1
|
Hiepen C, Benamar M, Barrasa-Fano J, Condor M, Ilhan M, Münch J, Hastar N, Kerkhoff Y, Harms GS, Mielke T, Koenig B, Block S, Rocks O, Abdelilah-Seyfried S, Van Oosterwyck H, Knaus P. Endothelial tip-cell position, filopodia formation and biomechanics require BMPR2 expression and signaling. Commun Biol 2025; 8:21. [PMID: 39779836 PMCID: PMC11711618 DOI: 10.1038/s42003-024-07431-8] [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] [Academic Contribution Register] [Received: 01/02/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Blood vessel formation relies on biochemical and mechanical signals, particularly during sprouting angiogenesis when endothelial tip cells (TCs) guide sprouting through filopodia formation. The contribution of BMP receptors in defining tip-cell characteristics is poorly understood. Our study combines genetic, biochemical, and molecular methods together with 3D traction force microscopy, which reveals an essential role of BMPR2 for actin-driven filopodia formation and mechanical properties of endothelial cells (ECs). Targeting of Bmpr2 reduced sprouting angiogenesis in zebrafish and BMPR2-deficient human ECs formed fewer filopodia, affecting cell migration and actomyosin localization. Spheroid assays revealed a reduced sprouting of BMPR2-deficient ECs in fibrin gels. Even more strikingly, in mosaic spheroids, BMPR2-deficient ECs failed to acquire tip-cell positions. Yet, 3D traction force microscopy revealed that these distinct cell behaviors of BMPR2-deficient tip cells cannot be explained by differences in force-induced matrix deformations, even though these cells adopted distinct cone-shaped morphologies. Notably, BMPR2 positively regulates local CDC42 activity at the plasma membrane to promote filopodia formation. Our findings reveal that BMPR2 functions as a nexus integrating biochemical and biomechanical processes crucial for TCs during angiogenesis.
Collapse
Affiliation(s)
- Christian Hiepen
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany.
- Westphalian University of Applied Sciences, August-Schmidt-Ring 10, 45665, Recklinghausen, Germany.
| | - Mounir Benamar
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany
| | - Jorge Barrasa-Fano
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Celestijnenlaan 300 C, 3001, Leuven, Belgium
| | - Mar Condor
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Celestijnenlaan 300 C, 3001, Leuven, Belgium
| | - Mustafa Ilhan
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany
- Berlin School of Integrative Oncology, Augustenburger Platz 1, D-13353, Berlin, Germany
| | - Juliane Münch
- Universität Potsdam, Institute of Biochemistry and Biology, Karl-Liebknecht Strasse 24-25, 14476, Potsdam-Golm, Germany
| | - Nurcan Hastar
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany
| | - Yannic Kerkhoff
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany
| | - Gregory S Harms
- Universitätsmedizin, Johannes Gutenberg-Universität Mainz, Cell Biology Unit, Imaging Core Facility and the Research Center for Immune Intervention, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Thorsten Mielke
- Max-Planck-Institute for Molecular Genetics, Microscopy & Cryo-Electron Microscopy, Ihnestraße 63-73, 14195, Berlin, Germany
| | - Benjamin Koenig
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany
- Leibniz Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Stephan Block
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany
| | - Oliver Rocks
- Charité - Universitätsmedizin Berlin, Systemic Cell Dynamics, Charitéplatz 1, 10117, Berlin, Germany
| | - Salim Abdelilah-Seyfried
- Universität Potsdam, Institute of Biochemistry and Biology, Karl-Liebknecht Strasse 24-25, 14476, Potsdam-Golm, Germany
| | - Hans Van Oosterwyck
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Celestijnenlaan 300 C, 3001, Leuven, Belgium
- KU Leuven, Prometheus Division of Skeletal Tissue Engineering, Leuven, Belgium
| | - Petra Knaus
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany.
| |
Collapse
|
2
|
Prummel KD, Woods K, Kholmatov M, Schmitt EC, Vlachou EP, Poschmann G, Stühler K, Wehner R, Schmitz M, Winter S, Oelschlaegel U, Schwartz LS, Moura PL, Hellström-Lindberg E, Theobald M, Trowbridge JJ, Platzbecker U, Zaugg JB, Guezguez B. Inflammatory Mesenchymal Stromal Cells and IFN-responsive T cells are key mediators of human bone marrow niche remodeling in CHIP and MDS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.27.625734. [PMID: 39651275 PMCID: PMC11623587 DOI: 10.1101/2024.11.27.625734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 12/11/2024]
Abstract
Somatic mutations in hematopoietic stem/progenitor cells (HSPCs) can lead to clonal hematopoiesis of indeterminate potential (CHIP), potentially progressing to myelodysplastic syndromes (MDS). Here, we investigated how CHIP and MDS remodel the human bone marrow (BM) niche relative to healthy elderly donors, using single cell and anatomical analyses in a large BM cohort. We found distinct inflammatory remodeling of the BM in CHIP and MDS. Furthermore, the stromal compartment progressively lost its HSPC-supportive adipogenic CXCL12-abundant reticular cells while an inflammatory mesenchymal stroma cell (iMSCs) population emerged in CHIP, which expanded in MDS. iMSCs exhibited distinct functional signatures in CHIP and MDS, retaining residual HSPC-support and angiogenic activity in MDS, corresponding with an increase in microvasculature in the MDS niche. Additionally, an IFN-responsive T cell population was linked to fueling inflammation in the stroma. Overall, these findings open new avenues for early intervention in hematological malignancies.
Collapse
|
3
|
Huang Z, Huang J, Lin Y, Deng Y, Yang L, Zhang X, Huang H, Sun Q, Liu H, Liang H, Lv Z, He B, Hu F. Construction and validation of a TAMRGs prognostic signature for gliomas by integrated analysis of scRNA and bulk RNA sequencing data. Brain Res 2024; 1846:149237. [PMID: 39270996 DOI: 10.1016/j.brainres.2024.149237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/31/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND This study aimed to construct and validate a prognostic model based on tumor associated macrophage-related genes (TAMRGs) by integrating single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing (bulk RNA-seq) data. METHODS The scRNA-seq data of three inhouse glioma tissues were used to identify the tumor-associated macrophages (TAMs) marker genes, the DEGs from the The Cancer Genome Atlas (TCGA) - Genotype-Tissue Expression (GTEx) dataset were used to further select TAMs marker genes. Subsequently, a TAMRG-score was constructed by Least absolute shrinkage and selection operator (LASSO) regression and multivariate Cox regression analysis in the TCGA dataset and validated in the Chinese Glioma Genome Atlas (CGGA) dataset. RESULTS We identified 186 TAMs marker genes, and a total of 6 optimal prognostic genes including CKS2, LITAF, CTSB, TWISTNB, PPIF and G0S2 were selected to construct a TAMRG-score. The high TAMRG-score was significantly associated with worse prognosis (log-rank test, P<0.001). Moreover, the TAMRG-score outperformed the other three models with AUC of 0.808. Immune cell infiltration, TME scores, immune checkpoints, TMB and drug susceptibility were significantly different between TAMRG-score groups. In addition, a nomogram were constructed by combing the TAMRG-score and clinical information (Age, Grade, IDH mutation and 1p19q codeletion) to predict the survival of glioma patients with AUC of 0.909 for 1-year survival. CONCLUSION The high TAMRG-score group was associated with a poor prognosis. A nomogram by incorporating TMARG-score could precisely predict glioma survival, and provide evidence for personalized treatment of glioma.
Collapse
Affiliation(s)
- Zhicong Huang
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, PR China
| | - Jingyao Huang
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, PR China
| | - Ying Lin
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, PR China
| | - Ying Deng
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, PR China
| | - Longkun Yang
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, PR China
| | - Xing Zhang
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, PR China
| | - Hao Huang
- Department of Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, Guangdong, PR China
| | - Qian Sun
- Department of Neurosurgery, The Tumor Hospital Affiliated to Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Hui Liu
- Department of Neurosurgery, The Tumor Hospital Affiliated to Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Hongsheng Liang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Zhonghua Lv
- Department of Neurosurgery, The Tumor Hospital Affiliated to Harbin Medical University, Harbin, Heilongjiang, PR China.
| | - Baochang He
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, PR China.
| | - Fulan Hu
- Department of Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, Guangdong, PR China.
| |
Collapse
|
4
|
Dong Y, Li J, Jiang Q, He S, Wang B, Yi Q, Cheng X, Gao X, Bai Y. Structure, ingredient, and function-based biomimetic scaffolds for accelerated healing of tendon-bone interface. J Orthop Translat 2024; 48:70-88. [PMID: 39185339 PMCID: PMC11342074 DOI: 10.1016/j.jot.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 03/25/2024] [Revised: 06/11/2024] [Accepted: 07/16/2024] [Indexed: 08/27/2024] Open
Abstract
Background Tendon-bone interface (TBI) repair is slow and challenging owing to its hierarchical structure, gradient composition, and complex function. In this work, enlightened by the natural characteristics of TBI microstructure and the demands of TBI regeneration, a structure, composition, and function-based scaffold was fabricated. Methods: The biomimetic scaffold was designed based on the "tissue-inducing biomaterials" theory: (1) a porous scaffold was created with poly-lactic-co-glycolic-acid, nano-hydroxyapatite and loaded with BMP2-gelatinmp to simulate the bone (BP); (2) a hydrogel was produced from sodium alginate, type I collagen, and loaded with TGF-β3 to simulate the cartilage (CP); (3) the L-poly-lactic-acid fibers were oriented to simulate the tendon (TP). The morphology of tri-layered constructs, gelation kinetics, degradation rate, release kinetics and mechanical strength of the scaffold were characterized. Then, bone marrow mesenchymal stem cells (MSCs) and tenocytes (TT-D6) were cultured on the scaffold to evaluate its gradient differentiation inductivity. A rat Achilles tendon defect model was established, and BMSCs seeded on scaffolds were implanted into the lesionsite. The tendon-bone lesionsite of calcaneus at 4w and 8w post-operation were obtained for gross observation, radiological evaluation, biomechanical and histological assessment. Results The hierarchical microstructures not only endowed the scaffold with gradual composition and mechanical properties for matching the regional biophysical characteristics of TBI but also exhibited gradient differentiation inductivity through providing regional microenvironment for cells. Moreover, the scaffold seeded with cells could effectively accelerate healing in rat Achilles tendon defects, attributable to its enhanced differentiation performance. Conclusion The hierarchical scaffolds simulating the structural, compositional, and cellular heterogeneity of natural TBI tissue performed therapeutic effects on promoting regeneration of TBI and enhancing the healing quality of Achilles tendon. The translational potential of this article The novel scaffold showed the great efficacy on tendon to bone healing by offering a structural and compositional microenvironment. The results meant that the hierarchical scaffold with BMSCs may have a great potential for clinical application.
Collapse
Affiliation(s)
- YuHan Dong
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - JiangFeng Li
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma, Burn and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Qiang Jiang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - SiRong He
- School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Bin Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - QiYing Yi
- Laboratory Animal Center, Chongqing Medical University, Chongqing, 400016, China
| | - XiTing Cheng
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Xiang Gao
- College of Stomatology, Chongqing Medical University, Chongqing, 400016, China
| | - Yan Bai
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
5
|
Wu C, Yan J, Ge C, Xie L, He Y, Zhao Z, Deng Y, Dong Q, Yin L. Macrophage membrane-reversibly camouflaged nanotherapeutics accelerate fracture healing by fostering MSCs recruitment and osteogenic differentiation. J Nanobiotechnology 2024; 22:411. [PMID: 38997706 PMCID: PMC11241938 DOI: 10.1186/s12951-024-02679-y] [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] [Academic Contribution Register] [Received: 05/08/2024] [Accepted: 06/30/2024] [Indexed: 07/14/2024] Open
Abstract
The fracture healing outcome is largely dependent on the quantities as well as osteogenic differentiation capacities of mesenchymal stem cells (MSCs) at the lesion site. Herein, macrophage membrane (MM)-reversibly cloaked nanocomplexes (NCs) are engineered for the lesion-targeted and hierarchical co-delivery of short stromal derived factor-1α peptide (sSDF-1α) and Ckip-1 small interfering RNA (Ckip-1 siRNA, siCkip-1) to promote bone repair by concurrently fostering recruitment and osteogenic differentiation of endogenous MSCs. To construct the NCs, a membrane-penetrating α-helical polypeptide first assembles with siCkip-1, and the cationic NCs are sequentially coated with catalase and an outer shell of sSDF-1α-anchored MM. Due to MM-assisted inflammation homing, intravenously injected NCs could efficiently accumulate at the fractured femur, where catalase decomposes the local hydrogen peroxide to generate oxygen bubbles that drives the shedding of sSDF-1α-anchored MM in the extracellular compartment. The exposed, cationic inner core thus enables robust trans-membrane delivery into MSCs to induce Ckip-1 silencing. Consequently, sSDF-1α-guided MSCs recruitment cooperates with siCkip-1-mediated osteogenic differentiation to facilitate bone formation and accelerate bone fracture healing. This study provides an enlightened strategy for the hierarchical co-delivery of macromolecular drugs into different cellular compartments, and it also renders a promising modality for the management of fracture healing.
Collapse
Affiliation(s)
- Cheng Wu
- Department of Orthopedics, The Second Affiliated Hospital, Soochow University, Suzhou, 215004, China
- Department of Orthopedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Jing Yan
- Department of Gastroenterology, The Second Affiliated Hospital, Soochow University, Suzhou, 215004, China
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Chenglong Ge
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Lucheng Xie
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Yunjie He
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Ziyin Zhao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Yekun Deng
- Department of Orthopedics, The Second Affiliated Hospital, Soochow University, Suzhou, 215004, China
| | - Qirong Dong
- Department of Orthopedics, The Second Affiliated Hospital, Soochow University, Suzhou, 215004, China.
| | - Lichen Yin
- Department of Gastroenterology, The Second Affiliated Hospital, Soochow University, Suzhou, 215004, China.
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
6
|
Gong JS, Zhu GQ, Zhang Y, Chen B, Liu YW, Li HM, He ZH, Zou JT, Qian YX, Zhu S, Hu XY, Rao SS, Cao J, Xie H, Wang ZX, Du W. Aptamer-functionalized hydrogels promote bone healing by selectively recruiting endogenous bone marrow mesenchymal stem cells. Mater Today Bio 2023; 23:100854. [PMID: 38024846 PMCID: PMC10665677 DOI: 10.1016/j.mtbio.2023.100854] [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] [Academic Contribution Register] [Received: 09/03/2023] [Revised: 10/22/2023] [Accepted: 11/01/2023] [Indexed: 12/01/2023] Open
Abstract
Bone regeneration heavily relies on bone marrow mesenchymal stem cells (BMSCs). However, recruiting endogenous BMSCs for in situ bone regeneration remains challenging. In this study, we developed a novel BMSC-aptamer (BMSC-apt) functionalized hydrogel (BMSC-aptgel) and evaluated its functions in recruiting BMSCs and promoting bone regeneration. The functional hydrogels were synthesized between maleimide-terminated 4-arm polyethylene glycols (PEG) and thiol-flanked PEG crosslinker, allowing rapid in situ gel formation. The aldehyde group-modified BMSC-apt was covalently bonded to a thiol-flanked PEG crosslinker to produce high-density aptamer coverage on the hydrogel surface. In vitro and in vivo studies demonstrated that the BMSC-aptgel significantly increased BMSC recruitment, migration, osteogenic differentiation, and biocompatibility. In vivo fluorescence tomography imaging demonstrated that functionalized hydrogels effectively recruited DiR-labeled BMSCs at the fracture site. Consequently, a mouse femur fracture model significantly enhanced new bone formation and mineralization. The aggregated BMSCs stimulated bone regeneration by balancing osteogenic and osteoclastic activities and reduced the local inflammatory response via paracrine effects. This study's findings suggest that the BMSC-aptgel can be a promising and effective strategy for promoting in situ bone regeneration.
Collapse
Affiliation(s)
- Jiang-Shan Gong
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
| | - Guo-Qiang Zhu
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
| | - Yu Zhang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Bei Chen
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yi-Wei Liu
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
| | - Hong-Ming Li
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
| | - Ze-Hui He
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
| | - Jing-Tao Zou
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
| | - Yu-Xuan Qian
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
| | - Sheng Zhu
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
| | - Xin-Yue Hu
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
| | - Shan-Shan Rao
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, Hunan, 410008, China
| | - Jia Cao
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, Hunan, 410008, China
| | - Hui Xie
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, Hunan, 410008, China
| | - Zhen-Xing Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, Hunan, 410008, China
| | - Wei Du
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, Hunan, 410008, China
- Department of Rehabilitation Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| |
Collapse
|
7
|
Wang X, Zhu X, Wang D, Li X, Wang J, Yin G, Huang Z, Pu X. Identification of a Specific Phage as Growth Factor Alternative Promoting the Recruitment and Differentiation of MSCs in Bone Tissue Regeneration. ACS Biomater Sci Eng 2023; 9:2426-2437. [PMID: 37023478 DOI: 10.1021/acsbiomaterials.2c01538] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 04/08/2023]
Abstract
Inefficient use and loss of exogenously implanted mesenchymal stem cells (MSCs) are major concerns in MSCs-based bone tissue engineering. It is a promising approach to overcome the above issues by recruiting and regulation of endogenous MSCs. However, there are few substances that can recruit MSCs effectively and specifically to the site of bone injury. In this study, we identified a phage clone (termed P11) with specific affinity for MSCs through phage display biopanning, and further investigated the effects of P11 on the cytological behavior of MSCs and macrophages. The results showed that P11 could bind MSCs specifically and promote the proliferation and migration of MSCs. Meanwhile, P11 could polarize macrophages to the M1 phenotype and significantly changed their morphology, which further enhanced the chemotaxis of MSCs. Additionally, RNA-seq results revealed that P11 could promote the secretion of osteogenesis-related markers in MSCs through the TPL2-MEK-ERK signaling pathway. Altogether, P11 has great potential to be used as growth factor alternatives in bone tissue engineering, with the advantages of cheaper and stable activity. Our study also advances the understanding of the effects of phages on macrophages and MSCs, and provides a new idea for the development in the field of phage-based tissue engineering.
Collapse
Affiliation(s)
- Xingming Wang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Xiupeng Zhu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, People's Republic of China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People's Republic of China
| | - Danni Wang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Xiaoxu Li
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Juan Wang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Guangfu Yin
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Zhongbing Huang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Ximing Pu
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| |
Collapse
|
8
|
Popkov A, Kononovich N, Dubinenko G, Gorbach E, Shastov A, Tverdokhlebov S, Popkov D. Long Bone Defect Filling with Bioactive Degradable 3D-Implant: Experimental Study. Biomimetics (Basel) 2023; 8:biomimetics8020138. [PMID: 37092390 PMCID: PMC10123725 DOI: 10.3390/biomimetics8020138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/27/2023] [Revised: 03/26/2023] [Accepted: 03/26/2023] [Indexed: 03/30/2023] Open
Abstract
Previously, 3D-printed bone grafts made of titanium alloy with bioactive coating has shown great potential for the restoration of bone defects. Implanted into a medullary canal titanium graft with cellular structure demonstrated stimulation of the reparative osteogenesis and successful osseointegration of the graft into a single bone-implant block. The purpose of this study was to investigate osseointegration of a 3D-printed degradable polymeric implant with cellular structure as preclinical testing of a new technique for bone defect restoration. During an experimental study in sheep, a 20 mm-long segmental tibial defect was filled with an original cylindrical implant with cellular structure made of polycaprolactone coated with hydroxyapatite. X-ray radiographs demonstrated reparative bone regeneration from the periosteum lying on the periphery of cylindrical implant to its center in a week after the surgery. Cellular structure of the implant was fully filled with newly-formed bone tissue on the 4th week after the surgery. The bone tissue regeneration from the proximal and distal bone fragments was evident on 3rd week. This provides insight into the use of bioactive degradable implants for the restoration of segmental bone defects. Degradable implant with bioactive coating implanted into a long bone segmental defect provides stimulation of reparative osteogenesis and osseointegration into the single implant-bone block.
Collapse
|
9
|
He Y, Li F, Jiang P, Cai F, Lin Q, Zhou M, Liu H, Yan F. Remote control of the recruitment and capture of endogenous stem cells by ultrasound for in situ repair of bone defects. Bioact Mater 2023; 21:223-238. [PMID: 36157244 PMCID: PMC9465026 DOI: 10.1016/j.bioactmat.2022.08.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/28/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 12/02/2022] Open
Abstract
Stem cell-based tissue engineering has provided a promising platform for repairing of bone defects. However, the use of exogenous bone marrow mesenchymal stem cells (BMSCs) still faces many challenges such as limited sources and potential risks. It is important to develop new approach to effectively recruit endogenous BMSCs and capture them for in situ bone regeneration. Here, we designed an acoustically responsive scaffold (ARS) and embedded it into SDF-1/BMP-2 loaded hydrogel to obtain biomimetic hydrogel scaffold complexes (BSC). The SDF-1/BMP-2 cytokines can be released on demand from the BSC implanted into the defected bone via pulsed ultrasound (p-US) irradiation at optimized acoustic parameters, recruiting the endogenous BMSCs to the bone defected or BSC site. Accompanied by the daily p-US irradiation for 14 days, the alginate hydrogel was degraded, resulting in the exposure of ARS to these recruited host stem cells. Then another set of sinusoidal continuous wave ultrasound (s-US) irradiation was applied to excite the ARS intrinsic resonance, forming highly localized acoustic field around its surface and generating enhanced acoustic trapping force, by which these recruited endogenous stem cells would be captured on the scaffold, greatly promoting them to adhesively grow for in situ bone tissue regeneration. Our study provides a novel and effective strategy for in situ bone defect repairing through acoustically manipulating endogenous BMSCs. We designed ARS and embedded it into SDF-1/BMP-2 loaded hydrogel to form BSC. The BSC can release SDF-1/BMP-2 by p-US irradiation for recruitment of endogenous BMSCs and capture them by s-US irradiation. The in situ repair of bone defects were successfully realized by US-mediated control of the recruitment and capture of BMSCs.
Collapse
Affiliation(s)
- Yanni He
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, PR China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Fei Li
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Peng Jiang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Feiyan Cai
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Qin Lin
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Meijun Zhou
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, PR China
| | - Hongmei Liu
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, PR China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
- Corresponding author. Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, PR China.
| | - Fei Yan
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
- Corresponding author. Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China.
| |
Collapse
|
10
|
Kim J, Choi HS, Kim YM, Song SC. Thermo-Responsive Nanocomposite Bioink with Growth-Factor Holding and its Application to Bone Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2203464. [PMID: 36526612 DOI: 10.1002/smll.202203464] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 06/03/2022] [Revised: 11/03/2022] [Indexed: 06/17/2023]
Abstract
Three-dimensional (3D) bioprinting, which is being increasingly used in tissue engineering, requires bioinks with tunable mechanical properties, biological activities, and mechanical strength for in vivo implantation. Herein, a growth-factor-holding poly(organophosphazene)-based thermo-responsive nanocomposite (TNC) bioink system is developed. The mechanical properties of the TNC bioink are easily controlled within a moderate temperature range (5-37 °C). During printing, the mechanical properties of the TNC bioink, which determine the 3D printing resolution, can be tuned by varying the temperature (15-30 °C). After printing, TNC bioink scaffolds exhibit maximum stiffness at 37 °C. Additionally, because of its shear-thinning and self-healing properties, TNC bioinks can be extruded smoothly, demonstrating good printing outcomes. TNC bioink loaded with bone morphogenetic protein-2 (BMP-2) and transforming growth factor-beta1 (TGF-β1), key growth factors for osteogenesis, is used to print a scaffold that can stimulate biological activity. A biological scaffold printed using TNC bioink loaded with both growth factors and implanted on a rat calvarial defect model reveals significantly improved bone regenerative effects. The TNC bioink system is a promising next-generation bioink platform because its mechanical properties can be tuned easily for high-resolution 3D bioprinting with long-term stability and its growth-factor holding capability has strong clinical applicability.
Collapse
Affiliation(s)
- Jun Kim
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Hoon-Seong Choi
- Research Animal Resource Center, Research Resources Division, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Young-Min Kim
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Soo-Chang Song
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
- Nexgel Biotech, Co., Ltd, Seoul, 02792, Republic of Korea
| |
Collapse
|
11
|
Xu J, Fahmy-Garcia S, Wesdorp MA, Kops N, Forte L, De Luca C, Misciagna MM, Dolcini L, Filardo G, Labberté M, Vancíková K, Kok J, van Rietbergen B, Nickel J, Farrell E, Brama PAJ, van Osch GJVM. Effectiveness of BMP-2 and PDGF-BB Adsorption onto a Collagen/Collagen-Magnesium-Hydroxyapatite Scaffold in Weight-Bearing and Non-Weight-Bearing Osteochondral Defect Bone Repair: In Vitro, Ex Vivo and In Vivo Evaluation. J Funct Biomater 2023; 14:jfb14020111. [PMID: 36826910 PMCID: PMC9961206 DOI: 10.3390/jfb14020111] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/20/2023] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Despite promising clinical results in osteochondral defect repair, a recently developed bi-layered collagen/collagen-magnesium-hydroxyapatite scaffold has demonstrated less optimal subchondral bone repair. This study aimed to improve the bone repair potential of this scaffold by adsorbing bone morphogenetic protein 2 (BMP-2) and/or platelet-derived growth factor-BB (PDGF-BB) onto said scaffold. The in vitro release kinetics of BMP-2/PDGF-BB demonstrated that PDGF-BB was burst released from the collagen-only layer, whereas BMP-2 was largely retained in both layers. Cell ingrowth was enhanced by BMP-2/PDFG-BB in a bovine osteochondral defect ex vivo model. In an in vivo semi-orthotopic athymic mouse model, adding BMP-2 or PDGF-BB increased tissue repair after four weeks. After eight weeks, most defects were filled with bone tissue. To further investigate the promising effect of BMP-2, a caprine bilateral stifle osteochondral defect model was used where defects were created in weight-bearing femoral condyle and non-weight-bearing trochlear groove locations. After six months, the adsorption of BMP-2 resulted in significantly less bone repair compared with scaffold-only in the femoral condyle defects and a trend to more bone repair in the trochlear groove. Overall, the adsorption of BMP-2 onto a Col/Col-Mg-HAp scaffold reduced bone formation in weight-bearing osteochondral defects, but not in non-weight-bearing osteochondral defects.
Collapse
Affiliation(s)
- Jietao Xu
- Department of Orthopedics and Sports Medicine, Erasmus MC, University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Shorouk Fahmy-Garcia
- Department of Orthopedics and Sports Medicine, Erasmus MC, University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Marinus A. Wesdorp
- Department of Orthopedics and Sports Medicine, Erasmus MC, University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Nicole Kops
- Department of Orthopedics and Sports Medicine, Erasmus MC, University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Lucia Forte
- Fin-Ceramica Faenza S.p.A, 48018 Faenza, Italy
| | | | | | | | - Giuseppe Filardo
- Applied and Translational Research Center, IRCCS Rizzoli Orthopaedic Institute, 40136 Bologna, Italy
| | - Margot Labberté
- School of Veterinary Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Karin Vancíková
- School of Veterinary Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Joeri Kok
- Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Bert van Rietbergen
- Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Joachim Nickel
- Department Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, 97070 Würzburg, Germany
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Pieter A. J. Brama
- School of Veterinary Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Gerjo J. V. M. van Osch
- Department of Orthopedics and Sports Medicine, Erasmus MC, University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Otorhinolaryngology, Erasmus MC, University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Biomechanical Engineering, Delft University of Technology, 2628 CD Delft, The Netherlands
- Correspondence: ; Tel.: +31-107043661
| |
Collapse
|
12
|
Kim J, Kim Y, Song S. One-Step Preparation of an Injectable Hydrogel Scaffold System Capable of Sequential Dual-Growth Factor Release to Maximize Bone Regeneration. Adv Healthc Mater 2023; 12:e2202401. [PMID: 36453668 PMCID: PMC11468681 DOI: 10.1002/adhm.202202401] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/19/2022] [Revised: 11/28/2022] [Indexed: 12/02/2022]
Abstract
Numerous growth factors are involved in the natural bone healing process, which is precisely controlled in a time- and concentration-dependent manner. Mimicking the secretion pattern of growth factors could be an effective means to maximize the bone regeneration effect. However, achieving the sequential delivery of various growth factors without the use of multiple materials or complex scaffold designs is challenging. Herein, an injectable poly(organophosphazene) hydrogel scaffold (IPS) encapsulating bone morphogenetic protein (BMP)-2 and TGFβ-1 (IPS_BT) is studied to mimic the sequential secretion of growth factors involved in natural bone healing. The IPS_BT system is designed to release TGFβ-1 slowly while retaining BMP-2 for a longer period of time. When IPS_BT is injected in vivo, the hydrogel is replaced by bone tissue. In addition, angiogenic (CD31 and alpha-smooth muscle actin (α-SMA)) and stemness (Nanog and SOX2) markers are highly upregulated in the early stages of bone regeneration. The IPS system developed here has promising applications in tissue engineering because 1) various amounts of the growth factors can be loaded in one step, 2) the release pattern of each growth factor can be controlled via differences in their molecular interactions, and 3) the injected IPS can be degraded and replaced with regenerated bone tissue.
Collapse
Affiliation(s)
- Jun Kim
- Center for BiomaterialsBiomedical Research InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Division of Bio‐Medical Science and TechnologyKIST SchoolKorea University of Science and TechnologySeoul02792Republic of Korea
| | - Young‐Min Kim
- Center for BiomaterialsBiomedical Research InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Division of Bio‐Medical Science and TechnologyKIST SchoolKorea University of Science and TechnologySeoul02792Republic of Korea
| | - Soo‐Chang Song
- Center for BiomaterialsBiomedical Research InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Division of Bio‐Medical Science and TechnologyKIST SchoolKorea University of Science and TechnologySeoul02792Republic of Korea
- Nexgel Biotech, Co., Ltd.Seoul02792Republic of Korea
| |
Collapse
|
13
|
Gan Q, Chen L, Bei HP, Ng SW, Guo H, Liu G, Pan H, Liu C, Zhao X, Zheng Z. Artificial cilia for soft and stable surface covalent immobilization of bone morphogenetic protein-2. Bioact Mater 2023; 24:551-562. [PMID: 36714333 PMCID: PMC9845954 DOI: 10.1016/j.bioactmat.2022.12.029] [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] [Academic Contribution Register] [Received: 08/30/2022] [Revised: 12/29/2022] [Accepted: 12/29/2022] [Indexed: 01/14/2023] Open
Abstract
Preservation of growth factor sensitivity and bioactivity (e.g., bone morphogenetic protein-2 (BMP-2)) post-immobilization to tissue engineering scaffolds remains a great challenge. Here, we develop a stable and soft surface modification strategy to address this issue. BMP-2 (a model growth factor) is covalently immobilized onto homogeneous poly (glycidyl methacrylate) (PGMA) polymer brushes which are grafted onto substrate surfaces (Au, quartz glass, silica wafer, or common biomaterials) via surface-initiated atom transfer radical polymerization. This surface modification method multiplies the functionalized interfacial area; it is simple, fast, gentle, and has little effect on the loaded protein owing to the cilia motility. The immobilized BMP-2 (i-BMP-2) on the surface of homogeneous PGMA polymer brushes exhibits excellent bioactivity (⁓87% bioactivity of free BMP-2 in vitro and 20%-50% higher than scaffolds with free BMP-2 in vivo), with conformation and secondary structure well-preserved after covalent immobilization and ethanol sterilization. Moreover, the osteogenic activity of i-BMP-2 on the nanoline pattern (PGMA-poly (N-isopropylacrylamide)) shows ⁓110% bioactivity of free BMP-2. This is superior compared to conventional protein covalent immobilization strategies in terms of both bioactivity preservation and therapeutic efficacy. PGMA polymer brushes can be used to modify surfaces of different tissue-engineered scaffolds, which facilitates in situ immobilization of growth factors, and accelerates repair of a wide range of tissue types.
Collapse
Affiliation(s)
- Qi Gan
- Laboratory for Advanced Interfacial Materials and Devices, School of Fashion and Textiles, The Hong Kong Polytechnic University, Kowloon, 99077, Hong Kong Special Administrative Region of China,Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Lina Chen
- Laboratory for Advanced Interfacial Materials and Devices, School of Fashion and Textiles, The Hong Kong Polytechnic University, Kowloon, 99077, Hong Kong Special Administrative Region of China
| | - Ho-Pan Bei
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong Special Administrative Region of China
| | - Sze-Wing Ng
- Laboratory for Advanced Interfacial Materials and Devices, School of Fashion and Textiles, The Hong Kong Polytechnic University, Kowloon, 99077, Hong Kong Special Administrative Region of China
| | - Han Guo
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201204, PR China
| | - Guoqiang Liu
- Laboratory for Advanced Interfacial Materials and Devices, School of Fashion and Textiles, The Hong Kong Polytechnic University, Kowloon, 99077, Hong Kong Special Administrative Region of China
| | - Hao Pan
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China,Corresponding author.
| | - Xin Zhao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong Special Administrative Region of China,Corresponding author.
| | - Zijian Zheng
- Laboratory for Advanced Interfacial Materials and Devices, School of Fashion and Textiles, The Hong Kong Polytechnic University, Kowloon, 99077, Hong Kong Special Administrative Region of China,Department of Applied Biology and Chemical Technology, Faculty of Science, The Hong Kong Polytechnic University, Kowloon, 99077, Hong Kong Special Administrative Region of China,Corresponding author. Laboratory for Advanced Interfacial Materials and Devices, School of Fashion and Textiles, The Hong Kong Polytechnic University, Kowloon, 99077, Hong Kong Special Administrative Region of China.
| |
Collapse
|
14
|
Chen M, Chen Y, Wei C. Nanoparticles based composite coatings with tunable vascular endothelial growth factor and bone morphogenetic protein-2 release for bone regeneration. J Biomed Mater Res A 2022; 111:1044-1053. [PMID: 36565172 DOI: 10.1002/jbm.a.37489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/13/2022] [Revised: 12/06/2022] [Accepted: 12/14/2022] [Indexed: 12/25/2022]
Abstract
Bone healing is a complex cascade involving precisely coordinated spatiotemporal presentation of multiple growth factors (GFs), including osteogenic and angiogenic GFs, and each stage of bone healing requires varying types and content of GFs. In this study, we fabricated a composite nanocoating with tunable vascular endothelial growth factor (VEGF) and bone morphogenetic protein-2 (BMP-2) that was coated on the surface of a polydopamine (PDA)-decorated tertiary calcium phosphate (TCP) scaffold using VEGF-loaded chitosan/bovine serum albumin nanoparticles (CS/BSA-NPs) and BMP-2-loaded poly-L-lysine/oxidized alginate nanoparticles (PLL/OALG-NPs). It was found that VEGF could be efficiently released to promote vascularization in early bone repair stages due to the rapid biodegradation of CS/BSA-NPs, while bone formation can be promoted by a sustained release of BMP-2 from the slowly degrading PLL/OALG-NPs. The composite coating and TCP scaffold can be conjugated due to the excellent adhesive property of PDA. The composite coating can achieve the rapid release of VEGF and sustained release of BMP-2, which can activate GFs for accelerating bone healing.
Collapse
Affiliation(s)
- Mingcong Chen
- Department of Orthopaedics and Traumatology, Shenzhen University General Hospital, Shenzhen, China
| | - Yang Chen
- Department of Surgery, First People's Hospital of Foshan, Foshan, China
| | - Cheng Wei
- Department of Orthopaedics and Traumatology, Shenzhen University General Hospital, Shenzhen, China
| |
Collapse
|
15
|
Jia Y, Wang G, Yan W, Kong B, Xu Y, Wang C, Tang D, Xi X. Psoralen suppresses the phosphorylation of amyloid precursor protein (APP) to inhibit myelosuppression. Biomed Pharmacother 2022; 153:113381. [DOI: 10.1016/j.biopha.2022.113381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/14/2022] [Revised: 06/26/2022] [Accepted: 07/06/2022] [Indexed: 11/26/2022] Open
|
16
|
Wu S, Chen Z, Yu X, Duan X, Chen J, Liu G, Gong M, Xing F, Sun J, Huang S, Zhou X. A sustained release of BMP2 in urine-derived stem cells enhances the osteogenic differentiation and the potential of bone regeneration. Regen Biomater 2022; 9:rbac015. [PMID: 35529046 PMCID: PMC9070791 DOI: 10.1093/rb/rbac015] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/20/2021] [Revised: 01/25/2022] [Accepted: 01/30/2022] [Indexed: 02/05/2023] Open
Abstract
Cell-based tissue engineering is one of the optimistic approaches to replace current treatments for bone defects. Urine-derived stem cells (USCs) are obtained non-invasively and become one of the promising seed cells for bone regeneration. An injectable BMP2-releasing chitosan microspheres/type I collagen hydrogel (BMP2-CSM/Col I hydrogel) was fabricated. USCs proliferated in a time-dependent fashion, spread with good extension and interconnected with each other in different hydrogels both for 2D and 3D models. BMP2 was released in a sustained mode for more than 28 days. Sustained-released BMP2 increased the ALP activities and mineral depositions of USCs in 2D culture, and enhanced the expression of osteogenic genes and proteins in 3D culture. In vivo, the mixture of USCs and BMP2-CSM/Col I hydrogels effectively enhanced bone regeneration, and the ratio of new bone volume to total bone volume was 38% after 8 weeks of implantation. Our results suggested that BMP2-CSM/Col I hydrogels promoted osteogenic differentiation of USCs in 2D and 3D culture in vitro and USCs provided a promising cell source for bone tissue engineering in vivo. As such, USCs-seeded hydrogel scaffolds are regarded as an alternative approach in the repair of bone defects.
Collapse
Affiliation(s)
- Shuang Wu
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Zhao Chen
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Xi Yu
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Xin Duan
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Jialei Chen
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Guoming Liu
- Department of Orthopedics, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Min Gong
- Department of Orthopedics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610000, China
| | - Fei Xing
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Jiachen Sun
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Shishu Huang
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Xiang Zhou
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610000, China
| |
Collapse
|
17
|
Łukowicz K, Zagrajczuk B, Truchan K, Niedzwiedzki Ł, Cholewa-Kowalska K, Osyczka AM. Chemical Compounds Released from Specific Osteoinductive Bioactive Materials Stimulate Human Bone Marrow Mesenchymal Stem Cell Migration. Int J Mol Sci 2022; 23:ijms23052598. [PMID: 35269740 PMCID: PMC8909964 DOI: 10.3390/ijms23052598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/20/2022] [Revised: 02/15/2022] [Accepted: 02/23/2022] [Indexed: 01/12/2023] Open
Abstract
In this work, a poly(L-lactide-co-glycolide) (PLGA)-based composite was enriched with one of the following sol-gel bioactive glasses (SBG) at 50 wt.%: A1—40 mol% SiO2, 60 mol% CaO, CaO/SiO2 ratio of 1.50; S1—80 mol% SiO2, 20 mol% CaO, CaO/SiO2 ratio of 0.25; A2—40 mol% SiO2, 54 mol% CaO, 6 mol% P2O5, CaO/SiO2 ratio of 1.35; S2—80 mol% SiO2,16 mol% CaO, 4 mol% P2O5, CaO/SiO2 ratio of 0.20. The composites and PLGA control sheets were then soaked for 24 h in culture media, and the obtained condition media (CM) were used to treat human bone marrow stromal cells (hBMSCs) for 72 h. All CMs from the composites increased ERK 1/2 activity vs. the control PLGA CM. However, expressions of cell migration-related c-Fos, osteopontin, matrix metalloproteinase-2, C-X-C chemokine receptor type 4, vascular endothelial growth factor, and bone morphogenetic protein 2 were significantly increased only in cells treated with the CM from the A1/PLGA composite. This CM also significantly increased the rate of human BMSC migration but did not affect cell metabolic activity. These results indicate important biological markers that are upregulated by products released from the bioactive composites of a specific chemical composition, which may eventually prompt osteoprogenitor cells to colonize the bioactive material and accelerate the process of tissue regeneration.
Collapse
Affiliation(s)
- Krzysztof Łukowicz
- Department Biology and Cell Imaging, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland; (K.Ł.); (K.T.)
| | - Barbara Zagrajczuk
- Department of Glass Technology and Amorphous Coatings, Faculty of Materials Science and Ceramics, AGH University of Science and Technology, Mickiewicza Ave. 30, 30-059 Krakow, Poland; (B.Z.); (K.C.-K.)
| | - Karolina Truchan
- Department Biology and Cell Imaging, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland; (K.Ł.); (K.T.)
| | - Łukasz Niedzwiedzki
- Department of Orthopedics and Physiotherapy, Faculty of Health Sciences, Jagiellonian University Medical College, Kopernika 19e, 31-501 Krakow, Poland;
| | - Katarzyna Cholewa-Kowalska
- Department of Glass Technology and Amorphous Coatings, Faculty of Materials Science and Ceramics, AGH University of Science and Technology, Mickiewicza Ave. 30, 30-059 Krakow, Poland; (B.Z.); (K.C.-K.)
| | - Anna M. Osyczka
- Department Biology and Cell Imaging, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland; (K.Ł.); (K.T.)
- Correspondence:
| |
Collapse
|
18
|
Abstract
Tumorigenesis is a long-term and multistage process that often leads to the formation of metastases. During this pathological course, two major events appear to be crucial: primary tumour growth and metastatic expansion. In this context, despite research and clinical advances during the past decades, bone cancers remain a leading cause of death worldwide among paediatric cancer patients. Osteosarcomas are the most common malignant bone tumours in children and adolescents. Notwithstanding advances in therapeutic treatments, many patients succumb to these diseases. In particular, less than 30% of patients who demonstrate metastases at diagnosis or are poor responders to chemotherapy survive 5 years after initial diagnosis. LIM kinases (LIMKs), comprising LIMK1 and LIMK2, are common downstream effectors of several signalization pathways, and function as a signalling node that controls cytoskeleton dynamics through the phosphorylation of the cofilin family proteins. In recent decades, several reports have indicated that the functions of LIMKs are mainly implicated in the regulation of actin microfilament and the control of microtubule dynamics. Previous studies have thus identified LIMKs as cancer-promoting regulators in multiple organ cancers, such as breast cancer or prostate cancer. This review updates the current understanding of LIMK involvement in osteosarcoma progression.
Collapse
|
19
|
Aghali A. Craniofacial Bone Tissue Engineering: Current Approaches and Potential Therapy. Cells 2021; 10:cells10112993. [PMID: 34831216 PMCID: PMC8616509 DOI: 10.3390/cells10112993] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/27/2021] [Revised: 10/16/2021] [Accepted: 10/22/2021] [Indexed: 01/10/2023] Open
Abstract
Craniofacial bone defects can result from various disorders, including congenital malformations, tumor resection, infection, severe trauma, and accidents. Successfully regenerating cranial defects is an integral step to restore craniofacial function. However, challenges managing and controlling new bone tissue formation remain. Current advances in tissue engineering and regenerative medicine use innovative techniques to address these challenges. The use of biomaterials, stromal cells, and growth factors have demonstrated promising outcomes in vitro and in vivo. Natural and synthetic bone grafts combined with Mesenchymal Stromal Cells (MSCs) and growth factors have shown encouraging results in regenerating critical-size cranial defects. One of prevalent growth factors is Bone Morphogenetic Protein-2 (BMP-2). BMP-2 is defined as a gold standard growth factor that enhances new bone formation in vitro and in vivo. Recently, emerging evidence suggested that Megakaryocytes (MKs), induced by Thrombopoietin (TPO), show an increase in osteoblast proliferation in vitro and bone mass in vivo. Furthermore, a co-culture study shows mature MKs enhance MSC survival rate while maintaining their phenotype. Therefore, MKs can provide an insight as a potential therapy offering a safe and effective approach to regenerating critical-size cranial defects.
Collapse
Affiliation(s)
- Arbi Aghali
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA;
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47908, USA
| |
Collapse
|
20
|
Sun T, Meng C, Ding Q, Yu K, Zhang X, Zhang W, Tian W, Zhang Q, Guo X, Wu B, Xiong Z. In situ bone regeneration with sequential delivery of aptamer and BMP2 from an ECM-based scaffold fabricated by cryogenic free-form extrusion. Bioact Mater 2021; 6:4163-4175. [PMID: 33997500 PMCID: PMC8099605 DOI: 10.1016/j.bioactmat.2021.04.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/13/2020] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 12/11/2022] Open
Abstract
In situ tissue engineering is a powerful strategy for the treatment of bone defects. It could overcome the limitations of traditional bone tissue engineering, which typically involves extensive cell expansion steps, low cell survival rates upon transplantation, and a risk of immuno-rejection. Here, a porous scaffold polycaprolactone (PCL)/decellularized small intestine submucosa (SIS) was fabricated via cryogenic free-form extrusion, followed by surface modification with aptamer and PlGF-2123-144*-fused BMP2 (pBMP2). The two bioactive molecules were delivered sequentially. The aptamer Apt19s, which exhibited binding affinity to bone marrow-derived mesenchymal stem cells (BMSCs), was quickly released, facilitating the mobilization and recruitment of host BMSCs. BMP2 fused with a PlGF-2123-144 peptide, which showed "super-affinity" to the ECM matrix, was released in a slow and sustained manner, inducing BMSC osteogenic differentiation. In vitro results showed that the sequential release of PCL/SIS-pBMP2-Apt19s promoted cell migration, proliferation, alkaline phosphatase activity, and mRNA expression of osteogenesis-related genes. The in vivo results demonstrated that the sequential release system of PCL/SIS-pBMP2-Apt19s evidently increased bone formation in rat calvarial critical-sized defects compared to the sequential release system of PCL/SIS-BMP2-Apt19s. Thus, the novel delivery system shows potential as an ideal alternative for achieving cell-free scaffold-based bone regeneration in situ.
Collapse
Key Words
- 3D, three-dimensional
- Apt19s, aptamer 19s
- Aptamer
- BMD, bone mineral density
- BMP2
- BMP2, bone morphogenic protein 2
- BMSC, bone marrow-derived mesenchymal stem cell
- Bone regeneration in situ
- CLSM, confocal laser scanning microscopy
- CSD, critical-sized calvarial defect
- Cell recruitment
- Controlled delivery
- ECM, decellularized matrix
- FBS, fetal bovine serum
- FDA, US Food and Drug Administration
- FITC, fluorescein isothiocyanate
- FTIR, Fourier transform infrared
- H&E, hematoxylin and eosin
- HA, hydroxyapatite
- PCL, polycaprolactone
- PVDF, polyvinylidene difluoride
- Rh6G, rhodamine 6G
- SIS, small intestine submucosa
- pBMP2, PlGF-2123-144*-fused BMP2
- ssDNA, single-stranded DNA
Collapse
Affiliation(s)
- Tingfang Sun
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chunqing Meng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiuyue Ding
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Keda Yu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xianglin Zhang
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Wancheng Zhang
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Wenqing Tian
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Qi Zhang
- Wuhan Hi-tech Medical Tissue Research Center, Wuhan, 430206, China
| | - Xiaodong Guo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bin Wu
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zekang Xiong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
21
|
Endo K, Horiuchi K, Katano H, Ozeki N, Sakamaki Y, Koga H, Sekiya I. Intra-articular Injection of PDGF-BB Explored in a Novel in Vitro Model Mobilizes Mesenchymal Stem Cells From the Synovium Into Synovial Fluid in Rats. Stem Cell Rev Rep 2021; 17:1768-1779. [PMID: 33772387 DOI: 10.1007/s12015-021-10156-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 03/14/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Drugs that can induce mesenchymal stem cell (MSC) mobilization from synovium into synovial fluid will enable regenerative medicine in joints without use of exogenous MSCs. An in vitro synovial MSC migration model had previously been developed for screening but had problems in practical application. We herein developed a novel in vitro model, explored cytokines for synovial MSC mobilization with this model, and verified whether MSCs in synovial fluid increase following intra-articular injection of the cytokine. METHODS Human synovial MSCs embedded in a mixture of Matrigel and type 1 collagen hydrogel were placed on a culture insert and then put in medium containing migration factor. Of the six cytokines, we identified the one that mobilizes the highest number of MSCs. PDGF-BB or PBS was injected into rat knees, and 48 h later, synovial fluid was collected and cultured. The cells were examined for MSC properties. RESULTS PDGF-BB was the most effective for synovial MSC mobilization among six cytokines. The effect of PDGF-BB was inhibited by a PRGFR inhibitor. Injection of PDGF-BB into rat knees increased colony-forming cells in the synovial fluid. These cells had surface epitopes and multipotency comparable to MSCs and a higher capacity for proliferation, colony formation, and chondrogenesis. CONCLUSIONS This novel in vitro model recapitulated the migration of MSCs from synovium into synovial fluid. Our exploration of cytokines revealed that PDGF-BB strongly induced in vitro synovial MSC migration, while intra-articular injection of PDGF-BB increased in vivo MSC numbers in synovial fluid in rats.
Collapse
Affiliation(s)
- Kentaro Endo
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, 113-8510, Tokyo, Japan
| | - Kiyotaka Horiuchi
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, 113-8510, Tokyo, Japan
| | - Hisako Katano
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, 113-8510, Tokyo, Japan
| | - Nobutake Ozeki
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, 113-8510, Tokyo, Japan
| | - Yuriko Sakamaki
- Research Core Tokyo Medical and Dental University, Tokyo, Japan
| | - Hideyuki Koga
- Department of Joint Surgery and Sports Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ichiro Sekiya
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, 113-8510, Tokyo, Japan.
| |
Collapse
|
22
|
Evaluation of Acellular Dermal Matrix (ADM) as a Scaf-fold for Adipose-Derived Stem Cell Transfer in the Rat Model. World J Plast Surg 2021. [DOI: 10.52547/wjps.10.2.67] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/24/2022] Open
|
23
|
Human Periodontal Ligament Stem Cell-Derived Exosomes Promote Bone Regeneration by Altering MicroRNA Profiles. Stem Cells Int 2020; 2020:8852307. [PMID: 33293963 PMCID: PMC7691010 DOI: 10.1155/2020/8852307] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/17/2020] [Revised: 10/14/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023] Open
Abstract
The role and underlying mechanism of exosomes derived from human periodontal ligament stem cells (PDLSC) in osteogenesis are unclear. In the present study, we identified the exosomes derived from PDLSCs and found that osteogenic induction can enhance the osteogenic ability of PDLSC-derived exosomes in promoting the osteogenic differentiation of rat bone marrow stem cells (BMSCs). To investigate the underlying mechanism, we analyzed the exosomal miRNA expression profiles of undifferentiated and osteogenic differentiated PDLSCs by RNA sequencing. The results showed that seventy-two miRNAs were upregulated and thirty-five miRNAs were downregulated after osteogenic induction. The results of Gene Ontology analysis and pathway analysis demonstrated that the target genes of differentially expressed exosomal miRNAs participate in the regulation of a variety of biological processes, such as catalytic activity, protein binding, metabolic processes, cell development, and differentiation, and are enriched in osteogenic differentiation-related pathways, such as MAPK signaling, AMPK signaling, and insulin signaling pathways. Our results reveal for the first time that the exosomal miRNAs derived from osteogenic differentiated PDLSCs may promote the osteogenic differentiation of BMSCs, which provides a basis for further research on the regulatory function of exosomal miRNA of PDLSCs during osteogenesis.
Collapse
|
24
|
Xue Y, Li Z, Wang Y, Zhu X, Hu R, Xu W. Role of the HIF‑1α/SDF‑1/CXCR4 signaling axis in accelerated fracture healing after craniocerebral injury. Mol Med Rep 2020; 22:2767-2774. [PMID: 32945380 PMCID: PMC7453606 DOI: 10.3892/mmr.2020.11361] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/16/2019] [Accepted: 06/22/2020] [Indexed: 12/21/2022] Open
Abstract
The hypoxic state of the brain tissue surrounding craniocerebral injury induces an increase in the secretion of HIF-1α during the healing process. HIF-1α can promote mesenchymal stem cell (MSC) migration to ischemic and hypoxic sites by regulating the expression levels of molecules such as stromal cell-derived factor-1 (SDF-1) in the microenvironment. Stem cells express the SDF-1 receptor C-X-C chemokine receptor type 4 (CXCR4) and serve a key role in tissue repair, as well as a number of physiological and pathological processes. The present study aimed to determine the role of HIF-1α/SDF-1/CXCR4 signaling in the process of accelerated fracture healing during craniocerebral injury. Cultured MSCs underwent HIF-1α knockdown to elucidate its effect on the proliferative ability of MSCs, and the effect of SDF-1 in MSCs was investigated. It was also determined whether HIF-1α could promote osteogenesis via SDF-1/CXCR4 signaling and recruit MSCs. The results indicated that HIF-1α knockdown suppressed MSC proliferation in vitro, and SDF-1 promoted cell migration via binding to CXCR4. Furthermore, HIF-1α knockdown inhibited MSC migration via SDF-1/CXCR4 signaling. Considering the wide distribution and diversity of roles of SDF-1 and CXCR4, the present results may form a basis for the development of novel strategies for the treatment of craniocerebral injury.
Collapse
Affiliation(s)
- Yonghua Xue
- Department of Neurosurgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Zhikun Li
- Department of Orthopedic Surgery, Tongren Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200331, P.R. China
| | - Yi Wang
- Department of Orthopedic Surgery, Tongren Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200331, P.R. China
| | - Xiaodong Zhu
- Department of Orthopedic Surgery, Tongren Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200331, P.R. China
| | - Ruixi Hu
- Department of Orthopedic Surgery, Tongren Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200331, P.R. China
| | - Wei Xu
- Department of Orthopedic Surgery, Tongren Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200331, P.R. China
| |
Collapse
|
25
|
Zimmermann A, Hercher D, Regner B, Frischer A, Sperger S, Redl H, Hacobian A. Evaluation of BMP-2 Minicircle DNA for Enhanced Bone Engineering and Regeneration. Curr Gene Ther 2020; 20:55-63. [PMID: 32338217 DOI: 10.2174/1566523220666200427121350] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/05/2020] [Revised: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND To date, the significant osteoinductive potential of bone morphogenetic protein 2 (BMP-2) non-viral gene therapy cannot be fully exploited therapeutically. This is mainly due to weak gene delivery and brief expression peaks restricting the therapeutic effect. OBJECTIVE Our objective was to test the application of minicircle DNA, allowing prolonged expression potential. It offers notable advantages over conventional plasmid DNA. The lack of bacterial sequences and the resulting reduction in size, enables safe usage and improved performance for tissue regeneration. METHODS We inserted an optimized BMP-2 gene cassette with minicircle plasmid technology. BMP-2 minicircle plasmids were produced in E. coli yielding plasmids lacking bacterial backbone elements. Comparative studies of these BMP-2 minicircles and conventional BMP-2 plasmids were performed in vitro in cell systems, including bone marrow derived stem cells. Tests performed included gene expression profiles and cell differentiation assays. RESULTS A C2C12 cell line transfected with the BMP-2-Advanced minicircle showed significantly elevated expression of osteocalcin, alkaline phosphatase (ALP) activity, and BMP-2 protein amount when compared to cells transfected with conventional BMP-2-Advanced plasmid. Furthermore, the plasmids show suitability for stem cell approaches by showing significantly higher levels of ALP activity and mineralization when introduced into human bone marrow stem cells (BMSCs). CONCLUSION We have designed a highly bioactive BMP-2 minicircle plasmid with the potential to fulfil clinical requirements for non-viral gene therapy in the field of bone regeneration.
Collapse
Affiliation(s)
- Alice Zimmermann
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - David Hercher
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - Benedikt Regner
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - Amelie Frischer
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - Simon Sperger
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - Ara Hacobian
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| |
Collapse
|
26
|
Cheng Y, Qiao Y, Shen P, Gao B, Liu X, Kong X, Zhang S, Wu J. Fabrication and in vitro biological activity of functional pH-sensitive double-layered nanoparticles for dental implant application. J Biomater Appl 2020; 34:1409-1421. [PMID: 32054386 DOI: 10.1177/0885328220903615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/19/2022]
Affiliation(s)
- Yicheng Cheng
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, China
- Department of Stomatology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Youbei Qiao
- Department of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Peng Shen
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, China
- Department of Stomatology, Clinical department of Aerospace City, Northern Beijing Medical District, Chinese PLA General Hospital, Beijing, China
| | - Bo Gao
- Department of Stomatology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xianghui Liu
- Department of Stomatology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiangwei Kong
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Shaofeng Zhang
- Department of Stomatology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jiang Wu
- Department of Stomatology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
27
|
Li Z, Liu S, Fu T, Peng Y, Zhang J. Microtubule destabilization caused by silicate via HDAC6 activation contributes to autophagic dysfunction in bone mesenchymal stem cells. Stem Cell Res Ther 2019; 10:351. [PMID: 31775910 PMCID: PMC6880487 DOI: 10.1186/s13287-019-1441-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/11/2019] [Revised: 09/01/2019] [Accepted: 10/03/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Silicon-modified biomaterials have been extensively studied in bone tissue engineering. In recent years, the toxicity of silicon-doped biomaterials has gradually attracted attention but requires further elucidation. This study was designed to explore whether high-dose silicate can induce a cytotoxicity effect in bone mesenchymal stem cells (BMSCs) and the role of autophagy in its cytotoxicity and mechanism. METHODS Morphologic changes and cell viability of BMSCs were detected after different doses of silicate exposure. Autophagic proteins (LC3, p62), LC3 turnover assay, and RFP-GFP-LC3 assay were applied to detect the changes of autophagic flux following silicate treatment. Furthermore, to identify the potential mechanism of autophagic dysfunction, we tested the acetyl-α-tubulin protein level and histone deacetylase 6 (HDAC6) activity after high-dose silicate exposure as well as the changes in microtubule and autophagic activity after HDAC6 siRNA was applied. RESULTS It was found that a high dose of silicate could induce a decrease in cell viability; LC3-II and p62 simultaneously increased after high-dose silicate exposure. A high concentration of silicate could induce autophagic dysfunction and cause autophagosomes to accumulate via microtubule destabilization. Results showed that acetyl-α-tubulin decreased significantly with high-dose silicate treatment, and inhibition of HDAC6 activity can restore microtubule structure and autophagic flux. CONCLUSIONS Microtubule destabilization caused by a high concentration of silicate via HDAC6 activation contributed to autophagic dysfunction in BMSCs, and inhibition of HDAC6 exerted a cytoprotection effect through restoration of the microtubule structure and autophagic flux.
Collapse
Affiliation(s)
- Zheng Li
- Department of Orthopaedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai, China
| | - Shuhao Liu
- Department of Orthopaedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai, China
| | - Tengfei Fu
- Department of Orthopaedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai, China
| | - Yi Peng
- Department of Orthopaedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai, China
| | - Jian Zhang
- Department of Orthopaedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai, China.
| |
Collapse
|
28
|
Kim SY, Bae EB, Huh JW, Ahn JJ, Bae HY, Cho WT, Huh JB. Bone Regeneration Using a Three-Dimensional Hexahedron Channeled BCP Block Combined with Bone Morphogenic Protein-2 in Rat Calvarial Defects. MATERIALS 2019; 12:ma12152435. [PMID: 31370160 PMCID: PMC6696350 DOI: 10.3390/ma12152435] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 07/03/2019] [Revised: 07/25/2019] [Accepted: 07/29/2019] [Indexed: 12/14/2022]
Abstract
It is important to obtain sufficient bone mass before implant placement on alveolar bone, and synthetic bone such as biphasic calcium phosphate (BCP) has been studied to secure this. This study used a BCP block bone with a specific structure of the three-dimensional (3D) hexahedron channel and coating with recombinant human bone morphogenetic protein-2 (rhBMP-2) impregnated carboxymethyl cellulose (CMC) was used to examine the enhancement of bone regeneration of this biomaterial in rat calvarial defect. After the preparation of critical-size calvarial defects in fifteen rats, defects were divided into three groups and were implanted with the assigned specimen (n = 5): Boneplant (untreated 3D hexahedron channeled BCP block), Boneplant/CMC (3D hexahedron channeled BCP block coated with CMC), and Boneplant/CMC/BMP (3D hexahedron channeled BCP block coated with CMC containing rhBMP-2). After 4 weeks, the volumetric, histologic, and histometric analyses were conducted to measure the newly formed bone. Histologically, defects in the Boneplant/CMC/BMP group were almost completely filled with new bone compared to the Boneplant and Boneplant/CMC groups. The new bone volume (P < 0.05) and area (P < 0.001) in the Boneplant/CMC/BMP group (20.12% ± 2.17, 33.79% ± 3.66) were much greater than those in the Boneplant (10.77% ± 4.8, 16.48% ± 9.11) and Boneplant/CMC (10.72% ± 3.29, 16.57% ± 8.94) groups, respectively. In conclusion, the 3D hexahedron channeled BCP block adapted rhBMP-2 with carrier CMC showed high possibility as an effective bone graft material.
Collapse
Affiliation(s)
- So-Yeun Kim
- Department of Prosthodontics, Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Korea
| | - Eun-Bin Bae
- Department of Prosthodontics, Dental Research Institute, Dental and Life Science Institute, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Jae-Woong Huh
- Department of Prosthodontics, Dental Research Institute, Dental and Life Science Institute, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Seroun Dental Clinic, Suyeong-ro, Nam-gu, Busan 48445, Korea
| | - Jong-Ju Ahn
- Department of Prosthodontics, Dental Research Institute, Dental and Life Science Institute, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Hyun-Young Bae
- Department of Prosthodontics, Dental Research Institute, Dental and Life Science Institute, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Won-Tak Cho
- Department of Prosthodontics, Dental Research Institute, Dental and Life Science Institute, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Jung-Bo Huh
- Department of Prosthodontics, Dental Research Institute, Dental and Life Science Institute, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea.
| |
Collapse
|
29
|
Gadalla D, Goldstein AS. Improving the Osteogenicity of PCL Fiber Substrates by Surface-Immobilization of Bone Morphogenic Protein-2. Ann Biomed Eng 2019; 48:1006-1015. [PMID: 31115719 DOI: 10.1007/s10439-019-02286-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/31/2019] [Accepted: 05/07/2019] [Indexed: 12/16/2022]
Abstract
Polycaprolactone (PCL) fiber scaffolds are attractive, albeit inert, substrates for ligament regeneration, that may be improved by incorporating trophic factors to guide tissue remodeling in vivo. In particular, immobilization of bone morphogenic protein-2 (BMP-2) to the scaffold surface may facilitate rapid and robust integration of the scaffold with adjacent bone tissues. As a first step toward testing this, model PCL surfaces were modified by the addition of heparin (Hep) and BMP-2 to facilitate osteoblastic differentiation. Specifically, Hep was combined with PCL at 0, 0.5, and 1 wt% (denoted as PCL, PCL-0.5Hep, and PCL-1Hep), cast into films, and then BMP-2 was immobilized to surfaces by either adsorption and covalent conjugation. Here, BMP-2 concentration increased systematically with incorporation of Hep, and higher concentrations were achieved by covalent conjugation. Next, blends were electrospun to form thin meshes with fiber diameters of 0.92, 0.62, and 0.54 μm for PCL, PCL-0.5Hep, and PCL-1Hep, respectively. Mesenchymal stem cells (MSCs) had no difficulty attaching to and proliferating on all meshes. Lastly, PCL-1Hep meshes were prepared with adsorbed or covalently conjugated BMP-2 and cultured with MSCs in the absence of osteogenic factors. Under these conditions, alkaline phosphatase activity and deposition of bone sialoprotein, osteopontin, and calcium minerals-markers of osteoblastic differentiation-were significantly higher on surfaces with immobilized BMP-2. Together, these data indicate that covalent immobilization of trophic factors confers bioactivity to scaffolds, which may be applied in a spatially controlled manner for ligament regeneration and bone integration.
Collapse
Affiliation(s)
- Dina Gadalla
- Department of Chemical Engineering, Virginia Tech, Suite 245 Goodwin Hall, 635 Prices Fork Road, Blacksburg, VA, 24061-0211, USA
| | - Aaron S Goldstein
- Department of Chemical Engineering, Virginia Tech, Suite 245 Goodwin Hall, 635 Prices Fork Road, Blacksburg, VA, 24061-0211, USA.
- School of Biomedical Engineering and Sciences, Virginia Tech, 235 Kelly Hall, 325 Stanger Street, Blacksburg, VA, 24061-0298, USA.
| |
Collapse
|