicate that ABOHA@ZnO hydrogel microspheres provide a promising strategy for treating infectious bone defects by combining antimicrobial, osteogenic.
Collapse
Affiliation(s)
- Xiaolong Lin
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Shuli Deng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Tao Fu
- Department of Oral and Maxillofacial Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, School of Stomatology and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, 310000, China
| | - Yuqing Lei
- Department of Minimally Invasive Interventional Radiology, The Second Affiliated Hospital, School of Biomedical Engineering Guangzhou Medical University, Guangzhou, Guangdong, 510260, China
| | - Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Jiapei Yao
- Department of Orthopedics, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Changzhou, 213000, China
| | - Yaojun Lu
- Department of Orthopedics, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Changzhou, 213000, China
| | - Yong Huang
- Department of Orthopedics, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Changzhou, 213000, China
| | - Jingjing Shang
- Department of Pharmacy, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Changzhou, Jiangsu, 213000, China
| | - Jingjing Chen
- Department of Orthopedics, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Changzhou, 213000, China
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases, NHC Key Laboratory of Antibody Technique, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Xindie Zhou
- Department of Orthopedics, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Changzhou, 213000, China
- Department of Orthopedics, Gonghe County Hospital of Traditional Chinese Medicine, Hainan Tibetan Autonomous Prefecture, Qinghai Province, 811800, China
| |
Collapse
|
5
|
Li Y, Han Q, Chen Y, Huang W, Chen Y, Wu Z, Tan F, Liu M, Zou J. Effect of intraarticular human umbilical cord mesenchymal stem cells transplantation on cartilage degradation and matrix metalloproteinases in OA rat model. BMC Musculoskelet Disord 2025; 26:538. [PMID: 40450237 DOI: 10.1186/s12891-025-08797-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 05/22/2025] [Indexed: 06/03/2025] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a common cause of disability around the world, but the pathophysiology is still poorly understood. The study sought to investigate the effects of umbilical cord mesenchymal stem cells (UC-MSCs) injection in OA, with a focus on cartilage degradation. METHODS The serum matrix metalloproteinase (MMPs) and a disintegrin and metalloprotease domains with thrombospondins motifs (ADAMTS) levels in OA patients were examined using ELISA. The levels of MMPs and ADAMTS in peripheral blood mononuclear cells (PBMCs) following co-cultured with UC-MSC were determined by ELISA. The anterior cruciate ligament transection (ACLT) surgery was performed on the knee joints of a rat OA model, followed by intra-articular injection of UC-MSCs at 4 weeks and 8 weeks after surgery, and the changes of the severity of OA, tibiofemoral cartilage, and subchondral bone were observed by Safranin-O staining, hematoxylin and eosin (H&E) staining, and micro-CT imaging. RESULTS OA patients showed the higher protein levels of MMP2, 9, 13, ADAMTS4 and 5 than healthy controls. Furthermore, in vitro incubation of PBMC derived from OA patients with UC-MSCs down-regulated the protein expression of these proteases. Positive correlations were observed between serum MMP9 levels and high-sensitivity C-reactive protein (hsCRP) in OA patients, while a negative correlation between serum MMP13 and Alkaline phosphatase (ALKP) was observed in these patients. There was a negative correlation between OA patients' serum ADAMTS 4 and Lipoprotein (a)(Lp(a)). Western blotting and immunohistostaining were applied to assess the protein levels of matrix metalloproteinases and ADAMTSs in rat knee joints and these protein levels were significantly decreased in the UC-MSC intra-articular injection group. Micro-CT results showed in the OA rat model human UC-MSCs treatment alleviated the destruction of the tibial subchondral bone. CONCLUSION UC-MSCs decreased the level of several matrix proteases, slowing the progression of OA formation in rats. Our findings suggested that matrix metalloproteinases and ADAMTSs play a role in OA progression, UC-MSC exerted beneficial therapeutic effects on OA rat model by reducing the levels of these matrix metalloproteinases and ADAMTSs.
Collapse
Affiliation(s)
- Yanqun Li
- Dongguan EnlifeStem cell Biotechnology Research Institute, Nancheng, Dongguan, China
- Dongguan New type Research and development institution, Guangdong, China
| | - Qipeng Han
- Yuanpin Cell Biotechnology Group Co., Ltd., Hunan Province, Changsha, Hunan, China
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Yansha Chen
- Central Laboratory, Binhaiwan Central Hospital of Dongguan, Guangdong, China
| | - Wenlong Huang
- Dongguan Qingshi Biotechnology Co., LTD, Dongguan, China
| | - Yanqing Chen
- Central Laboratory, Binhaiwan Central Hospital of Dongguan, Guangdong, China
| | - Zhihuang Wu
- Central Laboratory, Binhaiwan Central Hospital of Dongguan, Guangdong, China
| | - Fengming Tan
- Dongguan EnlifeStem cell Biotechnology Research Institute, Nancheng, Dongguan, China
- Dongguan New type Research and development institution, Guangdong, China
| | - Muyun Liu
- National Engineering Research Center of Foundational Technologies for CGT Industry, Shenzhen, China.
| | - Jiangying Zou
- Dongguan EnlifeStem cell Biotechnology Research Institute, Nancheng, Dongguan, China.
- Dongguan New type Research and development institution, Guangdong, China.
| |
Collapse
|
6
|
Lu L, Xu Z, Miao Z, Zuo X, Shi D, Chang S, Luo P, Li G. Higher systemic immune-inflammation index associates with vertebral marrow proton density fat fraction in postmenopausal women. J Bone Miner Metab 2025:10.1007/s00774-025-01609-8. [PMID: 40425866 DOI: 10.1007/s00774-025-01609-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 04/30/2025] [Indexed: 05/29/2025]
Abstract
INTRODUCTION The systemic immune-inflammation index (SII) may influence bone homeostasis through inflammatory modulation. Although bone marrow adipocytes regulate bone metabolism via adipokine secretion, their interaction with SII remains unexplored. We investigated the SII-marrow adiposity relationship in postmenopausal women. MATERIALS AND METHODS This retrospective study included 187 postmenopausal women. Lumbar spine MRI using chemical shift encoding generated proton density fat fraction (PDFF) maps, with bone mineral density (BMD) measured by dual x-ray absorptiometry. The relationship between SII and marrow PDFF was evaluated through multivariable-adjusted linear regression, smooth curve fittings, and threshold analysis. RESULTS The results revealed a negative correlation between marrow PDFF values and BMD (r = - 0.438, P < 0.001). After accounting for age, time since menopause, body mass index, physical activity, C-reactive protein, interleukin (IL)-1β, IL-6, tumor necrosis factor-α, and BMD in the regression analysis, each unit increase in SII was found to be inked to an increase of 0.247 (β = 0.247; 95% confidence interval [CI], 0.212 to 0.281; P <0.001) in PDFF. After converting SII to a categorical variable (quartiles), participants in the highest SII quartile had a 16.8% higher vertebral marrow PDFF than those in the lowest SII quartile (β = 16.753, 95% CI: 11.036-18.522, P <0.001). Furthermore, a curvilinear relationship and threshold effect were also identified. Turning point was identified at the SII value of 441 on the adjusted smooth curve. CONCLUSIONS SII levels were positively associated with marrow adiposity in postmenopausal women.
Collapse
Affiliation(s)
- Li Lu
- Department of Radiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai, 200437, China
| | - Zheng Xu
- Changshou Community Health Center, Shanghai, China
| | - Zeyang Miao
- Department of Radiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai, 200437, China
| | - Xiaoyong Zuo
- Department of Radiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai, 200437, China
| | - Dan Shi
- Department of Geriatrics, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shixin Chang
- Department of Radiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai, 200437, China
| | - Peng Luo
- Department of Radiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai, 200437, China.
| | - Guanwu Li
- Department of Radiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai, 200437, China.
| |
Collapse
|
7
|
Tambun DV, Tanandika J, Carlita C, Ayub FA, Ramadhani R, Dewi RS, Djais A, Gultom F, Sunarso S, Amir LR. Titanium Surface Roughness Mediated Macrophages Polarization-Influenced Osteogenic Differentiation of Periodontal Ligament-Derived Mesenchymal Stromal Cells. Eur J Dent 2025. [PMID: 40334680 DOI: 10.1055/s-0045-1804889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025] Open
Abstract
OBJECTIVES Implant surface topography significantly influences cell behavior, including macrophages and bone cell interactions. The polarization of macrophages, key immune cells, is influenced by implant surface characteristics. This research aimed to examine periodontal ligament mesenchymal stromal cells (PDL MSCs) responses to the polarized macrophages induced by titanium surface roughness. MATERIALS AND METHODS RAW 264.7 macrophages were cultured with various surface roughness of titanium disks. Macrophage adhesion and polarization were evaluated by scanning electron microscope, gene expressions profiling, and flow cytometry. PDL MSCs were treated with conditioned medium of macrophages and analyzed with 3-[4,5-dimethylthiazol-2yl]-2,5-diphenyl-2H-tetrazolium bromide assay, real-time polymerase chain reaction, and Alizarin red staining. STATISTICAL ANALYSIS Data was statistically analyzed using GraphPad Prism 9 for Windows 11. The one-way analysis of variance test was used to compare the groups. Dunn post hoc test was used to compare the difference between the groups when appropriate. Significance was accepted when p < 0.05. RESULTS Medium surface roughness (Ti-MR) consistently inhibited tumor necrosis factor-α, interleukin-1β (IL-1β), and IL-6 gene expressions (p < 0.001) and upregulated transforming growth factor-β, vascular epithelial growth factor, and IL-10 expressions (p < 0.01). Confirmatory flow cytometry analysis showed consistent results, with Ti-HR and Ti-MR exhibiting the highest population of CD163+ cells (99.1 and 90.7%, respectively), while Ti-LR exhibited the lowest M1/M2 ratio (0.93). Furthermore, treatment of RAW 264.7 conditioned medium increased osteopontin, alkaline phosphatase, collagen type-1 A-1 chain, osteocalcin, runt-related transcription factor-2, and bone sialoprotein gene expressions and calcium deposition (p < 0.01). CONCLUSION Titanium implant surface topography influences macrophage polarization and osteogenic differentiation of PDL MSCs, with Ti-MR being the most effective in polarizing macrophages toward M2 and inducing optimal osteogenic responses from PDL MSCs.
Collapse
Affiliation(s)
- Dodi V Tambun
- Dentistry Study Program, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Jovanka Tanandika
- Dentistry Study Program, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Carlita Carlita
- Dentistry Study Program, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Fakhrana A Ayub
- Department of Prosthodontics, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Ratna Ramadhani
- Department of Oral Biology, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Ratna Sari Dewi
- Department of Prosthodontics, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Ariadna Djais
- Department of Oral Biology, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Ferry Gultom
- Department of Oral Biology, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Sunarso Sunarso
- Department of Dental Material, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Lisa R Amir
- Department of Oral Biology, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
8
|
Zhu L, Yuan X. Associations between monocyte to HDL-C ratio and lumbar bone mineral density in alcohol dependent individuals with depression. Sci Rep 2025; 15:15817. [PMID: 40328867 PMCID: PMC12056117 DOI: 10.1038/s41598-025-00885-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 05/02/2025] [Indexed: 05/08/2025] Open
Abstract
Osteoporosis, a skeletal disorder that reduces bone density, is a significant health concern. Alcohol dependence, a chronic condition, exacerbates public health problems due to its widespread occurrence and association with various comorbidities, including depression. This study aims to explore the relationship between monocyte to high-density lipoprotein cholesterol ratio (MHR) and lumbar bone mineral density (BMD)in individuals with alcohol dependence and depression. From 2009 to 2018, 49,693 participants were enrolled, and after screening, the study included 2,055 individuals with alcohol-dependency and depression. In this study, multivariate regression analysis was employed to assess the association between monocyte to HDL-C ratio and lumbar bone mineral density. Additionally, we conducted interaction tests and subgroup analysis. The result showed a negative correlation between MHR and lumbar BMD, which remained significant even after adjusting for covariates. Individuals with less than a 9th-grade education showed a positive link with MHR, while those with a college degree or higher had a negative link. This relationship remained significant in the fully adjusted model. A U-shaped relationship between MHR and lumbar BMD was observed in individuals with a high school diploma/GED, after adjusting for various factors. The intricate correlation between MHR and lumbar BMD may suggest the presence of biological interplays and disparities in socioeconomic or behavioral aspects. This underscores the necessity for tailored public health strategies that cater to various educational demographics.
Collapse
Affiliation(s)
- Lili Zhu
- Department of Infection Management, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiaofeng Yuan
- Department of Spine Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China.
- The Third Affiliated Hospital of Soochow University, Changzhou, China.
| |
Collapse
|
9
|
Liu A, Sun Y, Qi X, Zhou Y, Zhou J, Li Z, Wu X, Zou Z, Lv X, Li H, Li Y. Nonlinear association between liver fat content and lumbar bone mineral density in overweight and obese individuals: evidence from a large-scale health screening data in China. Endocrine 2025; 88:446-456. [PMID: 39869295 PMCID: PMC12069136 DOI: 10.1007/s12020-025-04168-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/14/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND The impact of fatty liver disease on lumbar bone mineral density (BMD) represents an intriguing area of study, particularly in light of established research linking obesity to bone metabolism. However, there remains limited investigation into the correlation between quantifying liver fat content (LFC) and lumbar BMD among overweight and obese populations, particularly within the Chinese demographic. This study aims to accurately quantify LFC and investigate its association with lumbar BMD in overweight or obese individuals. METHODS This cross-sectional study was conducted at the Health Management Center of Henan Provincial People's Hospital from January 2019 to February 2023, involving 6996 participants with a body mass index (BMI) of 24 kg/m² or higher. LFC and lumbar BMD were assessed using computed tomography. The study utilized one-way ANOVA, subgroup analysis, multifactor regression analysis, smooth curve fitting, and threshold and saturation effect analysis to explore the relationship between LFC and lumbar BMD. Furthermore, inflammatory cell analysis was included to investigate the potential mediating role of inflammatory cells in the association between LFC and lumbar BMD. RESULTS After adjusting for confounding variables, multivariate regression analysis revealed a significant negative association between LFC and lumbar BMD (β = -0.323, 95% CI: -0.464 to -0.183, P < 0.001). Particularly, participants in the highest baseline LFC quartile (Q4 group) exhibited a more pronounced negative impact on lumbar BMD compared to those in the lowest quartile (Q1 group) (β = -5.026, 95% CI: -7.040 to -3.012, P < 0.001). Threshold saturation effect analysis identified a turning point in the LFC-BMD relationship (K = 5.4). Below this point, LFC showed a positive correlation with lumbar BMD (β = 0.962, 95% CI: 0.016-1.907, P < 0.05), whereas above it, LFC was significantly negatively correlated with lumbar BMD (β = -0.405, 95% CI: -0.558 to -0.253, P < 0.001). Additionally, mediation analysis indicated that leukocytes and monocytes potentially mediated the association between LFC and lumbar BMD, with mediation ratios of -5.78 and -6.68%, respectively. CONCLUSION Among individuals categorized as overweight or obese, elevated levels of LFC were associated with reduced lumbar BMD, particularly noticeable above a threshold of 5.4%. Additionally, various types of inflammatory cells are presumed to exert a substantial mediating influence on the correlation between LFC and lumbar BMD.
Collapse
Affiliation(s)
- Ao Liu
- Department of Medical Imaging, People's Hospital of Zhengzhou University, #7 Wei Wu Road, Zhengzhou, 450003, China
| | - Yongbing Sun
- Department of Medical Imaging, People's Hospital of Zhengzhou University, #7 Wei Wu Road, Zhengzhou, 450003, China
| | - Xin Qi
- Department of Medical Imaging, Henan Provincial People's Hospital, Xinxiang Medical College, Zhengzhou, 450003, China
| | - Yang Zhou
- Department of Medical Imaging, People's Hospital of Zhengzhou University, #7 Wei Wu Road, Zhengzhou, 450003, China
| | - Jing Zhou
- Department of Health Management, Chronic Health Management Laboratory, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Zhonglin Li
- Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Xiaoling Wu
- Department of Nuclear Medicine, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Zhi Zou
- Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Xue Lv
- Department of Health Management, Chronic Health Management Laboratory, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Hao Li
- Department of Health Management, Fuwai Central China Cardiovascular Hospital, #1 Fuwai Avenue, Zhengzhou, 451464, China
| | - Yongli Li
- Department of Health Management, Chronic Health Management Laboratory, Henan Provincial People's Hospital, Zhengzhou, 450003, China.
| |
Collapse
|
10
|
Wang T, Zhang M, Guo J, Wei H, Li W, Luo Y. Alginate/bacterial cellulose/GelMA scaffolds with aligned nanopatterns and hollow channel networks for vascularized bone repair. Int J Biol Macromol 2025; 308:142578. [PMID: 40154692 DOI: 10.1016/j.ijbiomac.2025.142578] [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: 11/06/2024] [Revised: 03/16/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Designed macropores and nanopatterned surfaces are important architectural cues in three-dimensional (3D) scaffolds for promoting vascularization and bone regeneration. However, the fabrication of 3D scaffolds with both controlled nanopatterned surfaces and designed macropores remains a challenge, especially for hydrogel-based scaffolds. Herein, alginate (Alg)/bacterial cellulose (BC)/ Gelatin Methacryloyl (GelMA) composite scaffold with fully interconnected Hollow Channel Networks And An Aligned Nanopatterned Surface (HCAS) is fabricated using 3D printing, surface crosslinking, and prestretching/drying-induced orientation. The highly aligned nanofibrous structures significantly enhance the mechanical properties, as well as the structural stability of the hydrogel scaffold. In vitro experiments prove that the HCAS scaffold exhibits apparently enhanced angiogenic and osteogenic properties compared to the control groups since the aligned nanopatterns and hollow channels can activate the cyclic AMP-dependent Ras-related protein 1 (cAMP-RAP1) and mitogen-activated protein kinase (MAPK) pathways, respectively, and jointly promote the downstream phosphoinositide 3-kinase/hypoxia-inducible factor-1 (PI3K/HIF-1) pathway. In vivo experiments also show that HCAS scaffold significantly promotes vascularization and bone regeneration, further verifying the joint effect of the aligned nanopatterned surface and fully interconnected hollow channels in promoting vascularization and osteogenesis. Thus, the HCAS scaffold demonstrates that a cell- and growth factor-free approach can also promote satisfactory vascularization and bone regeneration, simply by creating nanopatterned surfaces and designed hollow channels within hydrogel scaffolds.
Collapse
Affiliation(s)
- Tianyu Wang
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), The First Affiliated Hospital of Shenzhen University, School of Medicine, Shenzhen University, Shenzhen 518055, China
| | - Mengqi Zhang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Jiali Guo
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Hao Wei
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Wencui Li
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), The First Affiliated Hospital of Shenzhen University, School of Medicine, Shenzhen University, Shenzhen 518055, China.
| | - Yongxiang Luo
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), The First Affiliated Hospital of Shenzhen University, School of Medicine, Shenzhen University, Shenzhen 518055, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China.
| |
Collapse
|
11
|
Huang Y, Mao J, Li Z, Wang W, Ni Z, Cai F, Tang J, Wang W, Zhang L, Zhou L, Jiang X, Wu J, Guo Q, Rui M, Huang Z, Jiang H, Wang L, Xi K, Gu Y, Chen L. Signal Converter-Based Therapy Platform Promoting Aging Bone Healing by Improving Permeability of the Mitochondrial Membrane. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2500156. [PMID: 40289881 DOI: 10.1002/adma.202500156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/19/2025] [Indexed: 04/30/2025]
Abstract
The aging microenvironment promotes persistent inflammation and loss of intrinsic regenerative capacity. These are major obstacles to effective bone tissue repair in older adults. This study aims to explore how physical thermal stimulation can effectively delay the bone marrow mesenchymal stem cells (BMSCs) aging process. Based on this, an implantable physical signal-converter platform is designed as a therapeutic system that enables stable heat signals at the bone injury site under ultrasound stimulation (US). It is found that the therapeutic platform controllably reduces the mitochondrial outer membrane permeabilization of aging BMSCs, bidirectionally inhibiting mitochondrial reactive oxygen species and mitochondrial DNA (mtDNA) leakage. The leakage ratio of mtDNA decreases by 22.7%. This effectively mitigates the activation of the cGAS-STING pathway and its downstream NF-κB signaling induced by oxidative stress in aging BMSCs, thereby attenuating the pathological advancement of chronic inflammation. Thus, it effectively restores the metabolism and osteogenic differentiation of aging BMSCs in vitro, which is further confirmed in a rat model. In the GMPG/US group, the bone mineral density increases 2-3 times at 4 weeks in the rats femoral defect model. Therefore, this ultrasound-based signal-conversion platform provides a promising strategy for aging bone defect repair.
Collapse
Affiliation(s)
- Yiyang Huang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Jiannan Mao
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
- Department of Orthopedics, Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, 163 Shoushan Road, Jiang Yin, 214400, P. R. China
| | - Ziang Li
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Wenbo Wang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Zhengxia Ni
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Feng Cai
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Jincheng Tang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Wei Wang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Lichen Zhang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Liang Zhou
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Xinzhao Jiang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Jie Wu
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Qiangqiang Guo
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Min Rui
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
- Department of Orthopedics, Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, 163 Shoushan Road, Jiang Yin, 214400, P. R. China
| | - Ziyan Huang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Haochen Jiang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Lingjun Wang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Kun Xi
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Yong Gu
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Liang Chen
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| |
Collapse
|
12
|
Iaquinta MR, De Pace R, Benkhalqui A, D’Agostino A, Trevisiol L, Finotti A, Breveglieri G, Tognon M, Martini F, Mazzoni E. Secretome Release During In Vitro Bone Marrow-Derived Mesenchymal Stem Cell Differentiation Induced by Bio-Oss ® Collagen Material. Int J Mol Sci 2025; 26:3807. [PMID: 40332455 PMCID: PMC12027874 DOI: 10.3390/ijms26083807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/03/2025] [Accepted: 04/12/2025] [Indexed: 05/08/2025] Open
Abstract
Bone diseases represent a growing healthcare challenge due to population aging and lifestyle changes. Although bone has a natural regenerative capacity, approximately 10% of fractures fail to heal properly, requiring advanced therapeutic approaches. Bone tissue engineering (BTE) has advanced the use of osteoinductive and osteoconductive biomaterials to support bone regeneration. Among them, Bio-Oss® Collagen, a composite of bovine hydroxyapatite and collagen, has shown excellent biocompatibility and bioactivity properties. This study analyzes the effect of Bio-Oss® Collagen on human bone marrow-derived mesenchymal stem cells (hBMSCs), assessing its osteoinductive and immunomodulatory potential. After 7 days of culture, the biomaterial modulated the expression of key genes involved in osteogenesis and chondrogenesis, which are known for their role in bone formation and maturation. At the same time, a downregulation of genes associated with bone resorption was observed. Secretome analysis revealed a controlled release of pro-regenerative cytokines, suggesting a role of the biomaterial in modulating inflammation to promote bone regeneration. Furthermore, immunofluorescence confirmed the high expression of osteocalcin and osteopontin, which are key markers of bone mineralization. These findings indicate that Bio-Oss® Collagen supports osteogenesis and modulates the immune response, creating a microenvironment favorable for bone regeneration.
Collapse
Affiliation(s)
- Maria Rosa Iaquinta
- Laboratories of Cell Biology and Molecular Genetics, Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (M.R.I.); (M.T.); (F.M.)
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Raffaella De Pace
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Assia Benkhalqui
- Section of Dentistry and Maxillo-Facial Surgery, University of Verona, 37124 Verona, Italy; (A.B.); (A.D.)
| | - Antonio D’Agostino
- Section of Dentistry and Maxillo-Facial Surgery, University of Verona, 37124 Verona, Italy; (A.B.); (A.D.)
| | - Lorenzo Trevisiol
- Unit of Maxillo-Facial Surgery, Santa Chiara Regional Hospital, Provincial Healthcare Services Agency (APSS), 38122 Trento, Italy;
- Centre for Medical Sciences (CISMed), University of Trento, 38122 Trento, Italy
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, 44121 Ferrara, Italy; (A.F.); (G.B.)
| | - Giulia Breveglieri
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, 44121 Ferrara, Italy; (A.F.); (G.B.)
| | - Mauro Tognon
- Laboratories of Cell Biology and Molecular Genetics, Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (M.R.I.); (M.T.); (F.M.)
| | - Fernanda Martini
- Laboratories of Cell Biology and Molecular Genetics, Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (M.R.I.); (M.T.); (F.M.)
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
- Centre of Biotechnology, University of Ferrara, 44121 Ferrara, Italy
| | - Elisa Mazzoni
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy;
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
13
|
Zheng J, Ren Y, Ke J, Zhu G, Wang Z, Shi X, Wang Y. Engineered Extracellular Vesicles Derived from Juvenile Mice Enhance Mitochondrial Function in the Aging Bone Microenvironment and Achieve Rejuvenation. ACS NANO 2025; 19:13952-13967. [PMID: 40183704 DOI: 10.1021/acsnano.4c17989] [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: 04/05/2025]
Abstract
Aging-related bone degeneration and impaired healing capacity remain significant challenges in regenerative medicine, necessitating innovative, efficient, and targeted strategies to restore bone health. Here, we engineered extracellular vesicles (EVs) derived from the serum of pretreated juvenile mice, with the goals of reversing aging, enhancing osteogenic potential, and increasing bioavailability to rejuvenate the aging bone environment. First, we established bone healing models representing different phases of healing to identify the EV type with the highest potential for improving the bone microenvironment in older individuals. Second, we employed DSS6 for bone targeting to enhance the biological effects of the selected EVs in vivo. The engineered EVs effectively targeted bone repair sites and promoted fracture healing more effectively than unmodified EVs in older mice. RNA sequencing revealed that the translocase of outer mitochondrial membrane 7 (Tomm7) is crucial for the underlying mechanism. Silencing Tomm7 significantly diminished the positive regulatory effects of the EVs. Specifically, the engineered EVs may enhance mitochondrial function in aging cells by activating the Tomm7-mediated Pink1/Parkin mitophagy pathway, promoting stemness recovery in aging bone marrow stromal cells (BMSCs) and reversing the adverse conditions of the aging bone microenvironment. Overall, the developed engineered EVs derived from serum from juvenile mice offer an alternative approach for treating aging bones. The identified underlying biological mechanisms provide a valuable reference for precision treatment of aging bones in the future.
Collapse
Affiliation(s)
- Jiaqian Zheng
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
| | - Yipeng Ren
- Department of Stomatology, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, P. R. China
| | - Junhua Ke
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
| | - Guanglin Zhu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
| | - Zhen Wang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Department of Orthopedics, Xijing Hospital, Xi'an 710032, P. R. China
| | - Xuetao Shi
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
| | - Yingjun Wang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
| |
Collapse
|
14
|
Wang J, Xu S, Chen B, Qin Y. Advances in cell therapy for orthopedic diseases: bridging immune modulation and regeneration. Front Immunol 2025; 16:1567640. [PMID: 40276505 PMCID: PMC12018241 DOI: 10.3389/fimmu.2025.1567640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Orthopedic diseases pose significant challenges to public health due to their high prevalence, debilitating effects, and limited treatment options. Additionally, orthopedic tumors, such as osteosarcoma, chondrosarcoma, and Ewing sarcoma, further complicate the treatment landscape. Current therapies, including pharmacological treatments and joint replacement, address symptoms but fail to promote true tissue regeneration. Cell-based therapies, which have shown successful clinical results in cancers and other diseases, have emerged as a promising solution to repair damaged tissues and restore function in orthopedic diseases and tumors. This review discusses the advances and potential application of cell therapy for orthopedic diseases, with a particular focus on osteoarthritis, bone fractures, cartilage degeneration, and the treatment of orthopedic tumors. We explore the potential of mesenchymal stromal cells (MSCs), chondrocyte transplantation, engineered immune cells and induced pluripotent stem cells to enhance tissue regeneration by modulating the immune response and addressing inflammation. Ultimately, the integration of cutting-edge cell therapy, immune modulation, and molecular targeting strategies could revolutionize the treatment of orthopedic diseases and tumors, providing hope for patients seeking long-term solutions to debilitating conditions.
Collapse
Affiliation(s)
- Jing Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Shenghao Xu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Bo Chen
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Yanguo Qin
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin, China
- Joint International Research Laboratory of Ageing Active Strategy and Bionic Health in Northeast Asia of Ministry of Education, Jilin University, Changchun, Jilin, China
| |
Collapse
|
15
|
Sodré LI, Gall MEC, Elias MDB, de Oliveira LO, Lobo FATF, Carias RBV, Teodoro AJ. Osteogenic Effects of Bioactive Compounds Found in Fruits on Mesenchymal Stem Cells: A Review. Nutr Rev 2025; 83:675-691. [PMID: 39862385 DOI: 10.1093/nutrit/nuae209] [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] [Indexed: 01/27/2025] Open
Abstract
Phytochemicals, which are bioactive compounds contained in fruits, vegetables, and teas, have a positive effect on human health by having anti-inflammatory, antioxidant, and anticarcinogenic effects. Several studies have highlighted the ability of bioactive compounds to activate key cellular enzymes associated with important signaling pathways related to cell division and proliferation, as well as their role in inflammatory and immunological responses. Some phytochemicals are associated with increased proliferation, differentiation, and expression of markers related to osteogenesis, bone formation, and mineralization by activating various signaling pathways. The objective of this study was to clarify which bioactive compounds present in fruits have osteogenic effects on mesenchymal stem cells and the possible associated mechanisms. A literature search was conducted in the LILACS, MEDLINE, and PubMed databases for pertinent articles published between 2014 and 2024. This review included 34 articles that report the osteogenic effects of various bioactive compounds found in different fruits. All the articles reported that phytochemicals play a role in enhancing the regenerative properties of mesenchymal cells, such as proliferation, osteogenic differentiation, secretion of angiogenic factors, and extracellular matrix formation. This review highlights the potential of these phytochemicals in the prevention and treatment of bone diseases. However, more studies are recommended to identify and quantify the therapeutic dose of phytochemicals, investigate their mechanisms in humans, and ensure their safety and effectiveness for health, particularly for bone health.
Collapse
Affiliation(s)
- Lia Igel Sodré
- Graduate Program in Science of Nutrition, Fluminense Federal University, Niterói, RJ 24020-140, Brazil
| | - Maria Eduarda Cordebello Gall
- Graduate Program in Biotechnology, National Institute of Metrology Standardization and Industrial Quality, Xerém, RJ 25250-020, Brazil
| | - Monique de Barros Elias
- Graduate Program in Food and Nutrition Security, Fluminense Federal University/Faculty of Nutrition, Niterói, RJ 24020-140, Brazil
| | - Luana Oeby de Oliveira
- Programa de Pós-Graduação em Ciências Aplicadas a Produtos para a Saúde (PPG-CAPS)/Fluminense Federal University, Faculty of Nutrition, Niteroi, RJ 24020-140, Brazil
| | | | - Rosana Bizon Vieira Carias
- Regenerative Medicine Laboratory, Centro Universitário Arthur Sá Earp Neto, Petrópolis Medical School, Petrópolis, RJ 25680-120, Brazil
| | - Anderson Junger Teodoro
- Universidade Federal Fluminense (Fluminense Federal University), Nutrition and Dietetics Department, Food and Nutrition Integrated Center, Niterói, RJ CEP 24020-140, Brazil
| |
Collapse
|
16
|
Qi H, Zhang B, Lian F. 3D-printed bioceramic scaffolds for bone defect repair: bone aging and immune regulation. Front Bioeng Biotechnol 2025; 13:1557203. [PMID: 40242352 PMCID: PMC12000889 DOI: 10.3389/fbioe.2025.1557203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/06/2025] [Indexed: 04/18/2025] Open
Abstract
The management of bone defects, particularly in aging populations, remains a major clinical challenge. The immune microenvironment plays an important role in the repair of bone defects and a favorable immune environment can effectively promote the repair of bone defects. However, aging is closely associated with chronic low-grade systemic inflammation, which adversely affects bone healing. Persistent low-grade systemic inflammation critically regulates bone repair through all stages. This review explores the potential of 3D-printed bioceramic scaffolds in bone defect repair, focusing on their capacity to modulate the immune microenvironment and counteract the effects of bone aging. The scaffolds not only provide structural support for bone regeneration but also serve as effective carriers for anti-osteoporosis drugs, offering a novel therapeutic strategy for treating osteoporotic bone defects. By regulating inflammation and improving the immune response, 3D-printed bioceramic scaffolds may significantly enhance bone repair, particularly in the context of age-related bone degeneration. This approach underscores the potential of advanced biomaterials in addressing the dual challenges of bone aging and immune dysregulation, offering promising avenues for the development of effective treatments for bone defects in the elderly. We hope the concepts discussed in this review could offer novel therapeutic strategies for bone defect repair, and suggest promising avenues for the future development and optimization of bioceramic scaffolds.
Collapse
Affiliation(s)
- Haoran Qi
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Bo Zhang
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Feng Lian
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Hefei, Anhui, China
| |
Collapse
|
17
|
Menger MM, Manuschewski R, Hans S, Braun BJ, Kayali MKDE, Ehnert S, Ampofo E, Wrublewsky S, Menger MD, Histing T, Laschke MW. Age-related alterations of angiogenesis, inflammation and bone microarchitecture during fracture healing in mice. GeroScience 2025:10.1007/s11357-025-01584-y. [PMID: 40108067 DOI: 10.1007/s11357-025-01584-y] [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: 12/03/2024] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
The surgical treatment of geriatric patients represents a major challenge in traumatology. It is well known that aging affects fracture healing. However, the exact pathophysiology of age-related changes in angiogenesis, inflammation and bone remodeling remains still elusive. Therefore, we herein studied the differences of femoral fracture healing in young adult (3-4 months) and aged (16-18 months) CD-1 mice by using a stable closed femoral fracture model with intramedullary screw fixation. The callus tissue was analyzed by means of X-ray, micro-computed tomography (µCT), histology and immunohistochemistry. We found a deteriorated trabecular architecture and a reduced bone formation within the callus tissue of aged mice. Moreover, aged animals showed an increased number of tartrate-resistant acid phosphatase (TRAP)-positive osteoclasts at an early healing time point, whereas the fraction of mature α-smooth muscle actin (SMA)-positive microvessels was significantly reduced. Furthermore, the numbers of macrophages and granulocytes were higher in the callus tissue of aged animals at the end of the healing process. Taken together, these results demonstrate a delayed femoral fracture healing in aged CD-1 mice. This is most likely caused by an early overshooting osteoclast response, a decelerated maturation of the callus microvasculature and a late increased recruitment of pro-inflammatory cells. Targeting these alterations may contribute to the development of novel treatment approaches for the stimulation of bone regeneration in geriatric patients.
Collapse
Affiliation(s)
- Maximilian M Menger
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany.
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg, Germany.
| | - Ruben Manuschewski
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg, Germany
| | - Sandra Hans
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg, Germany
| | - Benedikt J Braun
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
| | - Moses K D El Kayali
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg, Germany
| | - Sabrina Ehnert
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
| | - Emmanuel Ampofo
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg, Germany
| | - Selina Wrublewsky
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg, Germany
| | - Tina Histing
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg, Germany
| |
Collapse
|
18
|
Sun H, Liu Y, Huang Y, Xiong K, Zhang Z, Wang W, Dai Y, Li J, Li Q, Wang S, Shi C. Echinococcus granulosus sensu lato promotes osteoclast differentiation through DUSP4-MAPK signaling in osseous echinococcosis. Front Microbiol 2025; 16:1558603. [PMID: 40177487 PMCID: PMC11961949 DOI: 10.3389/fmicb.2025.1558603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Osseous echinococcosis, caused by Echinococcus granulosus infection, is characterized by progressive bone destruction driven by abnormal osteoclast activation. Dual-specificity phosphatase 4 (DUSP4), a key negative regulator of the MAPK pathway, inhibits osteoclast differentiation and bone resorption. This study aimed to elucidate the role of DUSP4 in E. granulosus-induced bone loss. Methods In vitro, a co-culture system of E. granulosus protoscoleces (PSCs) and bone marrow-derived macrophages (BMMs) was established. Osteoclast differentiation and bone resorption were assessed using TRAP staining and F-actin immunofluorescence. Transcriptome sequencing identified DUSP4 as a key regulator. DUSP4 overexpression was performed to evaluate its effects on osteoclast markers and MAPK signaling (ERK, JNK, p38). In vivo, a mouse model of osseous echinococcosis was developed, and DUSP4 overexpression was achieved via lentiviral transduction. Bone destruction was analyzed using X-ray, micro-CT, and histology. Results PSCs significantly enhanced osteoclast differentiation and bone resorption, upregulated osteoclast markers (CTSK, NFATc1), and activated MAPK signaling. DUSP4 overexpression reversed these effects, reducing osteoclast activity and MAPK phosphorylation. In vivo, PSC infection caused severe bone destruction, which was mitigated by DUSP4 overexpression. Disscussion This study reveals the molecular mechanism by which Echinococcus granulosus drives abnormal osteoclast activation through the DUSP4-MAPK signaling axis. Parasitic infection suppresses DUSP4 expression, relieving its negative regulation of the MAPK pathway and leading to excessive osteoclast differentiation. Restoring DUSP4 expression effectively reverses abnormal MAPK pathway activation, reducing osteoclast bone resorption activity to physiological levels. These findings not only provide new insights into the pathological mechanisms of bone destruction in osseous echinococcosis but also establish DUSP4 as a critical therapeutic target for pathological bone resorption, laying the groundwork for host-directed treatment strategies for parasitic bone diseases.
Collapse
Affiliation(s)
- Haohao Sun
- The First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Yaqing Liu
- The First Affiliated Hospital of Shihezi University, Shihezi, China
- The Medical College of Shihezi University, Shihezi, China
| | - Yiping Huang
- The Medical College of Shihezi University, Shihezi, China
| | - Kangjun Xiong
- The Medical College of Shihezi University, Shihezi, China
| | - Zhendong Zhang
- The First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Weishan Wang
- The First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Yi Dai
- The First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Jing Li
- The First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Qi Li
- The First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Sibo Wang
- Xi’an Jiaotong University Affiliated Honghui Hospital, Xi’an, China
| | - Chenhui Shi
- The First Affiliated Hospital of Shihezi University, Shihezi, China
| |
Collapse
|
19
|
Wu D, Liu R, Cen X, Dong W, Chen Q, Lin J, Wang X, Ling Y, Mao R, Sun H, Huang R, Su H, Xu H, Qin D. Preclinical study of engineering MSCs promoting diabetic wound healing and other inflammatory diseases through M2 polarization. Stem Cell Res Ther 2025; 16:113. [PMID: 40038782 PMCID: PMC11881511 DOI: 10.1186/s13287-025-04248-y] [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: 08/29/2024] [Accepted: 02/20/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Diabetic foot ulcer (DFU) represents a common and severe complication of diabetes mellitus. Effective and safe treatments need to be developed. Mesenchymal stem cells (MSCs) have demonstrated crucial roles in tissue regeneration, wound repair and inflammation regulation. However, the function is limited. The safety and efficacy of gene-modified MSCs is unknown. Therefore, this study aimed to investigate whether genetically modified MSCs with highly efficient expression of anti-inflammatory factors promote diabetic wound repair by regulating macrophage phenotype transition. This may provide a new approach to treating diabetic wound healing. METHODS In this study, human umbilical cord-derived MSCs (hUMSCs) were genetically modified using recombinant lentiviral vectors to simultaneously overexpress three anti-inflammatory factors, interleukin (IL)-4, IL-10, IL-13 (MSCs-3IL). Cell counting kit-8, flow cytometry and differentiation assay were used to detect the criteria of MSCs. Overexpression efficiency was evaluated using flow cytometry, quantitative real-time PCR, Western blot, enzyme-linked immunosorbent assay, and cell scratch assay. We also assessed MSCs-3IL's ability to modulate Raw264.7 macrophage phenotype using flow cytometry and quantitative real-time PCR. In addition, we evaluated diabetic wound healing through healing rate calculation, HE staining, Masson staining, and immunohistochemical analysis of PCNA, F4/80, CD31, CD86, CD206, IL-4, IL-10 and IL-13. In addition, we evaluated the safety of the MSCs-3IL cells and the effect of the cells on several other models of inflammation. RESULTS MSCs-3IL efficiently expressed high levels of IL-4 and IL-10 (mRNA transcription increased by 15,000-fold and 800,000-fold, protein secretion 400 and 200 ng/mL), and IL-13 (mRNA transcription increased by 950,000-fold, protein secretion 6 ng/mL). MSCs-3IL effectively induced phenotypic polarization of pro-inflammatory M1-like macrophages (M1) towards anti-inflammatory M2-like macrophages (M2). The enhancement of function does not change the cell phenotype. The dynamic distribution in vivo was normal and no karyotype variation and tumor risk was observed. In a mouse diabetic wound model, MSCs-3IL promoted diabetic wound healing with a wound closure rate exceeding 96% after 14 days of cell treatment. The healing process was aided by altering macrophage phenotype (reduced CD86 and increased CD206 expression) and accelerating re-epithelialization. CONCLUSIONS In summary, our study demonstrates that genetically modified hUMSCs effectively overexpressed three key anti-inflammatory factors (IL-4, IL-10, IL-13). MSCs-3IL-based therapy enhances diabetic wound healing with high efficiency and safety. This suggests that genetically modified hUMSCs could be used as a novel therapeutic approach for DFU repair.
Collapse
Affiliation(s)
- Di Wu
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Rencun Liu
- Shandong Province Key Laboratory of Detection Technology for Tumour Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi, China
| | - Xiaotong Cen
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Wanwen Dong
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Qing Chen
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiali Lin
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Xia Wang
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yixia Ling
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Rui Mao
- Laboratory Animal Research Center, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Haitao Sun
- Neurosurgery Centre, Department of Cerebrovascular Surgery, Engineering Technology Research Centre of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, The National Key Clinical Specialty, Southern Medical University, Guangzhou, Guangdong, China
| | - Rui Huang
- The Fifth Clinical College, Guangzhou Medical University, Guangzhou, China
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
- , No.621 Gangwan Road, Huangpu District, Guangzhou, China.
| | - Hongjie Xu
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
- , No.621 Gangwan Road, Huangpu District, Guangzhou, China.
| | - Dajiang Qin
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China.
- , No.621 Gangwan Road, Huangpu District, Guangzhou, China.
| |
Collapse
|
20
|
Tchouto MN, Bucher CH, Mess AK, Haas S, Schmidt-Bleek K, Duda GN, Beule D, Milek M. Pronounced impairment of B cell differentiation during bone regeneration in adult immune experienced mice. Front Immunol 2025; 16:1511902. [PMID: 40098964 PMCID: PMC11911212 DOI: 10.3389/fimmu.2025.1511902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Alterations of the adaptive immune system have been shown to impact bone healing and may result in impaired healing in some patients. Apart from T cells, B cells are the key drivers of adaptive immunity. Therefore, their role in age-associated impairments of bone healing might be essential to understand delays during the healing process. B cells are essential for bone formation, and their dysfunction has been associated with aging or autoimmune diseases. But whether age-associated changes in B cell phenotypes are involved in bone regeneration is unknown. Methods Here, we aimed to characterize the role of immune aging in B cell phenotypes during the early inflammatory phase of bone healing. By comparing non-immune experienced with young and immune experienced mice we aimed to analyze the effect of gained immune experience on B cells. Our single cell proteo-genomics analysis quantified thousands of transcriptomes of cells that were isolated from post osteotomy hematoma and the proximal and distal bone marrow cavities, and enabled us to evaluate cell proportion, differential gene expression and cell trajectories. Results While the B cell proportion in young and non-immune experienced animals did not significantly change from 2 to 5 days post osteotomy in the hematoma, we found a significant decrease of the B cell proportion in the immune experienced mice, which was accompanied by the decreased expression of B cell specific genes, suggesting a specific response in immune experienced animals. Furthermore, we detected the most extensive B cell differentiation block in immune-experienced mice compared to non-immune experienced and young animals, predominantly in the transition from immature to mature B cells. Discussion Our results suggest that the pronounced impairment of B cell production found in immune experienced animals plays an important role in the initial phase leading to delayed bone healing. Therefore, novel therapeutic approaches may be able target the B cell differentiation defect to retain B cell functionality even in the immune experienced setting, which is prone to delayed healing.
Collapse
Affiliation(s)
- Mireille Ngokingha Tchouto
- Julius Wolff Institute of Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Core Unit Bioinformatics, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité – Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christian H. Bucher
- Julius Wolff Institute of Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ann-Kathrin Mess
- Julius Wolff Institute of Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité – Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Simon Haas
- Systems Hematology, Stem Cells & Precision Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute of Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Georg N. Duda
- Julius Wolff Institute of Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Miha Milek
- Core Unit Bioinformatics, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
21
|
Kong T, Seo SK, Han YS, Seo WM, Kim B, Kim J, Cho YJ, Lee S, Kang KS. Primed Mesenchymal Stem Cells by IFN-γ and IL-1β Ameliorate Acute Respiratory Distress Syndrome through Enhancing Homing Effect and Immunomodulation. Biomol Ther (Seoul) 2025; 33:311-324. [PMID: 39973472 PMCID: PMC11893491 DOI: 10.4062/biomolther.2025.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/21/2025] Open
Abstract
Acute Respiratory Distress Syndrome (ARDS) is a severe condition characterized by extensive lung inflammation and increased alveolar-capillary permeability, often triggered by infections or systemic inflammatory responses. Mesenchymal stem cells (MSCs)-based therapy holds promise for treating ARDS, as MSCs manifest immunomodulatory and regenerative properties that mitigate inflammation and enhance tissue repair. Primed MSCs, modified to augment specific functionalities, demonstrate superior therapeutic efficacy in targeted therapies compared to naive MSCs. This study explored the immunomodulatory potential of MSCs using mixed lymphocyte reaction (MLR) assays and co-culture experiments with M1/M2 macrophages. Additionally, RNA sequencing was employed to identify alterations in immune and inflammation-related factors in primed MSCs. The therapeutic effects of primed MSCs were assessed in an LPS-induced ARDS mouse model, and the underlying mechanisms were investigated through spatial transcriptomics analysis. The study revealed that MSCs primed with IFN-γ and IL-1β significantly enhanced the suppression of T cell activity compared to naive MSCs, concurrently inhibiting TNF-α while increasing IL-10 production in macrophages. Notably, combined treatment with these two cytokines resulted in a significant upregulation of immune and inflammation-regulating factors. Furthermore, our analyses elucidated the mechanisms behind the therapeutic effects of primed MSCs, including the inhibition of inflammatory cell infiltration in lung tissue, modulation of immune and inflammatory responses, and enhancement of elastin fiber formation. Signaling pathway analysis confirmed that efficacy could be enhanced by modulating NFκB and TNF-α signaling. In conclusion, in early-phase ARDS, primed MSCs displayed enhanced homing capabilities, improved lung function, and reduced inflammation.
Collapse
Affiliation(s)
- Taeho Kong
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Seoul 08590, Republic of Korea
| | - Su Kyoung Seo
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Seoul 08590, Republic of Korea
| | - Yong-Seok Han
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Seoul 08590, Republic of Korea
| | - Woo Min Seo
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Seoul 08590, Republic of Korea
| | - Bokyong Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| | - Jieun Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| | - Young-Jae Cho
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| | - Seunghee Lee
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Seoul 08590, Republic of Korea
| | - Kyung-Sun Kang
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Seoul 08590, Republic of Korea
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
22
|
Fan J, Xie Y, Liu D, Cui R, Zhang W, Shen M, Cao L. Crosstalk Between H-Type Vascular Endothelial Cells and Macrophages: A Potential Regulator of Bone Homeostasis. J Inflamm Res 2025; 18:2743-2765. [PMID: 40026304 PMCID: PMC11871946 DOI: 10.2147/jir.s502604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/30/2025] [Indexed: 03/05/2025] Open
Abstract
The crosstalk between H-type endothelial cells (ECs) and macrophages is critical for maintaining angiogenesis and osteogenesis in bone homeostasis. As core components of type H vessels, ECs respond to various pro-angiogenic signals, forming specialized vascular structures characterized by high expression of platelet-endothelial cell adhesion molecule-1 (CD31) and endothelial mucin (EMCN), thereby facilitating angiogenesis-osteogenesis coupling during bone formation. Macrophages, as key immune cells in the perivascular region, are primarily classified into the classically activated pro-inflammatory M1 phenotype and the selectively activated anti-inflammatory M2 phenotype, thereby performing dual functions in regulating local tissue homeostasis and innate immunity. In recent years, the complex crosstalk between type H vessel ECs and macrophages has garnered significant interest in the context of bone-related diseases. Orderly regulation of angiogenesis and bone immunity provides a new direction for preventing bone metabolic disorders such as osteoporosis and osteoarthritis. However, their interactions in bone homeostasis remain insufficiently understood, with limited clinical data available. This review comprehensively examines the intricate interactions between type H vessel ECs and macrophages with diverse phenotypes, and Insights into the signaling pathways that regulate their crosstalk, focusing on their roles in angiogenesis and osteogenesis. Furthermore, the review discusses recent interventions targeting this crosstalk and the challenges that remain. These insights may offer new perspectives on bone homeostasis and provide a theoretical foundation for developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Jiaxuan Fan
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Yaohui Xie
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Desun Liu
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Rui Cui
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Wei Zhang
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Mengying Shen
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Linzhong Cao
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| |
Collapse
|
23
|
Zhang D, Qu X, Chu F, Wang Z, Mou J, Yuan F. Regulation of macrophage-mediated osteogenesis by kaempferol liposomes in trauma-induced heterotopic ossification. Int J Pharm 2025; 671:125226. [PMID: 39842743 DOI: 10.1016/j.ijpharm.2025.125226] [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: 11/04/2024] [Revised: 01/08/2025] [Accepted: 01/13/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND Heterotopic ossification (HO) is characterized by abnormal bone formation outside the skeleton following injury or inherited disease, leading to limb dysfunction and neurological deficits. Current treatment options for HO are largely ineffective. METHODS A network pharmacological analysis was conducted to identify the active ingredients and protein targets in Astragalus and Cinnamon Twig Five-Substance Decoction (ACTFSD) on HO. Protein-protein interaction analysis and Gene Ontology Enrichment Analysis were used to investigate the key genes associated with the target proteins. Molecular docking was employed to validate the interactions between the core components and targets of ACTFSD. Kaempferol was encapsulated in liposomes synthesized via the thin-film dispersion method. In vitro and in vivo experiments were conducted to evaluate the therapeutic effects of kaempferol liposomes. RESULTS Kaempferol, an active ingredient in ACTFSD, effectively inhibits the PTGS2\ NF-κB pathway, regulates macrophage immune responses, and reduces cytokine released by macrophages induce abnormal osteogenesis in tendon stem cells (TDSCs), thereby slowing the formation of HO. Kaempferol liposomes demonstrate better therapeutic effect than kaempferol alone. CONCLUSION This study indicates that targeting macrophages may represent an effective therapeutic strategy for HO. Furthermore, kaempferol liposomes appear to be a promising treatment for trauma-induced HO. These findings provide potential new directions for future HO treatment strategies.
Collapse
Affiliation(s)
- Dazhen Zhang
- Key Laboratory of Bone Tissue Regeneration and Digital Medicine, Xuzhou Medical University, Xuzhou 221006 Jiangsu, China; Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006 Jiangsu, China; Xuzhou Medical University, Xuzhou 221006 Jiangsu, China
| | - Xinzhe Qu
- Key Laboratory of Bone Tissue Regeneration and Digital Medicine, Xuzhou Medical University, Xuzhou 221006 Jiangsu, China; Xuzhou Medical University, Xuzhou 221006 Jiangsu, China
| | - Fuchao Chu
- Key Laboratory of Bone Tissue Regeneration and Digital Medicine, Xuzhou Medical University, Xuzhou 221006 Jiangsu, China; Department of Orthopedics, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006 Jiangsu, China
| | - Zhenxin Wang
- Key Laboratory of Bone Tissue Regeneration and Digital Medicine, Xuzhou Medical University, Xuzhou 221006 Jiangsu, China
| | - Jie Mou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221006 Jiangsu, China.
| | - Feng Yuan
- Key Laboratory of Bone Tissue Regeneration and Digital Medicine, Xuzhou Medical University, Xuzhou 221006 Jiangsu, China; Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006 Jiangsu, China.
| |
Collapse
|
24
|
Xiang C, Cao J, Hu R, Li K, Meng T, Xia Y, Meng Q, Liu K, Liu L, Zhu X. Oleandrin inhibits osteoclast differentiation by targeting the LRP4/MAPK/NF-κB signalling pathway to treat osteoporosis. Int Immunopharmacol 2025; 148:114073. [PMID: 39823799 DOI: 10.1016/j.intimp.2025.114073] [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: 08/16/2024] [Revised: 12/04/2024] [Accepted: 01/08/2025] [Indexed: 01/20/2025]
Abstract
Osteoporosis is a common inflammation-related disease in which the release of proinflammatory cytokines promotes bone loss. Oleandrin is a monomer compound extracted from the leaves of the Nerium oleander plant, has been shown to exert an anti-inflammatory effect on a variety of inflammation-related diseases. However, its role in osteoporosis and the underlying mechanisms remain unclear. In this study, Oleandrin was shown to reduce bone loss in ovariectomy-induced osteoporotic mice in vivo. Additionally, Oleandrin inhibited RANKL-induced osteoclast differentiation in a concentration-dependent manner in vitro. Signalling pathway studies showed that Oleandrin could inhibit osteoclast differentiation by targeting MAPK and NF-κB signalling pathways. Further mechanistic studies showed that Oleandrin binds to low-density lipoprotein receptor-related protein 4 in osteoclast, thereby exerting inhibitory effects on osteoclast differentiation. In conclusion, this study lays the foundation for further research on the anti-inflammatory and anti-osteoporotic effects of Oleandrin on osteoporosis and its underlying mechanism and provides new possibilities for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Chongxin Xiang
- Department of Orthopedic, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Jiankang Cao
- Department of Pain, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Rui Hu
- Department of Orthopedic, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Kaixuan Li
- Department of Neurosurgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
| | - Tingyu Meng
- Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Yutao Xia
- Department of Oncology, YiDu Central Hospital of Weifang, Weifang, Shandong 262500, China
| | - Qunbo Meng
- Department of Orthopedic, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Kaiwen Liu
- Department of Orthopedic, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China.
| | - Lian Liu
- Department of Orthopedic, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China.
| | - Xuetao Zhu
- Department of Orthopedic, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Department of Emergency and Critical Care Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School of Nanjing Medical University, China.
| |
Collapse
|
25
|
Zhang D, Wang L, Tang L, Zhang Y, Zhang H, Zou L. Protective Effect of Methyl Sulfonyl Methane on the Progression of Age-Induced Bone Loss by Regulating Oxidative Stress-Mediated Bone Resorption. Antioxidants (Basel) 2025; 14:216. [PMID: 40002402 PMCID: PMC11851857 DOI: 10.3390/antiox14020216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Aging is associated with detrimental bone loss, often leading to fragility fractures, which may be driven by oxidative stress. In this study, the outcomes of comparing the differences among young, adult and aged C57BL/6J mice found that the trabecular bone volume was significantly lower in the aged mice compared to young mice, and the bone characteristics were significantly correlated with the oxidative status. To counteract the adverse effects of aging, methyl sulfonyl methane (MSM), a stable metabolite of dimethyl sulfoxide, was used to supplement the drinking water (400 mg/kg/day) of the aged mice (73 weeks old) for 8 weeks. The MSM supplementation improved the maximum load, bone microarchitecture, and mRNA levels of osteocyte-specific genes in the tibia. Furthermore, MSM reduced the serum level of the C-terminal telopeptide of type I collagen, a marker of bone resorption, and downregulated the mRNA levels of genes related to osteoclast proliferation and activity. MSM also decreased the levels of pro-inflammatory cytokines in both the serum and bone marrow. Importantly, the MSM-treated mice exhibited an enhanced antioxidant status, characterized by increased glutathione peroxidase (GPx) activity and glutathione concentration in plasma, erythrocytes and bone marrow. These improvements were linked to the activation of the nuclear factor E2 related factor 2 (Nrf2) pathway and its downstream antioxidant gene expression, including that of superoxide dismutase and GPx. These findings suggested that age-related bone loss is closely tied to oxidative stress, and MSM supplementation effectively reverses bone loss by mitigating oxidative stress-mediated bone resorption.
Collapse
Affiliation(s)
- Duo Zhang
- Physical Education Department, Civil Aviation Flight University of China, Guanghan 618307, China; (D.Z.); (L.T.); (Y.Z.)
| | - Leilei Wang
- Key Laboratory of Animal Biochemistry and Nutrition of Agriculture Ministry, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China;
| | - Lu Tang
- Physical Education Department, Civil Aviation Flight University of China, Guanghan 618307, China; (D.Z.); (L.T.); (Y.Z.)
| | - Yeting Zhang
- Physical Education Department, Civil Aviation Flight University of China, Guanghan 618307, China; (D.Z.); (L.T.); (Y.Z.)
| | - Huaiyong Zhang
- Key Laboratory of Animal Biochemistry and Nutrition of Agriculture Ministry, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China;
- Laboratory for Animal Nutrition and Animal Product Quality, Department of Animal Sciences and Aquatic Ecology, Ghent University, 9000 Ghent, Belgium
| | - Lin Zou
- Physical Education Department, Civil Aviation Flight University of China, Guanghan 618307, China; (D.Z.); (L.T.); (Y.Z.)
| |
Collapse
|
26
|
Shen X, Zhang H, Song Z, Dong Y, Ge X, Jin S, Guo S, Zhang P, Fu Y, Zhu Y, Xiao N, Wang D, Cheng J, Xu R, Jiang H. Enhancer-driven Shh signaling promotes glia-to-mesenchyme transition during bone repair. Bone Res 2025; 13:16. [PMID: 39865079 PMCID: PMC11770102 DOI: 10.1038/s41413-024-00396-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/19/2024] [Accepted: 12/01/2024] [Indexed: 01/28/2025] Open
Abstract
Plp1-lineage Schwann cells (SCs) of peripheral nerve play a critical role in vascular remodeling and osteogenic differentiation during the early stage of bone healing, and the abnormal plasticity of SCs would jeopardize the bone regeneration. However, how Plp1-lineage cells respond to injury and initiate the vascularized osteogenesis remains incompletely understood. Here, by employing single-cell transcriptional profiling combined with lineage-specific tracing models, we uncover that Plp1-lineage cells undergoing injury-induced glia-to-MSCs transition contributed to osteogenesis and revascularization in the initial stage of bone injury. Importantly, our data demonstrated that the Sonic hedgehog (Shh) signaling was responsible for the transition process initiation, which was strongly activated by c-Jun/SIRT6/BAF170 complex-driven Shh enhancers. Collectively, these findings depict an injury-specific niche signal-mediated Plp1-lineage cells transition towards Gli1+ MSCs and may be instructive for approaches to promote bone regeneration during aging or other bone diseases.
Collapse
Affiliation(s)
- Xin Shen
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, Jiangsu Province, China
| | - Hang Zhang
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, Jiangsu Province, China
| | - Zesheng Song
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, Jiangsu Province, China
| | - Yangjiele Dong
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, Jiangsu Province, China
| | - Xiao Ge
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, Jiangsu Province, China
| | - Shenghao Jin
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, Jiangsu Province, China
| | - Songsong Guo
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, Jiangsu Province, China
| | - Ping Zhang
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, Jiangsu Province, China
| | - Yu Fu
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, Jiangsu Province, China
| | - Yuchi Zhu
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, Jiangsu Province, China
| | - Na Xiao
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, Jiangsu Province, China
| | - Dongmiao Wang
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, Jiangsu Province, China
| | - Jie Cheng
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, Jiangsu Province, China
| | - Rongyao Xu
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, Jiangsu Province, China.
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, Jiangsu Province, China.
| | - Hongbing Jiang
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, Jiangsu Province, China.
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, Jiangsu Province, China.
| |
Collapse
|
27
|
Wang J, Zhang Y, Wang S, Wang X, Jing Y, Su J. Bone aging and extracellular vesicles. Sci Bull (Beijing) 2024; 69:3978-3999. [PMID: 39455324 DOI: 10.1016/j.scib.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/01/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024]
Abstract
Bone aging, a major global health concern, is the natural decline in bone mass and strength. Concurrently, extracellular vesicles (EVs), tiny membrane-bound particles produced by cells, have gained recognition for their roles in various physiological processes and age-related diseases. The interaction between EVs and bone aging is of growing interest, particularly their effects on bone metabolism, which become increasingly critical with advancing age. In this review, we explored the biology, types, and functions of EVs and emphasized their regulatory roles in bone aging. We examined the effects of EVs on bone metabolism and highlighted their potential as biomarkers for monitoring bone aging progression. Furthermore, we discussed the therapeutic applications of EVs, including targeted drug delivery and bone regeneration, and addressed the challenges associated with EV-based therapies, including the technical complexities and regulatory issues. We summarized the current research and clinical trials investigating the role of EVs in bone aging and suggested future research directions. These include the potential for personalized medicine using EVs and the integration of EV research with advanced technologies to enhance the management of age-related bone health. This analysis emphasized the transformative potential of EVs in understanding and managing bone aging, thereby marking a significant advancement in skeletal health research.
Collapse
Affiliation(s)
- Jian Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; School of Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China
| | - Yuanwei Zhang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China
| | - Sicheng Wang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China; Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai 200941, China
| | - Xinglong Wang
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721, USA.
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China.
| | - Jiacan Su
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
28
|
Li G, Wu J, Cheng X, Pei X, Wang J, Xie W. Nanoparticle-Mediated Gene Delivery for Bone Tissue Engineering. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2408350. [PMID: 39623813 DOI: 10.1002/smll.202408350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/13/2024] [Indexed: 03/17/2025]
Abstract
Critical-sized bone defects represent an urgent clinical problem, necessitating innovative treatment approaches. Gene-activated grafts for bone tissue engineering have emerged as a promising solution. However, traditional gene delivery methods are constrained by limited osteogenic efficacy and safety concerns. Recently, organic and inorganic nanoparticle (NP) vectors have attracted significant attention in bone tissue engineering for their safe, stable, and controllable gene delivery. Targeted gene delivery guided by insights into bone healing mechanisms, coupled with the multifunctional design of NPs, is crucial for enhancing therapeutic outcomes. Here, the theoretical foundations underlying NP-mediated gene therapy for enhancing bone healing across different histological stages are elucidated. Furthermore, the distinct attributes of functionalized NP vectors are discussed, and cutting-edge strategies aimed at optimizing gene delivery efficiency throughout the therapeutic process are highlighted. Additionally, the review addresses the unresolved challenges and prospects of this technology. This review may contribute to the continued development and clinical application of NP-mediated gene delivery for treating critical-sized bone defects.
Collapse
Affiliation(s)
- Guangzhao Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jiaxin Wu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xinting Cheng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Disease, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Xibo Pei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jian Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Wenjia Xie
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
29
|
Tyrina E, Yakubets D, Markina E, Buravkova L. Hippo Signaling Pathway Involvement in Osteopotential Regulation of Murine Bone Marrow Cells Under Simulated Microgravity. Cells 2024; 13:1921. [PMID: 39594669 PMCID: PMC11592674 DOI: 10.3390/cells13221921] [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: 10/14/2024] [Revised: 11/02/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
The development of osteopenia is one of the most noticeable manifestations of the adverse effects of space factors on crew members. The Hippo signaling pathway has been shown to play a central role in regulating the functional activity of cells through their response to mechanical stimuli. In the present study, the components of the Hippo pathway and the protective properties of osteodifferentiation inducers were investigated under simulated microgravity (smg) using a heterotypic bone marrow cell culture model, which allows for the maintenance of the close interaction between the stromal and hematopoietic compartments, present in vivo and of great importance for both the fate of osteoprogenitors and hematopoiesis. After 14 days of smg, the osteopotential and osteodifferentiation of bone marrow stromal progenitor cells, the expression of Hippo cascade genes and the immunocytochemical status of the adherent fraction of bone marrow cells, as well as the paracrine profile in the conditioned medium and the localization of Yap1 and Runx2 in mechanosensitive cells of the bone marrow were obtained. Simulated microgravity negatively affects stromal and hematopoietic cells when interacting in a heterotypic murine bone marrow cell culture. This is evidenced by the decrease in cell proliferation and osteopotential. Changes in the production of pleiotropic cytokines IL-6, GROβ and MCP-1 were revealed. Fourteen days of simulated microgravity induced a decrease in the nuclear translocation of Yap1 and the transcription factor Runx2 in the stromal cells of the intact group. Exposure to osteogenic induction conditions partially compensated for the negative effect of simulated microgravity. The data obtained will be crucial for understanding the effects of spaceflight on osteoprogenitor cell growth and differentiation via Hippo-Yap signaling.
Collapse
Affiliation(s)
- Ekaterina Tyrina
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia; (D.Y.); (L.B.)
| | | | - Elena Markina
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia; (D.Y.); (L.B.)
| | | |
Collapse
|
30
|
Bousch JF, Beyersdorf C, Schultz K, Windolf J, Suschek CV, Maus U. Proinflammatory Cytokines Enhance the Mineralization, Proliferation, and Metabolic Activity of Primary Human Osteoblast-like Cells. Int J Mol Sci 2024; 25:12358. [PMID: 39596421 PMCID: PMC11594863 DOI: 10.3390/ijms252212358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Osteoporosis is a progressive metabolic bone disease characterized by decreased bone density and microarchitectural deterioration, leading to an increased risk of fracture, particularly in postmenopausal women and the elderly. Increasing evidence suggests that inflammatory processes play a key role in the pathogenesis of osteoporosis and are strongly associated with the activation of osteoclasts, the cells responsible for bone resorption. In the present study, we investigated, for the first time, the influence of proinflammatory cytokines on the osteogenic differentiation, proliferation, and metabolic activity of primary human osteoblast-like cells (OBs) derived from the femoral heads of elderly patients. We found that all the proinflammatory cytokines, IL-1β, TNF-α, IL-6, and IL-8, enhanced the extracellular matrix mineralization of OBs under differentiation-induced cell culture conditions. In the cases of IL-1β and TNF-α, increased mineralization was correlated with increased osteoblast proliferation. Additionally, IL-1β- and TNF-α-increased osteogenesis was accompanied by a rise in energy metabolism due to improved glycolysis or mitochondrial respiration. In conclusion, we show here, for the first time, that, in contrast to findings obtained with cell lines, mesenchymal stem cells, or animal models, human OBs obtained from patients exhibited significantly enhanced osteogenesis upon exposure to proinflammatory cytokines, probably in part via a mechanism involving enhanced cellular energy metabolism. This study significantly contributes to the field of osteoimmunology by examining a clinically relevant cell model that can help to develop treatments for inflammation-related metabolic bone diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Uwe Maus
- Department for Orthopedics and Trauma Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Dusseldorf, Germany; (J.F.B.); (C.B.); (K.S.); (J.W.); (C.V.S.)
| |
Collapse
|
31
|
Menger MM, Speicher R, Hans S, Histing T, El Kayali MKD, Ehnert S, Menger MD, Ampofo E, Wrublewsky S, Laschke MW. Nlrp3 Deficiency Does Not Substantially Affect Femoral Fracture Healing in Mice. Int J Mol Sci 2024; 25:11788. [PMID: 39519338 PMCID: PMC11546854 DOI: 10.3390/ijms252111788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/20/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Inflammation has been recognized as major factor for successful bone regeneration. On the other hand, a prolonged or overshooting inflammatory response can also cause fracture healing failure. The nucleotide-binding oligomerization domain (NOD)-like receptor protein (NLRP)3 inflammasome plays a crucial role in inflammatory cytokine production. However, its role during fracture repair remains elusive. We investigated the effects of Nlrp3 deficiency on the healing of closed femoral fractures in Nlrp3-/- and wildtype mice. The callus tissue was analyzed by means of X-ray, biomechanics, µCT and histology, as well as immunohistochemistry and Western blotting at 2 and 5 weeks after surgery. We found a significantly reduced trabecular thickness at 2 weeks after fracture in the Nlrp3-/- mice when compared to the wildtype animals. However, the amount of bone tissue did not differ between the two groups. Additional immunohistochemical analyses showed a reduced number of CD68-positive macrophages within the callus tissue of the Nlrp3-/- mice at 2 weeks after fracture, whereas the number of myeloperoxidase (MPO)-positive granulocytes was increased. Moreover, we detected a significantly lower expression of vascular endothelial growth factor (VEGF) and a reduced number of microvessels in the Nlrp3-/- mice. The expression of the absent in melanoma (AIM)2 inflammasome was increased more than 2-fold in the Nlrp3-/- mice, whereas the expression of the pro-inflammatory cytokines interleukin (IL)-1β and IL-18 was not affected. Our results demonstrate that Nlrp3 deficiency does not markedly affect femoral fracture healing in mice. This is most likely due to the unaltered expression of pro-inflammatory cytokines and pro-osteogenic growth factors.
Collapse
Affiliation(s)
- Maximilian M. Menger
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany; (T.H.)
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (R.S.); (S.H.); (M.K.D.E.K.); (M.D.M.); (E.A.); (S.W.); (M.W.L.)
| | - Rouven Speicher
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (R.S.); (S.H.); (M.K.D.E.K.); (M.D.M.); (E.A.); (S.W.); (M.W.L.)
| | - Sandra Hans
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (R.S.); (S.H.); (M.K.D.E.K.); (M.D.M.); (E.A.); (S.W.); (M.W.L.)
| | - Tina Histing
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany; (T.H.)
| | - Moses K. D. El Kayali
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (R.S.); (S.H.); (M.K.D.E.K.); (M.D.M.); (E.A.); (S.W.); (M.W.L.)
| | - Sabrina Ehnert
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany;
| | - Michael D. Menger
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (R.S.); (S.H.); (M.K.D.E.K.); (M.D.M.); (E.A.); (S.W.); (M.W.L.)
| | - Emmanuel Ampofo
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (R.S.); (S.H.); (M.K.D.E.K.); (M.D.M.); (E.A.); (S.W.); (M.W.L.)
| | - Selina Wrublewsky
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (R.S.); (S.H.); (M.K.D.E.K.); (M.D.M.); (E.A.); (S.W.); (M.W.L.)
| | - Matthias W. Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (R.S.); (S.H.); (M.K.D.E.K.); (M.D.M.); (E.A.); (S.W.); (M.W.L.)
| |
Collapse
|
32
|
Chow SKH, Gao Q, Pius A, Morita M, Ergul Y, Murayama M, Shinohara I, Cekuc MS, Ma C, Susuki Y, Goodman SB. The Advantages and Shortcomings of Stem Cell Therapy for Enhanced Bone Healing. Tissue Eng Part C Methods 2024; 30:415-430. [PMID: 39311464 DOI: 10.1089/ten.tec.2024.0252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024] Open
Abstract
This review explores the regenerative potential of key progenitor cell types and therapeutic strategies to improve healing of complex fractures and bone defects. We define, summarize, and discuss the differentiation potential of totipotent, pluripotent, and multipotent stem cells, emphasizing the advantages and shortcomings of cell therapy for bone repair and regeneration. The fundamental role of mesenchymal stem cells is highlighted due to their multipotency to differentiate into the key lineage cells including osteoblasts, osteocytes, and chondrocytes, which are crucial for bone formation and remodeling. Hematopoietic stem cells (HSCs) also play a significant role; immune cells such as macrophages and T-cells modulate inflammation and tissue repair. Osteoclasts are multinucleated cells that are important to bone remodeling. Vascular progenitor (VP) cells are critical to oxygen and nutrient supply. The dynamic interplay among these lineages and their microenvironment is essential for effective bone restoration. Therapies involving cells that are more than "minimally manipulated" are controversial and include embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). ESCs, derived from early-stage embryos, possess pluripotent capabilities and have shown promise in preclinical studies for bone healing. iPSCs, reprogrammed from somatic cells, offer personalized medicine applications and can differentiate into various tissue-specific cell lines. Minimally manipulative cell therapy approaches such as the use of bone marrow aspirate concentrate (BMAC), exosomes, and various biomaterials for local delivery are explored for their effectiveness in bone regeneration. BMAC, which contains mostly immune cells but few mesenchymal and VPs, probably improves bone healing by facilitating paracrine-mediated intercellular communication. Exosome isolation harnesses the biological signals and cellular by-products that are a primary source for cell crosstalk and activation. Safe, efficacious, and cost-effective strategies to enhance bone healing using novel cellular therapies are part of a changing paradigm to modulate the inflammatory, repair, and regenerative pathways to achieve earlier more robust tissue healing and improved physical function. Impact Statement Stem cell therapy holds immense potential for bone healing due to its ability to regenerate damaged tissue. Nonmanipulated bone marrow aspirate contains mesenchymal stem cells that promote bone repair and reduce healing time. Induced pluripotent stem cells offer the advantage of creating patient-specific cells that can differentiate into osteoblasts, aiding in bone regeneration. Other delivery methods, such as scaffold-based techniques, enhance stem cell integration and function. Collectively, these approaches can improve treatment outcomes, reduce recovery periods, and advance our understanding of bone healing mechanisms, making them pivotal in orthopedic research and regenerative medicine.
Collapse
Affiliation(s)
- Simon Kwoon-Ho Chow
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Alexa Pius
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Mayu Morita
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Yasemin Ergul
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Masatoshi Murayama
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Issei Shinohara
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Mehmet Sertac Cekuc
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Chao Ma
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Yosuke Susuki
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
- Department of Bioengineering, Stanford University, Stanford, California, USA
| |
Collapse
|
33
|
Ge R, Liu C, Zhao Y, Wang K, Wang X. Endochondral Ossification for Spinal Fusion: A Novel Perspective from Biological Mechanisms to Clinical Applications. J Pers Med 2024; 14:957. [PMID: 39338212 PMCID: PMC11433020 DOI: 10.3390/jpm14090957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/07/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Degenerative scoliosis (DS), encompassing conditions like spondylolisthesis and spinal stenosis, is a common type of spinal deformity. Lumbar interbody fusion (LIF) stands as a conventional surgical intervention for this ailment, aiming at decompression, restoration of intervertebral height, and stabilization of motion segments. Despite its widespread use, the precise mechanism underlying spinal fusion remains elusive. In this review, our focus lies on endochondral ossification for spinal fusion, a process involving vertebral development and bone healing. Endochondral ossification is the key step for the successful vertebral fusion. Endochondral ossification can persist in hypoxic conditions and promote the parallel development of angiogenesis and osteogenesis, which corresponds to the fusion process of new bone formation in the hypoxic region between the vertebrae. The ideal material for interbody fusion cages should have the following characteristics: (1) Good biocompatibility; (2) Stable chemical properties; (3) Biomechanical properties similar to bone tissue; (4) Promotion of bone fusion; (5) Favorable for imaging observation; (6) Biodegradability. Utilizing cartilage-derived bone-like constructs holds promise in promoting bony fusion post-operation, thus warranting exploration in the context of spinal fusion procedures.
Collapse
Affiliation(s)
- Rile Ge
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, No. 95, Yong An Rd, Beijing 100050, China;
| | - Chenjun Liu
- Department of Spinal Surgery, Peking University People’s Hospital, 11th Xizhimen South Ave., Beijing 100044, China;
| | - Yuhong Zhao
- Beijing Key Laboratory of Lignocellulosic Chemistry, Beijing Forestry University, Beijing 100083, China;
| | - Kaifeng Wang
- Department of Spinal Surgery, Peking University People’s Hospital, 11th Xizhimen South Ave., Beijing 100044, China;
| | - Xiluan Wang
- Beijing Key Laboratory of Lignocellulosic Chemistry, Beijing Forestry University, Beijing 100083, China;
| |
Collapse
|
34
|
Murayama M, Chow SK, Lee ML, Young B, Ergul YS, Shinohara I, Susuki Y, Toya M, Gao Q, Goodman SB. The interactions of macrophages, lymphocytes, and mesenchymal stem cells during bone regeneration. Bone Joint Res 2024; 13:462-473. [PMID: 39237112 PMCID: PMC11377107 DOI: 10.1302/2046-3758.139.bjr-2024-0122.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/07/2024] Open
Abstract
Bone regeneration and repair are crucial to ambulation and quality of life. Factors such as poor general health, serious medical comorbidities, chronic inflammation, and ageing can lead to delayed healing and nonunion of fractures, and persistent bone defects. Bioengineering strategies to heal bone often involve grafting of autologous bone marrow aspirate concentrate (BMAC) or mesenchymal stem cells (MSCs) with biocompatible scaffolds. While BMAC shows promise, variability in its efficacy exists due to discrepancies in MSC concentration and robustness, and immune cell composition. Understanding the mechanisms by which macrophages and lymphocytes - the main cellular components in BMAC - interact with MSCs could suggest novel strategies to enhance bone healing. Macrophages are polarized into pro-inflammatory (M1) or anti-inflammatory (M2) phenotypes, and influence cell metabolism and tissue regeneration via the secretion of cytokines and other factors. T cells, especially helper T1 (Th1) and Th17, promote inflammation and osteoclastogenesis, whereas Th2 and regulatory T (Treg) cells have anti-inflammatory pro-reconstructive effects, thereby supporting osteogenesis. Crosstalk among macrophages, T cells, and MSCs affects the bone microenvironment and regulates the local immune response. Manipulating the proportion and interactions of these cells presents an opportunity to alter the local regenerative capacity of bone, which potentially could enhance clinical outcomes.
Collapse
Affiliation(s)
- Masatoshi Murayama
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Simon K. Chow
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Max L. Lee
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Bill Young
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Yasemin S. Ergul
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Issei Shinohara
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Yosuke Susuki
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Masakazu Toya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Bioengineering, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
35
|
Kang S, Woo Y, Seo Y, Yoo D, Kwon D, Park H, Lee SD, Yoo HY, Lee T. A Descriptive Review on the Potential Use of Diatom Biosilica as a Powerful Functional Biomaterial: A Natural Drug Delivery System. Pharmaceutics 2024; 16:1171. [PMID: 39339207 PMCID: PMC11434644 DOI: 10.3390/pharmaceutics16091171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Although various chemically synthesized materials are essential in medicine, food, and agriculture, they can exert unexpected side effects on the environment and human health by releasing certain toxic chemicals. Therefore, eco-friendly and biocompatible biomaterials based on natural resources are being actively explored. Recently, biosilica derived from diatoms has attracted attention in various biomedical fields, including drug delivery systems (DDS), due to its uniform porous nano-pattern, hierarchical structure, and abundant silanol functional groups. Importantly, the structural characteristics of diatom biosilica improve the solubility of poorly soluble substances and enable sustained release of loaded drugs. Additionally, diatom biosilica predominantly comprises SiO2, has high biocompatibility, and can easily hybridize with other DDS platforms, including hydrogels and cationic DDS, owing to its strong negative charge and abundant silanol groups. This review explores the potential applications of various diatom biosilica-based DDS in various biomedical fields, with a particular focus on hybrid DDS utilizing them.
Collapse
Affiliation(s)
- Sunggu Kang
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon-gu, Seoul 01897, Republic of Korea
| | - Yeeun Woo
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon-gu, Seoul 01897, Republic of Korea
| | - Yoseph Seo
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon-gu, Seoul 01897, Republic of Korea
| | - Daehyeon Yoo
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon-gu, Seoul 01897, Republic of Korea
| | - Daeryul Kwon
- Protist Research Division, Biological Resources Research Department, Nakdonggang National Institute of Biological Resources (NNIBR), 137, Donam 2-gil, Sangju-si 37242, Gyeongsangbuk-do, Republic of Korea
| | - Hyunjun Park
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon-gu, Seoul 01897, Republic of Korea
| | - Sang Deuk Lee
- Protist Research Division, Biological Resources Research Department, Nakdonggang National Institute of Biological Resources (NNIBR), 137, Donam 2-gil, Sangju-si 37242, Gyeongsangbuk-do, Republic of Korea
| | - Hah Young Yoo
- Department of Biotechnology, Sangmyung University, 20, Hongjimun 2-gil, Jongno-gu, Seoul 03016, Republic of Korea
| | - Taek Lee
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon-gu, Seoul 01897, Republic of Korea
| |
Collapse
|
36
|
Farazin A, Mahjoubi S. Dual-functional Hydroxyapatite scaffolds for bone regeneration and precision drug delivery. J Mech Behav Biomed Mater 2024; 157:106661. [PMID: 39018918 DOI: 10.1016/j.jmbbm.2024.106661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/25/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
Addressing infected bone defects remains a significant challenge in orthopedics, requiring effective infection control and bone defect repair. A promising therapeutic approach involves the development of dual-functional engineered biomaterials with drug delivery systems that combine antibacterial properties with osteogenesis promotion. The Hydroxyapatite composite scaffolds offer a one-stage treatment, eliminating the need for multiple surgeries and thereby streamlining the process and reducing treatment time. This review delves into the impaired bone repair mechanisms within pathogen-infected and inflamed microenvironments, providing a theoretical foundation for treating infectious bone defects. Additionally, it explores composite scaffolds made of antibacterial and osteogenic materials, along with advanced drug delivery systems that possess both antibacterial and bone-regenerative properties. By offering a comprehensive understanding of the microenvironment of infectious bone defects and innovative design strategies for dual-function scaffolds, this review presents significant advancements in treatment methods for infectious bone defects. Continued research and clinical validation are essential to refine these innovations, ensuring biocompatibility and safety, achieving controlled release and stability, and developing scalable manufacturing processes for widespread clinical application.
Collapse
Affiliation(s)
- Ashkan Farazin
- Department of Mechanical Engineering, Stevens Institute of Technology, Castle Point on Hudson, Hoboken, NJ, 07030, United States
| | - Soroush Mahjoubi
- Department of Civil and Environmental Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, United States; Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States.
| |
Collapse
|
37
|
Ding Z, Ma G, Zhou B, Cheng S, Tang W, Han Y, Chen L, Pang W, Chen Y, Yang D, Cao H. Targeting miR-29 mitigates skeletal senescence and bolsters therapeutic potential of mesenchymal stromal cells. Cell Rep Med 2024; 5:101665. [PMID: 39168101 PMCID: PMC11384963 DOI: 10.1016/j.xcrm.2024.101665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/07/2024] [Accepted: 07/08/2024] [Indexed: 08/23/2024]
Abstract
Mesenchymal stromal cell (MSC) senescence is a key factor in skeletal aging, affecting the potential of MSC applications. Identifying targets to prevent MSC and skeletal senescence is crucial. Here, we report increased miR-29 expression in bone tissues of aged mice, osteoporotic patients, and senescent MSCs. Genetic overexpression of miR-29 in Prx1-positive MSCs significantly accelerates skeletal senescence, reducing cortical bone thickness and trabecular bone mass, while increasing femur cross-sectional area, bone marrow adiposity, p53, and senescence-associated secretory phenotype (SASP) levels. Mechanistically, miR-29 promotes senescence by upregulating p53 via targeting Kindlin-2 mRNA. miR-29 knockdown in BMSCs impedes skeletal senescence, enhances bone mass, and accelerates calvarial defect regeneration, also reducing lipopolysaccharide (LPS)-induced organ injuries and mortality. Thus, our findings underscore miR-29 as a promising therapeutic target for senescence-related skeletal diseases and acute inflammation-induced organ damage.
Collapse
Affiliation(s)
- Zhen Ding
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Guixing Ma
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Bo Zhou
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Siyuan Cheng
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Wanze Tang
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yingying Han
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Litong Chen
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Wei Pang
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yangshan Chen
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Dazhi Yang
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Huiling Cao
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
38
|
Li N, Wang J, Feng G, Liu Y, Shi Y, Wang Y, Chen L. Advances in biomaterials for oral-maxillofacial bone regeneration: spotlight on periodontal and alveolar bone strategies. Regen Biomater 2024; 11:rbae078. [PMID: 39055303 PMCID: PMC11272181 DOI: 10.1093/rb/rbae078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 06/05/2024] [Accepted: 06/16/2024] [Indexed: 07/27/2024] Open
Abstract
The intricate nature of oral-maxillofacial structure and function, coupled with the dynamic oral bacterial environment, presents formidable obstacles in addressing the repair and regeneration of oral-maxillofacial bone defects. Numerous characteristics should be noticed in oral-maxillofacial bone repair, such as irregular morphology of bone defects, homeostasis between hosts and microorganisms in the oral cavity and complex periodontal structures that facilitate epithelial ingrowth. Therefore, oral-maxillofacial bone repair necessitates restoration materials that adhere to stringent and specific demands. This review starts with exploring these particular requirements by introducing the particular characteristics of oral-maxillofacial bones and then summarizes the classifications of current bone repair materials in respect of composition and structure. Additionally, we discuss the modifications in current bone repair materials including improving mechanical properties, optimizing surface topography and pore structure and adding bioactive components such as elements, compounds, cells and their derivatives. Ultimately, we organize a range of potential optimization strategies and future perspectives for enhancing oral-maxillofacial bone repair materials, including physical environment manipulation, oral microbial homeostasis modulation, osteo-immune regulation, smart stimuli-responsive strategies and multifaceted approach for poly-pathic treatment, in the hope of providing some insights for researchers in this field. In summary, this review analyzes the complex demands of oral-maxillofacial bone repair, especially for periodontal and alveolar bone, concludes multifaceted strategies for corresponding biomaterials and aims to inspire future research in the pursuit of more effective treatment outcomes.
Collapse
Affiliation(s)
- Nayun Li
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Union Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Oral and Maxillofacial Medical Devices and Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jinyu Wang
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Union Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Oral and Maxillofacial Medical Devices and Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guangxia Feng
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Union Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Oral and Maxillofacial Medical Devices and Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuqing Liu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Union Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Oral and Maxillofacial Medical Devices and Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yunsong Shi
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Union Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Oral and Maxillofacial Medical Devices and Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yifan Wang
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Union Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Oral and Maxillofacial Medical Devices and Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Union Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Oral and Maxillofacial Medical Devices and Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
39
|
Yang X, Zhang S, Lu J, Chen X, Zheng T, He R, Ye C, Xu J. Therapeutic potential of mesenchymal stem cell-derived exosomes in skeletal diseases. Front Mol Biosci 2024; 11:1268019. [PMID: 38903180 PMCID: PMC11187108 DOI: 10.3389/fmolb.2024.1268019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 05/16/2024] [Indexed: 06/22/2024] Open
Abstract
Skeletal diseases impose a considerable burden on society. The clinical and tissue-engineering therapies applied to alleviate such diseases frequently result in complications and are inadequately effective. Research has shifted from conventional therapies based on mesenchymal stem cells (MSCs) to exosomes derived from MSCs. Exosomes are natural nanocarriers of endogenous DNA, RNA, proteins, and lipids and have a low immune clearance rate and good barrier penetration and allow targeted delivery of therapeutics. MSC-derived exosomes (MSC-exosomes) have the characteristics of both MSCs and exosomes, and so they can have both immunosuppressive and tissue-regenerative effects. Despite advances in our knowledge of MSC-exosomes, their regulatory mechanisms and functionalities are unclear. Here we review the therapeutic potential of MSC-exosomes for skeletal diseases.
Collapse
Affiliation(s)
- Xiaobo Yang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Shaodian Zhang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Jinwei Lu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Xiaoling Chen
- Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Tian Zheng
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Rongxin He
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Chenyi Ye
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Jianbin Xu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| |
Collapse
|
40
|
Moghaddam A, Bahrami M, Mirzadeh M, Khatami M, Simorgh S, Chimehrad M, Kruppke B, Bagher Z, Mehrabani D, Khonakdar HA. Recent trends in bone tissue engineering: a review of materials, methods, and structures. Biomed Mater 2024; 19:042007. [PMID: 38636500 DOI: 10.1088/1748-605x/ad407d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 04/18/2024] [Indexed: 04/20/2024]
Abstract
Bone tissue engineering (BTE) provides the treatment possibility for segmental long bone defects that are currently an orthopedic dilemma. This review explains different strategies, from biological, material, and preparation points of view, such as using different stem cells, ceramics, and metals, and their corresponding properties for BTE applications. In addition, factors such as porosity, surface chemistry, hydrophilicity and degradation behavior that affect scaffold success are introduced. Besides, the most widely used production methods that result in porous materials are discussed. Gene delivery and secretome-based therapies are also introduced as a new generation of therapies. This review outlines the positive results and important limitations remaining in the clinical application of novel BTE materials and methods for segmental defects.
Collapse
Affiliation(s)
| | - Mehran Bahrami
- Department of Mechanical Engineering and Mechanics, Lehigh University, 27 Memorial Dr W, Bethlehem, PA 18015, United States of America
| | | | - Mehrdad Khatami
- Iran Polymer and Petrochemical Institute (IPPI), Tehran 14965-115, Iran
| | - Sara Simorgh
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Chimehrad
- Department of Mechanical & Aerospace Engineering, College of Engineering & Computer Science, University of Central Florida, Orlando, FL, United States of America
| | - Benjamin Kruppke
- Max Bergmann Center of Biomaterials and Institute of Materials Science, Technische Universität Dresden, 01069 Dresden, Germany
| | - Zohreh Bagher
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Davood Mehrabani
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Fars 71348-14336, Iran
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Fars 71345-1744, Iran
| | - Hossein Ali Khonakdar
- Iran Polymer and Petrochemical Institute (IPPI), Tehran 14965-115, Iran
- Max Bergmann Center of Biomaterials and Institute of Materials Science, Technische Universität Dresden, 01069 Dresden, Germany
| |
Collapse
|
41
|
Nie R, Zhang QY, Feng ZY, Huang K, Zou CY, Fan MH, Zhang YQ, Zhang JY, Li-Ling J, Tan B, Xie HQ. Hydrogel-based immunoregulation of macrophages for tissue repair and regeneration. Int J Biol Macromol 2024; 268:131643. [PMID: 38643918 DOI: 10.1016/j.ijbiomac.2024.131643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/10/2024] [Accepted: 04/14/2024] [Indexed: 04/23/2024]
Abstract
The rational design of hydrogel materials to modulate the immune microenvironment has emerged as a pivotal approach in expediting tissue repair and regeneration. Within the immune microenvironment, an array of immune cells exists, with macrophages gaining prominence in the field of tissue repair and regeneration due to their roles in cytokine regulation to promote regeneration, maintain tissue homeostasis, and facilitate repair. Macrophages can be categorized into two types: classically activated M1 (pro-inflammatory) and alternatively activated M2 (anti-inflammatory and pro-repair). By regulating the physical and chemical properties of hydrogels, the phenotypic transformation and cell behavior of macrophages can be effectively controlled, thereby promoting tissue regeneration and repair. A full understanding of the interaction between hydrogels and macrophages can provide new ideas and methods for future tissue engineering and clinical treatment. Therefore, this paper reviews the effects of hydrogel components, hardness, pore size, and surface morphology on cell behaviors such as macrophage proliferation, migration, and phenotypic polarization, and explores the application of hydrogels based on macrophage immune regulation in skin, bone, cartilage, and nerve tissue repair. Finally, the challenges and future prospects of macrophage-based immunomodulatory hydrogels are discussed.
Collapse
Affiliation(s)
- Rong Nie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Qing-Yi Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Zi-Yuan Feng
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Kai Huang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Chen-Yu Zou
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ming-Hui Fan
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yue-Qi Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ji-Ye Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jesse Li-Ling
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Medical Genetics, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Bo Tan
- Department of Orthopedic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, PR China
| | - Hui-Qi Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan 610212, PR China.
| |
Collapse
|
42
|
Li Z, Wang B, Wang R, Zhang Z, Xiong J, Wang X, Ma Y, Han L. Identification of PKM2 as a pyroptosis-related key gene aggravates senile osteoporosis via the NLRP3/Caspase-1/GSDMD signaling pathway. Int J Biochem Cell Biol 2024; 169:106537. [PMID: 38342404 DOI: 10.1016/j.biocel.2024.106537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/16/2023] [Accepted: 01/23/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUNDS Senile osteoporosis-alternatively labeled as skeletal aging-encompasses age-induced bone deterioration and loss of bone microarchitecture. Recent studies have indicated a potential association between senile osteoporosis and chronic systemic inflammation, and pyroptosis in bone marrow-derived mesenchymal stem cells is speculated to contribute to bone loss and osteoporosis. Therefore, targeting pyroptosis in stem cells may be a potential therapeutic strategy for treating osteoporosis. METHODS Initially, we conducted bioinformatics analysis to screen the GEO databases to identify the key gene associated with pyroptosis in senile osteoporosis. Next, we analyzed the relationship between altered proteins and clinical data. In vitro experiments were then performed to explore whether the downregulation of PKM2 expression could inhibit pyroptosis. Additionally, an aging-related mouse model of osteoporosis was established to validate the efficacy of a PKM2 inhibitor in alleviating osteoporosis progression. RESULTS We identified PKM2 as a key gene implicated in pyroptosis in senile osteoporosis patients through bioinformatics analysis. Further analyses of bone marrow and stem cells demonstrated significant PKM2 overexpression in senile osteoporosis patients. Silencing PKM2 expression inhibited pyroptosis in senile stem cells, of which the osteogenesis potential and angiogenic function were also primarily promoted. Moreover, the results in vivo demonstrated that administering PKM2 inhibitors suppressed pyroptosis in senile osteoporosis mice and mitigated senile osteoporosis progression. CONCLUSION Our study uncovered PKM2, a key pyroptosis marker of bone marrow mesenchymal stem cells in senile osteoporosis. Shikonin, a PKM2 inhibitor, was then identified as a potential drug candidate for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Zilin Li
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Wang
- Department of Rehabilitation, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruoyu Wang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhichao Zhang
- Department of Rehabilitation, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Xiong
- Department of Rehabilitation, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyun Wang
- Department of Rehabilitation, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Ma
- Department of Rehabilitation, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Lizhi Han
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical College, Anhui Key Laboratory of Tissue Transformation, Bengbu Medical College, Bengbu 233000, Anhui Province, China.
| |
Collapse
|
43
|
Chen P, Wang Y, Zhou B. Insights into targeting cellular senescence with senolytic therapy: The journey from preclinical trials to clinical practice. Mech Ageing Dev 2024; 218:111918. [PMID: 38401690 DOI: 10.1016/j.mad.2024.111918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 02/26/2024]
Abstract
Interconnected, fundamental aging processes are central to many illnesses and diseases. Cellular senescence is a mechanism that halts the cell cycle in response to harmful stimuli. Senescent cells (SnCs) can emerge at any point in life, and their persistence, along with the numerous proteins they secrete, can negatively affect tissue function. Interventions aimed at combating persistent SnCs, which can destroy tissues, have been used in preclinical models to delay, halt, or even reverse various diseases. Consequently, the development of small-molecule senolytic medicines designed to specifically eliminate SnCs has opened potential avenues for the prevention or treatment of multiple diseases and age-related issues in humans. In this review, we explore the most promising approaches for translating small-molecule senolytics and other interventions targeting senescence in clinical practice. This discussion highlights the rationale for considering SnCs as therapeutic targets for diseases affecting individuals of all ages.
Collapse
Affiliation(s)
- Peng Chen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China.
| | - Yulai Wang
- Department of Pharmacy, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, Hubei, P.R. China
| | - Benhong Zhou
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| |
Collapse
|
44
|
Dong Q, Fei X, Zhang H, Zhu X, Ruan J. Effect of Dimethyloxalylglycine on Stem Cells Osteogenic Differentiation and Bone Tissue Regeneration-A Systematic Review. Int J Mol Sci 2024; 25:3879. [PMID: 38612687 PMCID: PMC11011423 DOI: 10.3390/ijms25073879] [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: 02/20/2024] [Revised: 03/20/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Dimethyloxalylglycine (DMOG) has been found to stimulate osteogenesis and angiogenesis of stem cells, promoting neo-angiogenesis in bone tissue regeneration. In this review, we conducted a comprehensive search of the literature to investigate the effects of DMOG on osteogenesis and bone regeneration. We screened the studies based on specific inclusion criteria and extracted relevant information from both in vitro and in vivo experiments. The risk of bias in animal studies was evaluated using the SYRCLE tool. Out of the 174 studies retrieved, 34 studies met the inclusion criteria (34 studies were analyzed in vitro and 20 studies were analyzed in vivo). The findings of the included studies revealed that DMOG stimulated stem cells' differentiation toward osteogenic, angiogenic, and chondrogenic lineages, leading to vascularized bone and cartilage regeneration. Addtionally, DMOG demonstrated therapeutic effects on bone loss caused by bone-related diseases. However, the culture environment in vitro is notably distinct from that in vivo, and the animal models used in vivo experiments differ significantly from humans. In summary, DMOG has the ability to enhance the osteogenic and angiogenic differentiation potential of stem cells, thereby improving bone regeneration in cases of bone defects. This highlights DMOG as a potential focus for research in the field of bone tissue regeneration engineering.
Collapse
Affiliation(s)
- Qiannan Dong
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710000, China
- Center of Oral Public Health, College of Stomatology, Xi’an Jiaotong University, Xi’an 710000, China
| | - Xiuzhi Fei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710000, China
- Center of Oral Public Health, College of Stomatology, Xi’an Jiaotong University, Xi’an 710000, China
| | - Hengwei Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710000, China
- Center of Oral Public Health, College of Stomatology, Xi’an Jiaotong University, Xi’an 710000, China
| | - Ximei Zhu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710000, China
- Center of Oral Public Health, College of Stomatology, Xi’an Jiaotong University, Xi’an 710000, China
| | - Jianping Ruan
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710000, China
- Center of Oral Public Health, College of Stomatology, Xi’an Jiaotong University, Xi’an 710000, China
| |
Collapse
|
45
|
Yang T, Liu S, Ma H, Lai H, Wang C, Ni K, Lu Y, Li W, Hu X, Zhou Z, Lou C, He D. Carnitine functions as an enhancer of NRF2 to inhibit osteoclastogenesis via regulating macrophage polarization in osteoporosis. Free Radic Biol Med 2024; 213:174-189. [PMID: 38246515 DOI: 10.1016/j.freeradbiomed.2024.01.017] [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: 12/21/2023] [Accepted: 01/12/2024] [Indexed: 01/23/2024]
Abstract
Osteoporosis, which manifests as reduced bone mass and deteriorated bone quality, is common in the elderly population. It is characterized by persistent elevation of macrophage-associated inflammation and active osteoclast bone resorption. Currently, the roles of intracellular metabolism in regulating these processes remain unclear. In this study, we initially performed bioinformatics analysis and observed a significant increase in the proportion of M1 macrophages in bone marrow with aging. Further metabolomics analysis demonstrated a notable reduction in the expression of carnitine metabolites in aged macrophages, while carnitine was not detected in osteoclasts. During the differentiation process, osteoclasts took up carnitine synthesized by macrophages to regulate their own activity. Mechanistically, carnitine enhanced the function of Nrf2 by inhibiting the Keap1-Nrf2 interaction, reducing the proteasome-dependent ubiquitination and degradation of Nrf2. In silico molecular ligand docking analysis of the interaction between carnitine and Keap1 showed that carnitine binds to Keap1 to stabilize Nrf2 and enhance its function. In this study, we found that the decrease in carnitine levels in aging macrophages causes overactivation of osteoclasts, ultimately leading to osteoporosis. A decrease in serum carnitine levels in patients with osteoporosis was found to have good diagnostic and predictive value. Moreover, supplementation with carnitine was shown to be effective in the treatment of osteoporosis.
Collapse
Affiliation(s)
- Tao Yang
- The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, 289 Kuocang Road, Lishui, Zhejiang, 323000, PR China
| | - Shijie Liu
- The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, 289 Kuocang Road, Lishui, Zhejiang, 323000, PR China
| | - Haiwei Ma
- The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, 289 Kuocang Road, Lishui, Zhejiang, 323000, PR China
| | - Hehuan Lai
- The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, 289 Kuocang Road, Lishui, Zhejiang, 323000, PR China
| | - Chengdi Wang
- The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, 289 Kuocang Road, Lishui, Zhejiang, 323000, PR China
| | - Kainan Ni
- The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, 289 Kuocang Road, Lishui, Zhejiang, 323000, PR China
| | - Yahong Lu
- The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, 289 Kuocang Road, Lishui, Zhejiang, 323000, PR China
| | - Weiqing Li
- The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, 289 Kuocang Road, Lishui, Zhejiang, 323000, PR China
| | - Xingyu Hu
- The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, 289 Kuocang Road, Lishui, Zhejiang, 323000, PR China
| | - Zhiguo Zhou
- The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, 289 Kuocang Road, Lishui, Zhejiang, 323000, PR China
| | - Chao Lou
- The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, 289 Kuocang Road, Lishui, Zhejiang, 323000, PR China.
| | - Dengwei He
- The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, 289 Kuocang Road, Lishui, Zhejiang, 323000, PR China.
| |
Collapse
|
46
|
Song Q, Wang D, Li H, Wang Z, Sun S, Wang Z, Liu Y, Lin S, Li G, Zhang S, Zhang P. Dual-response of multi-functional microsphere system to ultrasound and microenvironment for enhanced bone defect treatment. Bioact Mater 2024; 32:304-318. [PMID: 37876555 PMCID: PMC10590728 DOI: 10.1016/j.bioactmat.2023.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/21/2023] [Accepted: 10/07/2023] [Indexed: 10/26/2023] Open
Abstract
Using bone tissue engineering strategies to achieve bone defect repair is a promising modality. However, the repair process outcomes are often unsatisfactory. Here we properly designed a multi-functional microsphere system, which could deliver bioactive proteins under the dual response of ultrasound and microenvironment, release microenvironment-responsive products on demand, reverse bone injury microenvironment, regulate the immune microenvironment, and achieve excellent bone defect treatment outcomes. In particular, the MnO2 introduced into the poly(lactic-co-glycolic acid) (PLGA) microspheres during synthesis could consume the acid produced by the degradation of PLGA to protect bone morphogenetic protein-2 (BMP-2). More importantly, MnO2 could consume reactive oxygen species (ROS) and produce Mn2+ and oxygen (O2), further promoting the repair of bone defects while reversing the microenvironment. Moreover, the reversal of the bone injury microenvironment and the depletion of ROS promoted the polarization of M1 macrophages to M2 macrophages, and the immune microenvironment was regulated. Notably, the ultrasound (US) irradiation used during treatment also allowed the on-demand release of microenvironment-responsive products. The multi-functional microsphere system combines the effects of on-demand delivery, reversal of bone injury microenvironment, and regulation of the immune microenvironment, providing new horizons for the clinical application of protein delivery and bone defect repair.
Collapse
Affiliation(s)
- Qingxu Song
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, 130021, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Dianwei Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Haoyu Li
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, 130021, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Zongliang Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Songjia Sun
- Department of Dermatology, Second Hospital of Jilin University, Changchun, 130022, China
| | - Zhenyu Wang
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yi Liu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Sien Lin
- Department of Orthopaedics and Traumatology and Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
| | - Gang Li
- Department of Orthopaedics and Traumatology and Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
| | - Shaokun Zhang
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Peibiao Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| |
Collapse
|
47
|
Wang W, Zheng X, Wang H, Zuo B, Chen S, Li J. Mechanical Unloading Promotes Osteoclastic Differentiation and Bone Resorption by Modulating the MSC Secretome to Favor Inflammation. Cell Transplant 2024; 33:9636897241236584. [PMID: 38501500 PMCID: PMC10953070 DOI: 10.1177/09636897241236584] [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: 10/22/2023] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 03/20/2024] Open
Abstract
Aging, space flight, and prolonged bed rest have all been linked to bone loss, and no effective treatments are clinically available at present. Here, with the rodent hindlimb unloading (HU) model, we report that the bone marrow (BM) microenvironment was significantly altered, with an increased number of myeloid cells and elevated inflammatory cytokines. In such inflammatory BM, the osteoclast-mediated bone resorption was greatly enhanced, leading to a shifted bone remodeling balance that ultimately ends up with disuse-induced osteoporosis. Using Piezo1 conditional knockout (KO) mice (Piezo1fl/fl;LepRCre), we proved that lack of mechanical stimuli on LepR+ mesenchymal stem cells (MSCs) is the main reason for the pathological BM inflammation. Mechanically, the secretome of MSCs was regulated by mechanical stimuli. Inadequate mechanical load leads to increased production of inflammatory cytokines, such as interleukin (IL)-1α, IL-6, macrophage colony-stimulating factor 1 (M-CSF-1), and so on, which promotes monocyte proliferation and osteoclastic differentiation. Interestingly, transplantation of 10% cyclic mechanical stretch (CMS)-treated MSCs into HU animals significantly alleviated the BM microenvironment and rebalanced bone remodeling. In summary, our research revealed a new mechanism underlying mechanical unloading-induced bone loss and suggested a novel stem cell-based therapy to potentially prevent disuse-induced osteoporosis.
Collapse
Affiliation(s)
- Wanyuji Wang
- Department of Cell Biology, Zunyi Medical University, Zunyi, China
| | - Xueling Zheng
- Department of Cell Biology, Zunyi Medical University, Zunyi, China
| | - Hehe Wang
- Department of Cell Biology, Zunyi Medical University, Zunyi, China
| | - Bin Zuo
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Sisi Chen
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiao Li
- Department of Cell Biology, Zunyi Medical University, Zunyi, China
| |
Collapse
|
48
|
Bi M, Yang K, Yu T, Wu G, Li Q. Cell-based mechanisms and strategies of co-culture system both in vivo and vitro for bone tissue engineering. Biomed Pharmacother 2023; 169:115907. [PMID: 37984308 DOI: 10.1016/j.biopha.2023.115907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023] Open
Abstract
The lack of a functional vascular supply has been identified as a major challenge limiting the clinical introduction of stem cell-based bone tissue engineering (BTE) for the repair of large-volume bone defects (LVBD). Various approaches have been explored to improve the vascular supply in tissue-engineered constructs, and the development of strategies that could effectively induce the establishment of a functional vascular supply has become a major goal of BTE research. One of the state-of-the-art methods is to incorporate both angiogenic and osteogenic cells in co-culture systems. This review clarifies the key concepts involved, summarises the cell types and models used to date, and systematically evaluates their performance. We also discuss the cell-to-cell communication between these two cell types and the strategies explored in BTE constructs with angiogenic and osteogenic cells to optimise their functions. In addition, we outline unresolved issues and remaining obstacles that need to be overcome for further development in this field and eventual successful repair of LVBD.
Collapse
Affiliation(s)
- Mengning Bi
- Department of Prosthetic Dentistry, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China; Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology Shanghai, China
| | - Kaiwen Yang
- Department of Prosthetic Dentistry, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China; Department of Oral Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai Key Laboratory of Stomatology &Shanghai Research Institute of Stomatology; National Clinical Research Center of Stomatology, Shanghai, China
| | - Tao Yu
- Department of Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Gang Wu
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam (VU), Amsterdam Movement Science (AMS), Amsterdam, the Netherlands; Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam (UvA) and Vrije Universiteit Amsterdam (VU), Amsterdam, the Netherlands.
| | - Qiong Li
- Department of Prosthetic Dentistry, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China.
| |
Collapse
|
49
|
Pius AK, Toya M, Gao Q, Lee ML, Ergul YS, Chow SKH, Goodman SB. Effects of Aging on Osteosynthesis at Bone-Implant Interfaces. Biomolecules 2023; 14:52. [PMID: 38254652 PMCID: PMC10813487 DOI: 10.3390/biom14010052] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/23/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
Joint replacement is a common surgery and is predominantly utilized for treatment of osteoarthritis in the aging population. The longevity of many of these implants depends on bony ingrowth. Here, we provide an overview of current techniques in osteogenesis (inducing bone growth onto an implant), which is affected by aging and inflammation. In this review we cover the biologic underpinnings of these processes as well as the clinical applications. Overall, aging has a significant effect at the cellular and macroscopic level that impacts osteosynthesis at bone-metal interfaces after joint arthroplasty; potential solutions include targeting prolonged inflammation, preventing microbial adhesion, and enhancing osteoinductive and osteoconductive properties.
Collapse
Affiliation(s)
- Alexa K. Pius
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA 94063, USA; (A.K.P.); (M.T.); (Q.G.); (M.L.L.); (Y.S.E.); (S.K.-H.C.)
| | - Masakazu Toya
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA 94063, USA; (A.K.P.); (M.T.); (Q.G.); (M.L.L.); (Y.S.E.); (S.K.-H.C.)
| | - Qi Gao
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA 94063, USA; (A.K.P.); (M.T.); (Q.G.); (M.L.L.); (Y.S.E.); (S.K.-H.C.)
| | - Max L. Lee
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA 94063, USA; (A.K.P.); (M.T.); (Q.G.); (M.L.L.); (Y.S.E.); (S.K.-H.C.)
| | - Yasemin Sude Ergul
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA 94063, USA; (A.K.P.); (M.T.); (Q.G.); (M.L.L.); (Y.S.E.); (S.K.-H.C.)
| | - Simon Kwoon-Ho Chow
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA 94063, USA; (A.K.P.); (M.T.); (Q.G.); (M.L.L.); (Y.S.E.); (S.K.-H.C.)
| | - Stuart Barry Goodman
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA 94063, USA; (A.K.P.); (M.T.); (Q.G.); (M.L.L.); (Y.S.E.); (S.K.-H.C.)
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
50
|
Bai L, Song P, Su J. Bioactive elements manipulate bone regeneration. BIOMATERIALS TRANSLATIONAL 2023; 4:248-269. [PMID: 38282709 PMCID: PMC10817798 DOI: 10.12336/biomatertransl.2023.04.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/21/2023] [Accepted: 12/08/2023] [Indexed: 01/30/2024]
Abstract
While bone tissue is known for its inherent regenerative abilities, various pathological conditions and trauma can disrupt its meticulously regulated processes of bone formation and resorption. Bone tissue engineering aims to replicate the extracellular matrix of bone tissue as well as the sophisticated biochemical mechanisms crucial for effective regeneration. Traditionally, the field has relied on external agents like growth factors and pharmaceuticals to modulate these processes. Although efficacious in certain scenarios, this strategy is compromised by limitations such as safety issues and the transient nature of the compound release and half-life. Conversely, bioactive elements such as zinc (Zn), magnesium (Mg) and silicon (Si), have garnered increasing interest for their therapeutic benefits, superior stability, and reduced biotic risks. Moreover, these elements are often incorporated into biomaterials that function as multifaceted bioactive components, facilitating bone regeneration via release on-demand. By elucidating the mechanistic roles and therapeutic efficacy of the bioactive elements, this review aims to establish bioactive elements as a robust and clinically viable strategy for advanced bone regeneration.
Collapse
Affiliation(s)
- Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Peiran Song
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| |
Collapse
|