1
|
Yuan X, Wu T, Lu T, Ye J. Osteogenesis Differentiation and Molecular Mechanism Study of a Si and Mg Dual-Ion System Based on mRNA Transcriptomic Sequencing Analysis. ACS APPLIED BIO MATERIALS 2025; 8:2482-2491. [PMID: 39985471 DOI: 10.1021/acsabm.4c01937] [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: 02/24/2025]
Abstract
Both silicon (Si) and magnesium (Mg) ions play essential roles in bone health. However, the precise mechanisms by which these two ions enhance osteogenic differentiation remain to be fully elucidated. Herein, a Si-Mg dual-ion system was designed to investigate the effects of Si and Mg ions on the cytological behavior of mouse bone marrow mesenchymal stem cells (mBMSCs). The molecular mechanism of the Si-Mg dual-ion system regulating osteogenic differentiation of mBMSCs was investigated by transcriptome sequencing technology. In the single-ion system, the Si group with concentrations of 1.5 and 0.75 mM exhibited good combined effects (cell proliferation, alkaline phosphatase (ALP) activity, and osteogenic differentiation gene expression (Runx2, OPN, and Col-I)) of mBMSCs. The Mg group with concentrations of 5 and 2.5 mM showed better combined effects (cell proliferation, ALP activity, and osteogenic differentiation gene expression) of mBMSCs. In the dual-ion system, the silicon (0.75 mM)-magnesium (2.5 mM) experimental group significantly enhanced the proliferation, ALP activity, and osteogenesis-related gene expression (Runx2, OPN, and Col-I) of mBMSCs. The analysis of transcriptome sequencing results showed that Mg ions had a certain pro-stem cell osteogenic differentiation regulatory effect. Si ions had a stronger regulation on osteogenic differentiation than the Mg ions. The regulation of osteogenic differentiation by Si-Mg dual ions was synergistically enhanced compared to that of a single ion. In addition, the transforming growth factor beta (TGF-β) signaling pathway and mitogen-activated protein kinase (MAPK) signaling pathway were involved in mediating the pro-stem cell osteogenic differentiation by Si-Mg dual ions. This study sheds light on investigating the molecular mechanism of dual-ion regulation of the osteogenic differentiation of mBMSCs and enriches the theory of ion-regulating osteogenic differentiation.
Collapse
Affiliation(s)
- Xinyuan Yuan
- School of Materials Science and Engineering and Key Laboratory of Biomedical Materials of Ministry of Education, South China University of Technology, Guangzhou 510641, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou 510006, P. R. China
| | - Tingting Wu
- Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, National Engineering Research Center for Healthcare Devices, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, P. R. China
| | - Teliang Lu
- Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, National Engineering Research Center for Healthcare Devices, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, P. R. China
| | - Jiandong Ye
- School of Materials Science and Engineering and Key Laboratory of Biomedical Materials of Ministry of Education, South China University of Technology, Guangzhou 510641, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou 510006, P. R. China
| |
Collapse
|
2
|
Jang SB, Kim Y, Yeo HC, Kang GH, An BC, Ryu Y, Chung MJ, Cho SG. Probiotic-Derived P8 Protein: Promoting Proliferation and Migration in Stem Cells and Keratinocytes. Int J Stem Cells 2025; 18:87-98. [PMID: 39491493 PMCID: PMC11867908 DOI: 10.15283/ijsc24107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 11/05/2024] Open
Abstract
Probiotics exert various effects on the body and provide different health benefits. Previous reports have demonstrated that the P8 protein (P8), isolated from Lactobacillus rhamnosus, has anticancer properties. However, its efficacy in stem cells and normal cells has not been reported. In this study, the effect of P8 on cell proliferation and wound healing was evaluated, investigating its underlying mechanism. Based on scratch assay results, we demonstrated that P8 treatment significantly increases wound healing by activating the cell cycle and promoting stem cell stemness. Cellular mechanisms were further investigated by culturing stem cells in a medium containing Lactobacillus-derived P8 protein, revealing its promotion of cell proliferation and migration. Also, it is found that P8 enhances the expression of stemness markers, such as OCT4 and SOX2, along with activation of the mitogen-activated protein kinase (MAPK) signaling and Hippo pathways. These results indicate that P8 can promote cell growth by increasing stem cell proliferation, migration, and stemness in a manner associated with MAPK and Hippo signaling, which could contribute to the increased wound healing after P8 treatment. Furthermore, P8 could promote wound healing in keratinocytes by activating the MAPK signaling pathways. These results suggest that P8 might be a promising candidate to enhance stem cell culture efficiency by activating cell proliferation, and enhance therapeutic effects in skin diseases.
Collapse
Affiliation(s)
- Soo Bin Jang
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
| | - Yoojung Kim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
| | - Han Cheol Yeo
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
| | | | | | - Yongku Ryu
- R&D Center, Cell Biotech Co., Ltd., Gimpo, Korea
| | | | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
- R&D Team, StemExOne Co., Ltd., Seoul, Korea
| |
Collapse
|
3
|
Yuan X, Wu T, Lu T, Ye J. Si and Zn dual ions upregulate the osteogenic differentiation of mBMSCs: mRNA transcriptomic sequencing analysis. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2024; 35:54. [PMID: 39251504 PMCID: PMC11383841 DOI: 10.1007/s10856-024-06825-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/10/2024] [Indexed: 09/11/2024]
Abstract
Both silicon (Si) and zinc (Zn) ions are essential elements to bone health and their mechanisms for promoting osteogenesis have aroused the extensive attention of researchers. Thereinto, the mechanism by which dual ions promote osteogenic differentiation remains to be elucidated. Herein, the effects of Si and Zn ions on the cytological behaviors of mBMSCs were firstly studied. Then, the molecular mechanism of Si-Zn dual ions regulating the osteogenic differentiation of mBMSCs was investigated via transcriptome sequencing technology. In the single-ion system, Si ion at the concentration of 1.5 mM (Si-1.5) had better comprehensive effects of cell proliferation, ALP activity and osteogenesis-related gene expression levels (ALP, Runx2, OCN, Col-I and BSP); Zn ion at the concentration of 50 μM (Zn-50) demonstrated better combining effects of cell proliferation, ALP activity and same osteogenic genes expression levels. In the dual-ion system, the Si (1.5 mM)-Zn (50 μM) group (Si1.5-Zn50) synthetically enhanced ALP activity and osteogenesis genes compared with single-ion groups. Analysis of the transcriptome sequencing results showed that Si ion had a certain effect on promoting the osteogenic differentiation of mBMSCs; Zn ion had a stronger effect of contributing to a better osteogenic differentiation of mBMSCs than that of Si ion; the Si-Zn dual ions had a synergistic enhancement on conducting to the osteogenic differentiation of mBMSCs compared to single ion (Si or Zn). This study offers a blueprint for exploring the regulation mechanism of osteogenic differentiation by dual ions.
Collapse
Affiliation(s)
- Xinyuan Yuan
- School of Materials Science and Engineering and Key Laboratory of Biomedical Materials of Ministry of Education, South China University of Technology, Guangzhou, PR China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, PR China
| | - Tingting Wu
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, PR China
| | - Teliang Lu
- School of Materials Science and Engineering and Key Laboratory of Biomedical Materials of Ministry of Education, South China University of Technology, Guangzhou, PR China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, PR China
| | - Jiandong Ye
- School of Materials Science and Engineering and Key Laboratory of Biomedical Materials of Ministry of Education, South China University of Technology, Guangzhou, PR China.
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, PR China.
| |
Collapse
|
4
|
Gao Q, Liu J, Wang M, Liu X, Jiang Y, Su J. Biomaterials regulates BMSCs differentiation via mechanical microenvironment. BIOMATERIALS ADVANCES 2024; 157:213738. [PMID: 38154401 DOI: 10.1016/j.bioadv.2023.213738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/11/2023] [Accepted: 12/16/2023] [Indexed: 12/30/2023]
Abstract
Bone mesenchymal stem cells (BMSCs) are crucial for bone tissue regeneration, the mechanical microenvironment of hard tissues, including bone and teeth, significantly affects the osteogenic differentiation of BMSCs. Biomaterials may mimic the microenvironment of the extracellular matrix and provide mechanical signals to regulate BMSCs differentiation via inducing the secretion of various intracellular factors. Biomaterials direct the differentiation of BMSCs via mechanical signals, including tension, compression, shear, hydrostatic pressure, stiffness, elasticity, and viscoelasticity, which can be transmitted to cells through mechanical signalling pathways. Besides, biomaterials with piezoelectric effects regulate BMSCs differentiation via indirect mechanical signals, such as, electronic signals, which are transformed from mechanical stimuli by piezoelectric biomaterials. Mechanical stimulation facilitates achieving vectored stem cell fate regulation, while understanding the underlying mechanisms remains challenging. Herein, this review summarizes the intracellular factors, including translation factors, epigenetic modifications, and miRNA level, as well as the extracellular factor, including direct and indirect mechanical signals, which regulate the osteogenic differentiation of BMSCs. Besides, this review will also give a comprehensive summary about how mechanical stimuli regulate cellular behaviours, as well as how biomaterials promote the osteogenic differentiation of BMSCs via mechanical microenvironments. The cellular behaviours and activated signal pathways will give more implications for the design of biomaterials with superior properties for bone tissue engineering. Moreover, it will also provide inspiration for the construction of bone organoids which is a useful tool for mimicking in vivo bone tissue microenvironments.
Collapse
Affiliation(s)
- Qianmin Gao
- Institute of Translational Medicine, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; Organoid Research Centre, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; National Centre for Translational Medicine (Shanghai) SHU Branch, NO.333 Nanchen Road, Shanghai University, Shanghai 200444, PR China
| | - Jinlong Liu
- Institute of Translational Medicine, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; Organoid Research Centre, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; National Centre for Translational Medicine (Shanghai) SHU Branch, NO.333 Nanchen Road, Shanghai University, Shanghai 200444, PR China
| | - Mingkai Wang
- Institute of Translational Medicine, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; Organoid Research Centre, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; National Centre for Translational Medicine (Shanghai) SHU Branch, NO.333 Nanchen Road, Shanghai University, Shanghai 200444, PR China
| | - Xiangfei Liu
- Department of Orthopedics, Shanghai Zhongye Hospital, NO. 456 Chunlei Road, Shanghai 200941, PR China.
| | - Yingying Jiang
- Institute of Translational Medicine, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China.
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; Organoid Research Centre, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; National Centre for Translational Medicine (Shanghai) SHU Branch, NO.333 Nanchen Road, Shanghai University, Shanghai 200444, PR China; Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, NO.1665 Kongjiang Road, Shanghai 200092, PR China.
| |
Collapse
|
5
|
Wang X, Liu S, Zhang W, Peng H, Zhang M, Li Y, Guo Q, Wang W, Huang N, Liu L, Liu D. Silicon nanowire array overcomes chemotherapeutic resistance by inducing the differentiation of breast cancer stem cells. J Biomed Mater Res B Appl Biomater 2023. [PMID: 36929288 DOI: 10.1002/jbm.b.35249] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/18/2023] [Accepted: 03/03/2023] [Indexed: 03/18/2023]
Abstract
Currently, traditional cancer treatment strategies are greatly challenged by the existence of cancer stem cells (CSCs), which are root cause of chemotherapy resistance. Differentiation therapy presents a novel therapeutic strategy for CSC-targeted therapy. However, there are very few studies on the induction of CSCs differentiation so far. Silicon nanowire array (SiNWA) with many unique properties is considered to be an excellent material for various applications ranging from biotechnology to biomedical applications. In this study, we report the SiNWA differentiates MCF-7-derived breast CSCs (BCSCs) into non-CSCs by modulating the morphology of cells. In vitro, the differentiated BCSCs lose the stemness properties and thus become sensitive to chemotherapeutic drugs, eventually leading to the death of BCSCs. Therefore, this work suggests a potential approach for overcoming chemotherapeutic resistance.
Collapse
Affiliation(s)
- Xiaotong Wang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, People's Republic of China
| | - Sisi Liu
- Cheng'an County Hospital of Traditional Chinese Medicine, Handan, People's Republic of China
| | - Wei Zhang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, People's Republic of China
| | - Haotong Peng
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, People's Republic of China
| | - Miao Zhang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, People's Republic of China
| | - Yaping Li
- College of Public Health, Hebei University, Baoding, People's Republic of China
| | - Qi Guo
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, People's Republic of China
| | - Wenjing Wang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, People's Republic of China
| | - Na Huang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, People's Republic of China
| | - LiYan Liu
- Medical Comprehensive Experimental Centrer, Hebei University, Baoding, People's Republic of China
| | - Dandan Liu
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, People's Republic of China
| |
Collapse
|
6
|
Zhang X, Chen JY, Pei X, Li YH, Feng H, He ZH, Xie WJ, Pei XB, Zhu Z, Wan QB, Wang J. One-Pot Facile Encapsulation of Dimethyloxallyl Glycine by Nanoscale Zeolitic Imidazolate Frameworks-8 for Enhancing Vascularized Bone Regeneration. Adv Healthc Mater 2023; 12:e2202317. [PMID: 36349826 DOI: 10.1002/adhm.202202317] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/31/2022] [Indexed: 11/10/2022]
Abstract
In the process of bone tissue regeneration, regulation of osteogenesis-angiogenesis coupling is of great importance. Therefore, dimethyloxallyl glycine (DMOG) is loaded by nanoscale zeolitic imidazolate frameworks-8 (ZIF-8) to obtain a drug-loading system that can promote osteogenesis-angiogenesis coupling. Characterization of the drug-loading nanoparticles (DMOG@ZIF-8) reveals that DMOG is successfully loaded into ZIF-8 by two different methods, and the DMOG@ZIF-8 is prepared using the one-pot method (OD@ZIF-8) achieves higher loading efficiency and longer release time than those prepared using the post-loading method (PD@ZIF-8). In vitro studies found that DMOG@ZIF-8 significantly enhances the migration, tube formation, and angiogenesis-related protein secretion of human umbilical vein endothelial cells as well as the extracellular matrix mineralization, alkaline phosphatase activity, and osteogenesis-related protein secretion of bone marrow mesenchymal stem cells. Moreover, OD@ZIF-8 nanoparticles are more efficient than PD@ZIF-8 nanoparticles in induction of osteogenesis-angiogenesis coupling. Then, in vivo cranial critical defect model shows that the addition of OD@ZIF-8 significantly promotes vascularized bone formation as indicated by the results including microcomputed tomographic, histological and immunofluorescence staining, and so on. Taken together, loading ZIF-8 with DMOG may be a promising solution for critical-sized bone defect reconstruction and the one-pot method is preferred in the preparation of such drug-loading system.
Collapse
Affiliation(s)
- Xin Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jun-Yu Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xiang Pei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China
| | - Ya-Hong Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Hao Feng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Zi-Han He
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Wen-Jia Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xi-Bo Pei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Zhou Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Qian-Bing Wan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jian Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, 610041, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
7
|
Han F, Li T, Li M, Zhang B, Wang Y, Zhu Y, Wu C. Nano-calcium silicate mineralized fish scale scaffolds for enhancing tendon-bone healing. Bioact Mater 2023; 20:29-40. [PMID: 35633872 PMCID: PMC9123220 DOI: 10.1016/j.bioactmat.2022.04.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 04/14/2022] [Accepted: 04/26/2022] [Indexed: 11/12/2022] Open
Abstract
Tendon-bone healing is essential for an effective rotator cuff tendon repair surgery, however, this remains a significant challenge due to the lack of biomaterials with high strength and bioactivity. Inspired by the high-performance exoskeleton of natural organisms, we set out to apply natural fish scale (FS) modified by calcium silicate nanoparticles (CS NPs) as a new biomaterial (CS-FS) to overcome the challenge. Benefit from its “Bouligand” microstructure, such FS-based scaffold maintained excellent tensile strength (125.05 MPa) and toughness (14.16 MJ/m3), which are 1.93 and 2.72 times that of natural tendon respectively, allowing it to well meet the requirements for rotator cuff tendon repair. Additionally, CS-FS showed diverse bioactivities by stimulating the differentiation and phenotypic maintenance of multiple types of cells participated into the composition of tendon-bone junction, (e.g. bone marrow mesenchymal stem cells (BMSCs), chondrocyte, and tendon stem/progenitor cells (TSPCs)). In both rat and rabbit rotator cuff tear (RCT) models, CS-FS played a key role in the tendon-bone interface regeneration and biomechanical function, which may be achieved by activating BMP-2/Smad/Runx2 pathway in BMSCs. Therefore, natural fish scale -based biomaterials are the promising candidate for clinical tendon repair due to their outstanding strength and bioactivity. Nano-calcium silicate mineralized fish scale scaffold was first developed for tendon defect repair. •CS-FS exhibited excellent mechanical properties superior to those of natural tendon. •CS-FS showed diverse bioactivities by stimulating the differentiation of multiple types of cells. •CS NPs accelerated tendon-bone interface tendon-bone healing enhancement and biomechanical recovery.
Collapse
|
8
|
Bladder Cancer Cells Exert Pleiotropic Effects on Human Adipose-Derived Stem Cells. Life (Basel) 2022; 12:life12040549. [PMID: 35455040 PMCID: PMC9025060 DOI: 10.3390/life12040549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/29/2022] [Accepted: 04/05/2022] [Indexed: 12/12/2022] Open
Abstract
Stem cell-based therapies are considered one of the most promising disciplines in biomedicine. Bladder cancer patients could benefit from therapies directed to promote healing after invasive surgeries or to lessen urinary incontinence, a common side effect of both cancer itself and the treatment. However, the local delivery of cells producing large amounts of paracrine factors may alter interactions within the microenvironment. For this reason, reconstructive cellular therapies for patients with a history of cancer carry a potential risk of tumor reactivation. We used an indirect co-culture model to characterize the interplay between adipose-derived stem cells and bladder cancer cells. Incubation with ASCs increased MCP-1 secretion by bladder cancer cells (from 2.1-fold to 8.1-fold, depending on the cell line). Cancer cell-derived factors altered ASC morphology. Cells with atypical shapes and significantly enlarged volumes appeared within the monolayer. Incubation in a conditioned medium (CM) containing soluble mediators secreted by 5637 and HB-CLS-1 bladder cancer cell lines decreased ASC numbers by 47.5% and 45.7%. A significant increase in adhesion to ECM components, accompanied by reduced motility and sheet migration, was also observed after incubation in CM from 5637 and HB-CLS-1 cells. No differences were observed when ASCs were co-cultured with HT-1376 cells. Our previous and present results indicate that soluble mediators secreted by ASCs and bladder cancer cells induce opposite effects influencing cells that represent the non-muscle-invasive urinary bladder.
Collapse
|
9
|
Fang H, Zhu D, Yang Q, Chen Y, Zhang C, Gao J, Gao Y. Emerging zero-dimensional to four-dimensional biomaterials for bone regeneration. J Nanobiotechnology 2022; 20:26. [PMID: 34991600 PMCID: PMC8740479 DOI: 10.1186/s12951-021-01228-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/26/2021] [Indexed: 12/17/2022] Open
Abstract
Bone is one of the most sophisticated and dynamic tissues in the human body, and is characterized by its remarkable potential for regeneration. In most cases, bone has the capacity to be restored to its original form with homeostatic functionality after injury without any remaining scarring. Throughout the fascinating processes of bone regeneration, a plethora of cell lineages and signaling molecules, together with the extracellular matrix, are precisely regulated at multiple length and time scales. However, conditions, such as delayed unions (or nonunion) and critical-sized bone defects, represent thorny challenges for orthopedic surgeons. During recent decades, a variety of novel biomaterials have been designed to mimic the organic and inorganic structure of the bone microenvironment, which have tremendously promoted and accelerated bone healing throughout different stages of bone regeneration. Advances in tissue engineering endowed bone scaffolds with phenomenal osteoconductivity, osteoinductivity, vascularization and neurotization effects as well as alluring properties, such as antibacterial effects. According to the dimensional structure and functional mechanism, these biomaterials are categorized as zero-dimensional, one-dimensional, two-dimensional, three-dimensional, and four-dimensional biomaterials. In this review, we comprehensively summarized the astounding advances in emerging biomaterials for bone regeneration by categorizing them as zero-dimensional to four-dimensional biomaterials, which were further elucidated by typical examples. Hopefully, this review will provide some inspiration for the future design of biomaterials for bone tissue engineering.
Collapse
Affiliation(s)
- Haoyu Fang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Daoyu Zhu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Qianhao Yang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yixuan Chen
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Changqing Zhang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Junjie Gao
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Science, Ningbo, Zhejiang, China.
| | - Youshui Gao
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
10
|
Capacitive electrical stimulation of a conducting polymeric thin film induces human mesenchymal stem cell osteogenesis. Biointerphases 2022; 17:011001. [PMID: 34979808 DOI: 10.1116/6.0001435] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Electroactive materials based on conductive polymers are promising options for tissue engineering and regenerative medicine applications. In the present work, the conducting copolymers of poly (3,4-ethylenedioxythiophene) and poly (d, l-lactic acid) (PEDOT-co-PDLLA) with PEDOT:PDLLA molar ratios of 1:50, 1:25, and 1:5 were synthesized and compared to the insulating macromonomer of EDOT-PDLLA as an experimental control. Bone marrow-derived human mesenchymal stem cells (hMSC-BM) were cultured on the copolymers and the macromonomer thin films inside a bioreactor that induced a capacitive electrical stimulation (CES) with an electric field of 100 mV/mm for 2 h per day for 21 days. Under CES, the copolymers exhibited good cell viability and promoted the differentiation from hMSC-BM to osteogenic lineages, revealed by higher mineralization mainly when the contents of conducting segments of PEDOT (i.e., copolymer with 1:25 and 1:5 PEDOT:PDLLA ratios) were increased. The results indicate that the intrinsic electrical conductivity of the substrates is an important key point for the effectiveness of the electric field generated by the CES, intending to promote the differentiation effect for bone cells.
Collapse
|
11
|
Icariin and Icariside II Reciprocally Stimulate Osteogenesis and Inhibit Adipogenesis of Multipotential Stromal Cells through ERK Signaling. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:8069930. [PMID: 34956384 PMCID: PMC8702327 DOI: 10.1155/2021/8069930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 11/17/2022]
Abstract
Herba Epimedii is a famous Chinese herbal medicine for treating bone diseases. Icariin and icariside II, the main chemical constituents, have attracted great attention from scientists for their potential as antiosteoporosis agents. Our study aimed to evaluate their effects on the lineage commitment of multipotential stromal cells (MSCs). The osteogenesis and adipogenesis of MSCs were assessed by ALP activity, calcium deposition, and adipocyte formation. The expression profiles and levels of osteogenic and adipogenic specific genes were evaluated by cDNA microarray and quantitative real-time PCR. The involvement of extracellular signal-regulated kinase (ERK) signaling was studied by enzyme-linked immunosorbent assay. Icariin and icariside II significantly increased ALP activity and mineralization during osteogenic differentiation of MSCs. Runx2, Col1, and Bmp2 were upregulated in the presence of icariin and icariside II. Meanwhile, they downregulated Pparg, Adipsin, and Cebpb expression during adipogenic differentiation. cDNA microarray revealed 57 differentially expressed genes during lineage commitment of MSCs. In addition, icariin and icariside II enhanced the phosphorylation of ERK, and the above biological effects were blocked by ERK inhibitor U0126. Icariin and icariside II may drive the final lineage commitment of MSCs towards osteogenesis and inhibit adipogenesis through the ERK signaling pathway. Both of them exert multiple osteoprotective effects and deserve more attention for their medicinal and healthcare prospects.
Collapse
|
12
|
Song S, Kim KY, Lee SH, Kim KK, Lee K, Lee W, Jeon H, Ko SH. Recent Advances in 1D Nanomaterial‐Based Bioelectronics for Healthcare Applications. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Sangmin Song
- Applied Nano and Thermal Science Lab Department of Mechanical Engineering Seoul National University 1 Gwanak-ro Gwanak-gu Seoul 151-742 Korea
- Center for Biomaterials Biomedical Research Institute Korea Institute of Science and Technology (KIST) 5, Hwarang-ro 14-gil Seongbuk-gu Seoul 02792 Korea
| | - Kyung Yeun Kim
- Applied Nano and Thermal Science Lab Department of Mechanical Engineering Seoul National University 1 Gwanak-ro Gwanak-gu Seoul 151-742 Korea
- Center for Biomaterials Biomedical Research Institute Korea Institute of Science and Technology (KIST) 5, Hwarang-ro 14-gil Seongbuk-gu Seoul 02792 Korea
| | - Sun Hee Lee
- Center for Biomaterials Biomedical Research Institute Korea Institute of Science and Technology (KIST) 5, Hwarang-ro 14-gil Seongbuk-gu Seoul 02792 Korea
| | - Kyun Kyu Kim
- Department of Chemical Engineering Stanford University Stanford CA 94305 USA
| | - Kyungwoo Lee
- Center for Biomaterials Biomedical Research Institute Korea Institute of Science and Technology (KIST) 5, Hwarang-ro 14-gil Seongbuk-gu Seoul 02792 Korea
| | - Wonryung Lee
- Center for Biomaterials Biomedical Research Institute Korea Institute of Science and Technology (KIST) 5, Hwarang-ro 14-gil Seongbuk-gu Seoul 02792 Korea
| | - Hojeong Jeon
- Center for Biomaterials Biomedical Research Institute Korea Institute of Science and Technology (KIST) 5, Hwarang-ro 14-gil Seongbuk-gu Seoul 02792 Korea
- KU-KIST Graduate School of Converging Science and Technology Korea University 145, Anam-ro Seongbuk-gu Seoul 02841 Korea
| | - Seung Hwan Ko
- Applied Nano and Thermal Science Lab Department of Mechanical Engineering Seoul National University 1 Gwanak-ro Gwanak-gu Seoul 151-742 Korea
- Institute of Advanced Machines and Design/Institute of Engineering Research Seoul National University Seoul 08826 Korea
| |
Collapse
|
13
|
El-Habashy S, Eltaher H, Gaballah A, Mehanna R, El-Kamel AH. Biomaterial-Based Nanocomposite for Osteogenic Repurposing of Doxycycline. Int J Nanomedicine 2021; 16:1103-1126. [PMID: 33603371 PMCID: PMC7887185 DOI: 10.2147/ijn.s298297] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 01/20/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Besides its antimicrobial action, doxycycline (DX) has lately been repurposed as a small-molecule drug for osteogenic purposes. However, osteogenic DX application is impeded by its dose-dependent cytotoxicity. Further, high-dose DX impairs cell differentiation and mineralization. PURPOSE Integrating DX into a biomaterial-based delivery system that can control its release would not only ameliorate its cytotoxic actions but also augment its osteogenic activity. In this work, we managed to engineer novel composite DX-hydroxyapatite-polycaprolactone nanoparticles (DX/HAp/PCL) to modify DX osteogenic potential. METHODS Employing a 23-factorial design, we first optimized HApN for surface-area attributes to maximize DX loading. Composite DX/HAp/PCL were then realized using a simple emulsification technique, characterized using various in vitro methods, and evaluated for in vitro osteogenesis. RESULTS The developed HApN exhibited a favorable crystalline structure, Ca:P elemental ratio (1.67), mesoporous nature, and large surface area. DX/HAp/PCL achieved the highest reported entrapment efficiency (94.77%±1.23%) of DX in PCL-based particles. The developed composite system achieved controlled release of the water-soluble DX over 24 days. Moreover, the novel composite nanosystem managed to significantly ameliorate DX cytotoxicity on bone-marrow stem cells, as well as enhance its overall proliferation potential. Alkaline phosphatase and mineralization assays revealed superior osteodifferentiation potential of the composite system. Quantification of gene expression demonstrated that while DX solution was able to drive bone-marrow stem cells down the osteogenic lineage into immature osteoblasts after 10-day culture, the innovative composite system allowed maturation of osteodifferentiated cells. To the best of our knowledge, this is the first work to elaborate the impact of DX on the expression of osteogenic genes: RUNX2, OSP, and BSP. Further, the osteogenicity of a DX-loaded particulate-delivery system has not been previously investigated. CONCLUSION Our findings indicate that repurposing low-dose DX in complementary biomaterial-based nanosystems can offer a prominent osteogenic candidate for bone-regeneration purposes.
Collapse
Affiliation(s)
- Salma El-Habashy
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Hoda Eltaher
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Ahmed Gaballah
- Microbiology Department, Medical Research Institute, Alexandria University, Alexandria, 21561, Egypt
| | - Radwa Mehanna
- Medical Physiology Department, Faculty of Medicine, Alexandria University, Alexandria, 21131, Egypt
- Center of Excellence for Research in Regenerative Medicine and Applications CERRMA, Faculty of Medicine, Alexandria University, Alexandria, 21131, Egypt
| | - Amal H El-Kamel
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| |
Collapse
|
14
|
Elashry MI, Baulig N, Wagner AS, Klymiuk MC, Kruppke B, Hanke T, Wenisch S, Arnhold S. Combined macromolecule biomaterials together with fluid shear stress promote the osteogenic differentiation capacity of equine adipose-derived mesenchymal stem cells. Stem Cell Res Ther 2021; 12:116. [PMID: 33579348 PMCID: PMC7879632 DOI: 10.1186/s13287-021-02146-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 01/06/2021] [Indexed: 11/10/2022] Open
Abstract
Background Combination of mesenchymal stem cells (MSCs) and biomaterials is a rapidly growing approach in regenerative medicine particularly for chronic degenerative disorders including osteoarthritis and osteoporosis. The present study examined the effect of biomaterial scaffolds on equine adipose-derived MSC morphology, viability, adherence, migration, and osteogenic differentiation. Methods MSCs were cultivated in conjunction with collagen CultiSpher-S Microcarrier (MC), nanocomposite xerogels B30 and combined B30 with strontium (B30Str) biomaterials in osteogenic differentiation medium either under static or mechanical fluid shear stress (FSS) culture conditions. The data were generated by histological means, live cell imaging, cell viability, adherence and migration assays, semi-quantification of alkaline phosphatase (ALP) activity, and quantification of the osteogenic markers runt-related transcription factor 2 (Runx2) and alkaline phosphatase (ALP) expression. Results The data revealed that combined mechanical FSS with MC but not B30 enhanced MSC viability and promoted their migration. Combined osteogenic medium with MC, B30, and B30Str increased ALP activity compared to cultivation in basal medium. Osteogenic induction with MC, B30, and B30Str resulted in diffused matrix mineralization. The combined osteogenic induction with biomaterials under mechanical FSS increased Runx2 protein expression either in comparison to those cells cultivated in BM or those cells induced under static culture. Runx2 and ALP expression was upregulated following combined osteogenic differentiation together with B30 and B30Str regardless of static or FSS culture. Conclusions Taken together, the data revealed that FSS in conjunction with biomaterials promoted osteogenic differentiation of MSCs. This combination may be considered as a marked improvement for clinical applications to cure bone defects. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02146-7.
Collapse
Affiliation(s)
- Mohamed I Elashry
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, Frankfurter Str. 98, 35392, Giessen, Germany.
| | - Nadine Baulig
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, Frankfurter Str. 98, 35392, Giessen, Germany
| | - Alena-Svenja Wagner
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, 35392, Giessen, Germany.,Institute of Veterinary Physiology and Biochemistry, Justus Liebig University of Giessen, 35392, Giessen, Germany
| | - Michele C Klymiuk
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, Frankfurter Str. 98, 35392, Giessen, Germany
| | - Benjamin Kruppke
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Str. 27, 01069, Dresden, Germany
| | - Thomas Hanke
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Str. 27, 01069, Dresden, Germany
| | - Sabine Wenisch
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, 35392, Giessen, Germany
| | - Stefan Arnhold
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, Frankfurter Str. 98, 35392, Giessen, Germany
| |
Collapse
|
15
|
El-Habashy SE, Eltaher HM, Gaballah A, Zaki EI, Mehanna RA, El-Kamel AH. Hybrid bioactive hydroxyapatite/polycaprolactone nanoparticles for enhanced osteogenesis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 119:111599. [PMID: 33321643 DOI: 10.1016/j.msec.2020.111599] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/20/2020] [Accepted: 09/23/2020] [Indexed: 02/05/2023]
Abstract
Hydroxyapatite nanoparticles (HApN) are largely employed as osteogenic inorganic material. Inorganic/polymeric hybrid nanostructures can provide versatile bioactivity for superior osteogenicity, particularly as nanoparticles. Herein, we present hybrid biomaterial-based hydroxyapatite/polycaprolactone nanoparticles (HAp/PCL NPs) realized using simple preparation techniques to augment HApN osteogenicity. Using wet chemical precipitation, we optimized HApN crystalline properties utilizing a 23-factorial design. Optimized HApN exhibited typical Ca/P elemental ratio with high reaction yield. Surface area analysis revealed their mesoporous nature and high surface area. Hybrid HAp/PCL NPs prepared using direct emulsification-solvent evaporation maintained HApN crystallinity with no observed chemical interactions. To the best of our knowledge, we are the first to elaborate the biocompatibility and osteogenicity of nanoparticulate hybrid HAp/PCL. Hybrid HAp/PCL NPs outperformed HApN regarding mesenchymal cell proliferation and osteodifferentiation with reduction of possible cytotoxicity. Unlike HApN, hybrid HAp/PCL NPs presented moderate expression of early osteogenic markers, Runx-2 and osteopontin and significantly elevated expression of the late osteogenic marker, bone sialoprotein after 10-day culture. Our results indicate that hybrid bioactive HAp/PCL NPs could offer a more prominent osteogenic potential than plain HApN for bone regenerative applications as a standalone nanoplatform or as part of complex engineered systems.
Collapse
Affiliation(s)
- Salma E El-Habashy
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Hoda M Eltaher
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Ahmed Gaballah
- Microbiology Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Eiman I Zaki
- Histology and Cell Biology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Radwa A Mehanna
- Medical Physiology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt; Center of Excellence for Research in Regenerative Medicine and Applications CERRMA, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Amal H El-Kamel
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
16
|
Moussi K, Abusamra DB, Yassine O, Merzaban J, Kosel J. Strain-induced Differentiation of Mesenchymal Stem Cells. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2020:2239-2244. [PMID: 33018453 DOI: 10.1109/embc44109.2020.9176273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Directing the fate of human mesenchymal stem/stromal cells (hMSCs) toward bone formation using mechanical strain is a promising approach in regenerative medicine related to bone diseases. Numerous studies have evaluated the effects of vibration or cyclic tensile strain on MSCs towards developing a mechanically-based method for stimulating differentiation. Here, we study the differentiation of hMSCs cultured on elastic polydimethylsiloxane (PDMS) membrane, which is magnetically actuated to induce periodically varying strain. The strain distribution across the membrane was calculated by finite-element modeling and demonstrates three main areas of different strain amplitudes. The strain effect on the hMSCs was evaluated by measuring the mineralization of differentiated hMSCs using Alizarin S red stain. The results indicate a strain-dependent differentiation of hMSCs, where the highest region of strain on the membrane resulted in the most accelerated differentiation. Osteogenic differentiation was achieved as early as two weeks, which is significantly sooner than control hMSCs treated with osteogenic media alone.
Collapse
|
17
|
Wang J, Chen X, Yang X, Guo B, Li D, Zhu X, Zhang X. Positive role of calcium phosphate ceramics regulated inflammation in the osteogenic differentiation of mesenchymal stem cells. J Biomed Mater Res A 2020; 108:1305-1320. [PMID: 32064734 DOI: 10.1002/jbm.a.36903] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 02/09/2020] [Accepted: 02/12/2020] [Indexed: 02/05/2023]
Abstract
Recently, researches have confirmed the crucial role of inflammatory response in Ca-P ceramic-induced osteogenesis, however, the underlying mechanism has not yet been fully understood. In this study, BCP and β-TCP ceramics were used as material models to investigate the effect of physicochemical properties on inflammatory response in vitro. The results showed that BCP and β-TCP could support macrophages attachment, proliferation, and spreading favorably, as well as promote gene expressions of inflammatory related cytokines (IL-1, IL-6, MCP-1, and TNF-α) and growth factors (TGF-β, FGF, PDGF, VEGF, IGF, and EGF). BCP showed a facilitating function on the gene expressions earlier than β-TCP. Further coculture experiments performed in vitro demonstrated that the CMs containing various increased cytokines for macrophages pre-culture could significantly promote MSCs osteogenic differentiation, which was confirmed by the gene expressions of osteogenic specific markers and the intracellular OCN product accumulation under the stimulation of BCP and β-TCP ceramics. Further evidence was found from the formation of mineralized nodules in BCM and TCM. In addition, this study showed a concise relationship between Ca-P ceramic induced inflammation and its osteoinductivity that the increased cytokines and growth factors from macrophages could promote MSCs osteogenic differentiation.
Collapse
Affiliation(s)
- Jing Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Xuening Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Xiao Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Bo Guo
- Department of Ophthalmology, West China Hospital of Sichuan University, Chengdu, China
| | - Danyang Li
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Liu D, Hong Y, Li Y, Hu C, Yip TC, Yu WK, Zhu Y, Fong CC, Wang W, Au SK, Wang S, Yang M. Targeted destruction of cancer stem cells using multifunctional magnetic nanoparticles that enable combined hyperthermia and chemotherapy. Theranostics 2020; 10:1181-1196. [PMID: 31938059 PMCID: PMC6956796 DOI: 10.7150/thno.38989] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 10/14/2019] [Indexed: 12/17/2022] Open
Abstract
Cancer stem cells (CSCs) have been implicated in cancer recurrence and therapy resistance. Therefore, a CSC-targeted therapy that disrupts the maintenance and survival of CSCs may offer an effective approach in killing tumor cells in primary tumors and preventing the metastasis caused by CSCs. Nanoparticles (NPs)-based thermotherapy and/or chemotherapy are promising therapeutic methods for cancer treatment. Methods: A silica-based multifunctional NP system was present, which encapsulated a chemotherapeutic agent and magnetic cores and coated with a specific antibody against the lung CSCs. The efficacy of this novel therapeutic strategy was systematically studied both in vitro and in vivo by simultaneous activating the combined thermotherapy and chemotherapy via CSC-targeted NPs. Results: These NPs were systematically administered and activated for targeted chemotherapy and thermotherapy by using an externally applied alternating magnetic field (AMF). The antibody-modified NPs targeted to lung CSCs with enhanced cellular uptake in vitro and extended accumulation in tumor in vivo. Up to 98% of lung CSCs was killed in vitro with 30-min application of AMF, due to the combined effects of hyperthermia and chemotherapeutic drug treatment. In in vivo models, this combined therapy significantly suppressed tumor growth and metastasis in lung CSC xenograft-bearing mice, with minimal side effects and adverse effects. Conclusion: With good biocompatibility and targeting capability, the nanodrug delivery system may offer a promising clinical platform for the combined thermotherapy and chemotherapy. This work demonstrated the feasibility of developing multifunctional nanomedicine targeting CSCs for effective cancer treatment.
Collapse
Affiliation(s)
- Dandan Liu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen 518057, China
- College of Chemistry and Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding 071002, China
| | - Yingcai Hong
- Department of Thoracic Surgery, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University, Shenzhen 510000, China
| | - Yaping Li
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen 518057, China
| | - Chong Hu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen 518057, China
| | - Tak-Chun Yip
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China
| | - Wai-Kin Yu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen 518057, China
| | - Yu Zhu
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Chi-Chun Fong
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
- Department of Thoracic Surgery, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University, Shenzhen 510000, China
| | - Weimao Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Siu-Kie Au
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China
| | - Shubin Wang
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Mengsu Yang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen 518057, China
| |
Collapse
|
19
|
Casanova MR, Alves da Silva M, Costa-Pinto AR, Reis RL, Martins A, Neves NM. Chondrogenesis-inductive nanofibrous substrate using both biological fluids and mesenchymal stem cells from an autologous source. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 98:1169-1178. [DOI: 10.1016/j.msec.2019.01.069] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 12/10/2018] [Accepted: 01/16/2019] [Indexed: 02/07/2023]
|
20
|
Silicate-based bioceramic scaffolds for dual-lineage regeneration of osteochondral defect. Biomaterials 2019; 192:323-333. [DOI: 10.1016/j.biomaterials.2018.11.025] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 11/12/2018] [Accepted: 11/14/2018] [Indexed: 01/26/2023]
|
21
|
Zhang C, Xie B, Zou Y, Zhu D, Lei L, Zhao D, Nie H. Zero-dimensional, one-dimensional, two-dimensional and three-dimensional biomaterials for cell fate regulation. Adv Drug Deliv Rev 2018; 132:33-56. [PMID: 29964080 DOI: 10.1016/j.addr.2018.06.020] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/01/2018] [Accepted: 06/25/2018] [Indexed: 02/06/2023]
Abstract
The interaction of biological cells with artificial biomaterials is one of the most important issues in tissue engineering and regenerative medicine. The interaction is strongly governed by physical and chemical properties of the materials and displayed with differentiated cellular behaviors, including cell self-renewal, differentiation, reprogramming, dedifferentiation, or transdifferentiation as a result. A number of engineered biomaterials with micro- or nano-structures have been developed to mimic structural components of cell niche and specific function of extra cellular matrix (ECM) over past two decades. In this review article, we briefly introduce the fabrication of biomaterials and their classification into zero-dimensional (0D), one-dimensional (1D), two-dimensional (2D) and three-dimensional (3D) ones. More importantly, the influence of different biomaterials on inducing cell self-renewal, differentiation, reprogramming, dedifferentiation, and transdifferentiation was discussed based on the progress at 0D, 1D, 2D and 3D levels, following which the current research limitations and research perspectives were provided.
Collapse
Affiliation(s)
- Can Zhang
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha 410082, China
| | - Bei Xie
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha 410082, China
| | - Yujian Zou
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha 410082, China
| | - Dan Zhu
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha 410082, China
| | - Lei Lei
- Department of Orthodontics, Xiangya Stomatological Hospital, Central South University, Changsha 410008, China.
| | - Dapeng Zhao
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha 410082, China.
| | - Hemin Nie
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha 410082, China; Shenzhen Research Institute of Hunan University, Nanshan Hi-new Technology and Industry Park, Shenzhen 518057, China.
| |
Collapse
|
22
|
Deng C, Zhu H, Li J, Feng C, Yao Q, Wang L, Chang J, Wu C. Bioactive Scaffolds for Regeneration of Cartilage and Subchondral Bone Interface. Theranostics 2018; 8:1940-1955. [PMID: 29556366 PMCID: PMC5858510 DOI: 10.7150/thno.23674] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 01/17/2018] [Indexed: 01/15/2023] Open
Abstract
The cartilage lesion resulting from osteoarthritis (OA) always extends into subchondral bone. It is of great importance for simultaneous regeneration of two tissues of cartilage and subchondral bone. 3D-printed Sr5(PO4)2SiO4 (SPS) bioactive ceramic scaffolds may achieve the aim of regenerating both of cartilage and subchondral bone. We hypothesized that strontium (Sr) and silicon (Si) ions released from SPS scaffolds play a crucial role in osteochondral defect reconstruction. Methods: SPS bioactive ceramic scaffolds were fabricated by a 3D-printing method. The SEM and ICPAES were used to investigate the physicochemical properties of SPS scaffolds. The proliferation and maturation of rabbit chondrocytes stimulated by SPS bioactive ceramics were measured in vitro. The stimulatory effect of SPS scaffolds for cartilage and subchondral bone regeneration was investigated in vivo. Results: SPS scaffolds significantly stimulated chondrocyte proliferation, and SPS extracts distinctly enhanced the maturation of chondrocytes and preserved chondrocytes from OA. SPS scaffolds markedly promoted the regeneration of osteochondral defects. The complex interface microstructure between cartilage and subchondral bone was obviously reconstructed. The underlying mechanism may be related to Sr and Si ions stimulating cartilage regeneration by activating HIF pathway and promoting subchondral bone reconstruction through activating Wnt pathway, as well as preserving chondrocytes from OA via inducing autophagy and inhibiting hedgehog pathway. Conclusion: Our findings suggest that SPS scaffolds can help osteochondral defect reconstruction and well reconstruct the complex interface between cartilage and subchondral bone, which represents a promising strategy for osteochondral defect regeneration.
Collapse
|
23
|
Xue R, Qian Y, Li L, Yao G, Yang L, Sun Y. Polycaprolactone nanofiber scaffold enhances the osteogenic differentiation potency of various human tissue-derived mesenchymal stem cells. Stem Cell Res Ther 2017. [PMID: 28646917 PMCID: PMC5482966 DOI: 10.1186/s13287-017-0588-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Polycaprolactone (PCL) has been regarded as a promising synthetic material for bone tissue engineering application. Owing to its unique biochemical properties and great compatibility, PCL fibers have come to be explored as a potential delivering scaffold for stem cells to support bone regeneration during clinical application. METHODS The human derived mesenchymal stem cells (MSCs) were obtained from umbilical cord (UC), bone marrow (BM), and adipose tissue (AD), respectively. The osteogenic differentiation potency of various human MSCs on this novel synthetic biomaterial was also investigated in vitro. RESULTS Here, we illustrated that those human UC-, BM-, and AD-derived MSCs exhibited fibroblast-like morphology and expressed characteristic markers. Impressively, PCL nanofiber scaffold could support those MSC adhesion and proliferation. Long-term culture on PCL nanofiber scaffold maintained the viability as well as accelerated the proliferation of those three different kinds of human MSCs. More importantly, the osteogenic differentiation potency of those human MSCs was increased significantly by culturing on PCL nanofiber scaffold. Of note, BM-derived MSCs demonstrated greater differentiation potency among the three kinds of MSCs. The Wnt/β-catenin and Smad3 signaling pathways contributed to the enhanced osteogenesis of human MSCs, which was activated consistently by PCL nanofiber scaffold. CONCLUSIONS The utilization of PCL nanofiber scaffold would provide a great application potential for MSC-based bone tissue repair by enhancing the osteogenic differentiation of human MSCs.
Collapse
Affiliation(s)
- Ruyue Xue
- Reproductive Medical Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yuna Qian
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, 400044, China
| | - Linhao Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Guidong Yao
- Reproductive Medical Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, 400044, China
| | - Yingpu Sun
- Reproductive Medical Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
24
|
Yan Q, Fang L, Wei J, Xiao G, Lv M, Ma Q, Liu C, Wang W. Silicon nanowires enhanced proliferation and neuronal differentiation of neural stem cell with vertically surface microenvironment. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2017; 28:1394-1407. [PMID: 28494208 DOI: 10.1080/09205063.2017.1329888] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Owing to its biocompatibility, noncytotoxicity, biodegradability and three-dimensional structure, vertically silicon nanowires (SiNWs) arrays are a promising scaffold material for tissue engineering, regenerative medicine and relevant medical applications. Recently, its osteogenic differentiation effects, reorganization of cytoskeleton and regulation of the fate on stem cells have been demonstrated. However, it still remains unknown whether SiNWs arrays could affect the proliferation and neuronal differentiation of neural stem cells (NSCs) or not. In the present study, we have employed vertically aligned SiNWs arrays as culture systems for NSCs and proved that the scaffold material could promote the proliferation and neuronal differentiation of NSCs while maintaining excellent cell viability and stemness. Immunofluorescence imaging analysis, Western blot and RT-PCR results reveal that NSCs proliferation and neuronal differentiation efficiency on SiNWs arrays are significant greater than that on silicon wafers. These results implicate SiNWs arrays could offer a powerful platform for NSCs research and NSCs-based therapy in the field of neural tissue engineering.
Collapse
Affiliation(s)
- Qiuting Yan
- a College of Pharmaceutical Sciences , Soochow University , Suzhou , China
| | - Lipao Fang
- b Institute of Neuroscience , Soochow University , Suzhou , China
| | - Jiyu Wei
- c NGS Laboratory , GENEWIZ, Inc. , Suzhou , China
| | - Guipeng Xiao
- c NGS Laboratory , GENEWIZ, Inc. , Suzhou , China
| | - Meihong Lv
- b Institute of Neuroscience , Soochow University , Suzhou , China
| | - Quanhong Ma
- b Institute of Neuroscience , Soochow University , Suzhou , China
| | - Chunfeng Liu
- b Institute of Neuroscience , Soochow University , Suzhou , China.,d Department of Neurology and Suzhou Clinical Research Centre of Neurological Disease , The Second Affiliated Hospital of Soochow University , Suzhou , China
| | - Wang Wang
- b Institute of Neuroscience , Soochow University , Suzhou , China
| |
Collapse
|
25
|
Wang JY, Chen WM, Wen CS, Hung SC, Chen PW, Chiu JH. Du-Huo-Ji-Sheng-Tang and its active component Ligusticum chuanxiong promote osteogenic differentiation and decrease the aging process of human mesenchymal stem cells. JOURNAL OF ETHNOPHARMACOLOGY 2017; 198:64-72. [PMID: 28040510 DOI: 10.1016/j.jep.2016.12.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 09/01/2016] [Accepted: 12/10/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Postmenopausal osteoporosis is the most common bone disease worldwide. Information concerning the effects of herbal medicines on mesenchymal cell osteogenesis and senescence remains lacking. AIM OF THIS STUDY This study was designed to investigate the effects of Du-Huo-Ji-Sheng-Tang (DHJST), a Chinese herbal medicine and its active component Ligusticum chuanxiong on osteogenic differentiation and the aging process of human mesenchymal cells (hMSCs). MATERIALS & METHODS hMSCs were used as in vitro model and osteogenesis was induced by administration of either osteogenesis inducing medium (OIM) or dexamethasone-depleted OIM (DDOIM) for 1-week or 2 weeks and the results were evaluated by measuring the formation of mineralization nodules. The effects of the compound recipe DHJST and its active component L. chuanxiong on hMSCs osteogenesis-related gene expression was determined by real-time PCR that targeted bone morphogenetic protein-2 (BMP2), RUNX2, ALP, COL-1, osteopontin (OPN), and osteocalcin (OCN). Antibodies against BMP-related signaling pathway proteins, such as BMP-2, ERK, SMAD 1/5/8, and RUNX2, were also detected at the protein level by Western blotting. Finally, the cumulative growth curve and senescence of the hMSCs were evaluated in order to assess the aging process. RESULTS L. chuanxiong increased osteogenic activity in hMSCs and up-regulated BMP-2 and RUNX2 gene expression via the activation of SMAD 1/5/8 and ERK signaling. Furthermore DHJST also showed a trend towards promoting the same effects in the same system. In the absence of dexamethasone, DHJST did activate SMAD 1/5/8 and ERK signaling and hence increased RUNX2 protein expression in hMSCs. In addition, both DHJST and L. chuanxiong delayed the hMSCs aging process by decreasing cell senescence. CONCLUSIONS We concluded that DHJST and its active component L. chuanxiong are able to promote osteogenic activity and decrease hMSCs senescence as cells age.
Collapse
Affiliation(s)
- Jir-You Wang
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC; Department of Orthopedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC.
| | - Wei-Ming Chen
- Department of Orthopedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Che-Sheng Wen
- Department of Orthopedics, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC
| | - Shih-Chieh Hung
- Department of Orthopedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Pei-Wen Chen
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Jen-Hwey Chiu
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC; Division of General Surgery, Departml;ent of Surgery, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC; Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| |
Collapse
|
26
|
Perez JE, Ravasi T, Kosel J. Mesenchymal stem cells cultured on magnetic nanowire substrates. NANOTECHNOLOGY 2017; 28:055703. [PMID: 28029098 DOI: 10.1088/1361-6528/aa52a3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Stem cells have been shown to respond to extracellular mechanical stimuli by regulating their fate through the activation of specific signaling pathways. In this work, an array of iron nanowires (NWs) aligned perpendicularly to the surface was fabricated by pulsed electrodepositon in porous alumina templates followed by a partial removal of the alumina to reveal 2-3 μm of the NWs. This resulted in alumina substrates with densely arranged NWs of 33 nm in diameter separated by 100 nm. The substrates were characterized by scanning electron microscopy (SEM) energy dispersive x-ray analysis and vibrating sample magnetometer. The NW array was then used as a platform for the culture of human mesenchymal stem cells (hMSCs). The cells were stained for the cell nucleus and actin filaments, as well as immuno-stained for the focal adhesion protein vinculin, and then observed by fluorescence microscopy in order to characterize their spreading behavior. Calcein AM/ethidium homodimer-1 staining allowed the determination of cell viability. The interface between the cells and the NWs was studied using SEM. Results showed that hMSCs underwent a re-organization of actin filaments that translated into a change from an elongated to a spherical cell shape. Actin filaments and vinculin accumulated in bundles, suggesting the attachment and formation of focal adhesion points of the cells on the NWs. Though the overall number of cells attached on the NWs was lower compared to the control, the attached cells maintained a high viability (>90%) for up to 6 d. Analysis of the interface between the NWs and the cells confirmed the re-organization of F-actin and revealed the adhesion points of the cells on the NWs. Additionally, a net of filopodia surrounded each cell, suggesting the probing of the array to find additional adhesion points. The cells maintained their round shape for up to 6 d of culture. Overall, the NW array is a promising nanostructured platform for studying and influencing hMSCs differentiation.
Collapse
Affiliation(s)
- Jose E Perez
- Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia. Division of Computer, Electrical and Mathematical Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | | | | |
Collapse
|
27
|
Dobbenga S, Fratila-Apachitei LE, Zadpoor AA. Nanopattern-induced osteogenic differentiation of stem cells - A systematic review. Acta Biomater 2016; 46:3-14. [PMID: 27667018 DOI: 10.1016/j.actbio.2016.09.031] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 09/07/2016] [Accepted: 09/22/2016] [Indexed: 01/16/2023]
Abstract
It is well known that biomaterials topography can influence the behavior of stem cells. Nevertheless, the fundamentals and the impact of nanoscale topography are just emerging. The main objective of this review has been to reveal the state-of-the-art on the effects of controlled nanoscale topographies (nanopatterns) on in vitro osteogenic differentiation of mesenchymal stem cells (MSCs) in the absence of osteogenic supplements. The findings indicate that nanopatterns with specific feature sizes, spatial arrangements, or shapes may induce osteogenic differentiation of MSCs. Regardless of substrate chemistry, nanopattern-induced osteogenic differentiation is associated with large focal adhesions, enhanced cell areas, and well organized cytoskeleton. These results suggest that earlier interactions between nanopattern features and cell receptors are involved, with effects on the entire cell structure and subsequent differentiation. Such events are possibly mediated by nanotopography-induced mechanotransduction pathways. The findings so far reveal that nanoscale topography has potential for directing differentiation of MSCs towards the osteogenic lineage in non-osteogenic media and should be harnessed for possible synergistic effects in bone regenerative therapies. STATEMENT OF SIGNIFICANCE The use of nanotopography to induce cellular responses represents a novel and rapidly growing area of research. Nevertheless, the findings and trends so far are difficult to identify and discuss mostly due to a non-systematic research approach. The present manuscript is providing a systematic review focused on nanopattern-induced osteogenic differentiation of mesenchymal stem cells. The coverage of the most relevant aspects including nanopatterns fabrication methods, their effects on osteogenic differentiation of mesenchymal stem cells as well as the related effects on adhesion and cell morphology has enabled an integrated discussion including the potential mechanotransduction mechanisms involved. Furthermore, a clear distinction between the studies that use only surface nanotopographies and the ones that mix nanotopographical features with osteogenic supplements has been made. This delineation is essential for revealing and understanding the role of biomaterial's nanotopography per se on stem cells differentiation based on which novel osteoinductive biomaterials can be developed.
Collapse
|
28
|
Xu L, Zhao Y, Wang M, Song W, Li B, Liu W, Jin X, Zhang H. Defocused low-energy shock wave activates adipose tissue-derived stem cells in vitro via multiple signaling pathways. Cytotherapy 2016; 18:1503-1514. [PMID: 27727017 DOI: 10.1016/j.jcyt.2016.08.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 08/14/2016] [Accepted: 08/21/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND AIMS We found defocused low-energy shock wave (DLSW) could be applied in regenerative medicine by activating mesenchymal stromal cells. However, the possible signaling pathways that participated in this process remain unknown. In the present study, DLSW was applied in cultured rat adipose tissue-derived stem cells (ADSCs) to explore its effect on ADSCs and the activated signaling pathways. METHODS After treating with DLSW, the cellular morphology and cytoskeleton of ADSCs were observed. The secretions of ADSCs were detected. The expressions of ADSC surface antigens were analyzed using flow cytometry. The expressions of proliferating cell nuclear antigen and Ki67 were analyzed using western blot. The expression of CXCR2 and the migrations of ADSCs in vitro and in vivo were detected. The phosphorylation of selected signaling pathways with or without inhibitors was also detected. RESULTS DLSW did not change the morphology and phenotype of ADSCs, and could promote the secretion, proliferation and migration of ADSCs. The phosphorylation levels were significantly higher in mitogen-activated protein kinases (MAPK) pathway, phosphoinositide 3-kinase (PI-3K)/AKT pathway and nuclear factor-kappa B (NF-κB) signaling pathway but not in Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway. Furthermore, ADSCs were not activated by DLSW after adding the inhibitors of these pathways simultaneously. CONCLUSIONS Our results demonstrated for the first time that DLSW could activate ADSCs through MAPK, PI-3K/AKT and NF-κB signaling pathways. Combination of DLSW and agonists targeting these pathways might improve the efficacy of ADSCs in regenerative medicine in the future.
Collapse
Affiliation(s)
- Lina Xu
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Yong Zhao
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Muwen Wang
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Wei Song
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Bo Li
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Wei Liu
- Department of Pharmacy, Peking University Third Hospital, Beijing, China; Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, USA
| | - Xunbo Jin
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Haiyang Zhang
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China; Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California, USA.
| |
Collapse
|
29
|
Wang C, Liu D, Zhang C, Sun J, Feng W, Liang XJ, Wang S, Zhang J. Defect-Related Luminescent Hydroxyapatite-Enhanced Osteogenic Differentiation of Bone Mesenchymal Stem Cells Via an ATP-Induced cAMP/PKA Pathway. ACS APPLIED MATERIALS & INTERFACES 2016; 8:11262-11271. [PMID: 27088570 DOI: 10.1021/acsami.6b01103] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Novel defect-related hydroxyapatite (DHAP), which combines the advantages of HAP and defect-related luminescence, has the potential application in tissue engineering and biomedical area, because of its excellent capability of monitoring the osteogenic differentiation and material biodegradation. Although the extracellular mechanism of DHAP minerals and PO4(3-) functioning in osteogenic differentiation has been widely studied, the intracellular molecular mechanism through which PO4(3-) mediates osteogenesis of bone mesenchymal stem cells (BMSCs) is not clear. We examined a previously unknown molecular mechanism through which PO4(3-) promoted osteogenesis of BMSCs with an emphasis on adenosine-triphosphate (ATP)-induced cAMP/PKA pathway. Our studies showed that DHAP could be uptaken into lysosome, in which PO4(3-) was released from DHAP, because of the acid environment of lysosome. The released PO4(3-) interacted with ADP to form ATP, and then degraded into adenosine, an ATP metabolite, which interacted with A2b adenosine receptor to activate the cAMP/PKA pathway, resulting in the high expression of osteogenesis-related genes, such as Runx2, BMP-2, and OCN. These findings first revealed the function of ATP-metabolism in bone physiological homeostasis, which may be developed to cure bone metabolic diseases.
Collapse
Affiliation(s)
- Chao Wang
- College of Chemistry and Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University , Baoding 071002, People's Republic of China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University , Baoding 071002, People's Republic of China
| | - Dandan Liu
- College of Chemistry and Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University , Baoding 071002, People's Republic of China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University , Baoding 071002, People's Republic of China
| | - Cuimiao Zhang
- College of Chemistry and Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University , Baoding 071002, People's Republic of China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University , Baoding 071002, People's Republic of China
| | - Jiadong Sun
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University , Baoding 071002, People's Republic of China
| | - Weipei Feng
- College of Chemistry and Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University , Baoding 071002, People's Republic of China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University , Baoding 071002, People's Republic of China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China , Beijing 100190, People's Republic of China
| | - Shuxiang Wang
- College of Chemistry and Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University , Baoding 071002, People's Republic of China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University , Baoding 071002, People's Republic of China
| | - Jinchao Zhang
- College of Chemistry and Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University , Baoding 071002, People's Republic of China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University , Baoding 071002, People's Republic of China
| |
Collapse
|
30
|
Schimke MM, Stigler R, Wu X, Waag T, Buschmann P, Kern J, Untergasser G, Rasse M, Steinmüller-Nethl D, Krueger A, Lepperdinger G. Biofunctionalization of scaffold material with nano-scaled diamond particles physisorbed with angiogenic factors enhances vessel growth after implantation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:823-833. [DOI: 10.1016/j.nano.2015.11.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 10/21/2015] [Accepted: 11/04/2015] [Indexed: 12/26/2022]
|
31
|
Xia L, Yin Z, Mao L, Wang X, Liu J, Jiang X, Zhang Z, Lin K, Chang J, Fang B. Akermanite bioceramics promote osteogenesis, angiogenesis and suppress osteoclastogenesis for osteoporotic bone regeneration. Sci Rep 2016; 6:22005. [PMID: 26911441 PMCID: PMC4766478 DOI: 10.1038/srep22005] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 02/05/2016] [Indexed: 11/09/2022] Open
Abstract
It is a big challenge for bone healing under osteoporotic pathological condition with impaired angiogenesis, osteogenesis and remodeling. In the present study, the effect of Ca, Mg, Si containing akermanite bioceramics (Ca2MgSi2O7) extract on cell proliferation, osteogenic differentiation and angiogenic factor expression of BMSCs derived from ovariectomized rats (BMSCs-OVX) as well as the expression of osteoclastogenic factors was evaluated. The results showed that akermanite could enhance cell proliferation, ALP activity, expression of Runx2, BMP-2, BSP, OPN, OCN, OPG and angiogenic factors including VEGF and ANG-1. Meanwhile, akermanite could repress expression of osteoclastogenic factors including RANKL and TNF-α. Moreover, akermanite could activate ERK, P38, AKT and STAT3 signaling pathways, while crosstalk among these signaling pathways was evident. More importantly, the effect of akermanite extract on RANKL-induced osteoclastogenesis was evaluated by TRAP staining and real-time PCR assay. The results showed that akermanite could suppress osteoclast formation and expression of TRAP, cathepsin K and NFATc1. The in vivo experiments revealed that akermanite bioceramics dramatically stimulated osteogenesis and angiogenesis in an OVX rat critical-sized calvarial defect model. All these results suggest that akermanite bioceramics with the effects of Mg and Si ions on osteogenesis, angiogenesis and osteoclastogenesis are promising biomaterials for osteoporotic bone regeneration.
Collapse
Affiliation(s)
- Lunguo Xia
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-maxillofacial Science, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, China
| | - Zhilan Yin
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Lixia Mao
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-maxillofacial Science, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, China
| | - Xiuhui Wang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Jiaqiang Liu
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-maxillofacial Science, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, China
| | - Xinquan Jiang
- Oral Bioengineering and regenerative medicine Lab, Shanghai Research Institute of Stomatology, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Zhiyuan Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Kaili Lin
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.,School &Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, China
| | - Jiang Chang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Bing Fang
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-maxillofacial Science, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, China
| |
Collapse
|
32
|
Chen X, Wang J, Wang J, Zhu X, Yang X, Fan Y, Zhang X. The positive role of macrophage secretion stimulated by BCP ceramic in the ceramic-induced osteogenic differentiation of pre-osteoblasts via Smad-related signaling pathways. RSC Adv 2016. [DOI: 10.1039/c6ra23362g] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The present study demonstrated that material-mediated immune responses, particularly macrophage secretion might play a vital role in material-induced osteogenesis.
Collapse
Affiliation(s)
- Xuening Chen
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu
- China
| | - Jing Wang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu
- China
| | - Jing Wang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu
- China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu
- China
| | - Xiao Yang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu
- China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu
- China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu
- China
| |
Collapse
|
33
|
Li W, Wei S, Liu C, Song M, Wu H, Yang Y. Regulation of the osteogenic and adipogenic differentiation of bone marrow-derived stromal cells by extracellular uridine triphosphate: The role of P2Y2 receptor and ERK1/2 signaling. Int J Mol Med 2015; 37:63-73. [PMID: 26531757 PMCID: PMC4687443 DOI: 10.3892/ijmm.2015.2400] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/20/2015] [Indexed: 11/13/2022] Open
Abstract
An imbalance in the osteogenesis and adipogenesis of bone marrow-derived stromal cells (BMSCs) is a crucial pathological factor in the development of osteoporosis. Growing evidence suggests that extracellular nucleotide signaling involving the P2 receptors plays a significant role in bone metabolism. The aim of the present study was to investigate the effects of uridine triphosphate (UTP) on the osteogenic and adipogenic differentiation of BMSCs, and to elucidate the underlying mechanisms. The differentiation of the BMSCs was determined by measuring the mRNA and protein expression levels of osteogenic- and adipogenic-related markers, alkaline phosphatase (ALP) staining, alizarin red staining and Oil Red O staining. The effects of UTP on BMSC differentiation were assayed using selective P2Y receptor antagonists, small interfering RNA (siRNA) and an intracellular signaling inhibitor. The incubation of the BMSCs with UTP resulted in a dose-dependent decrease in osteogenesis and an increase in adipogenesis, without affecting cell proliferation. Significantly, siRNA targeting the P2Y2 receptor prevented the effects of UTP, whereas the P2Y6 receptor antagonist (MRS2578) and siRNA targeting the P2Y4 receptor had little effect. The activation of P2Y receptors by UTP transduced to the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway. This transduction was prevented by the mitogen-activated protein kinase inhibitor (U0126) and siRNA targeting the P2Y2 receptor. U0126 prevented the effects of UTP on osteogenic- and adipogenic-related gene expression after 24 h of culture, as opposed to 3 to 7 days of culture. Thus, our data suggest that UTP suppresses the osteogenic and enhances the adipogenic differentiation of BMSCs by activating the P2Y2 receptor. The ERK1/2 signaling pathway mediates the early stages of this process.
Collapse
Affiliation(s)
- Wenkai Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Sheng Wei
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Chaoxu Liu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Mingyu Song
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Yong Yang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| |
Collapse
|
34
|
Li W, Li G, Zhang Y, Wei S, Song M, Wang W, Yuan X, Wu H, Yang Y. Role of P2 × 7 receptor in the differentiation of bone marrow stromal cells into osteoblasts and adipocytes. Exp Cell Res 2015; 339:367-79. [PMID: 26481422 DOI: 10.1016/j.yexcr.2015.10.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 10/07/2015] [Accepted: 10/09/2015] [Indexed: 01/05/2023]
Abstract
Imbalance in osteogenesis and adipogenesis of bone marrow stromal cells is a crucial pathological process of osteoporosis. P2 × 7-deficient mice were previously shown to exhibit an osteopenic phenotype and abnormal fat distribution, leading us to hypothesize that P2 × 7R activation was involved in the differentiation of BMSCs. Consequently, we investigated the effect of P2 × 7R activation on osteogenic and adipogenic differentiation of BMSCs in vitro, and established an ovariectomized (OVX) osteoporosis model to test P2 × 7R activation on adipocytes formation, trabecular and cortical bone parameters in vivo. Our results showed that activation of P2 × 7R by BzATP resulted in increase in the gene expression of osteoblastic markers, the activity of alkaline phosphatase and bone mineralization, and decrease in the gene expression of adipogenic markers and the number of adipocytes generated by BMSCs. MicroCT analysis showed that BzATP treatment ameliorated the micro-architecture of trabecular bones in OVX mice, while cortical bone parameters were unaffected. H&E staining analysis showed that was increase in the volume of trabecular bone and number of trabecular bone, and decrease in bone marrow adipocytes in BzATP-treated OVX mice compared with OVX mice. Also, activation of P2 × 7R transduced to ERK1/2 and JNK signaling pathways. This transduction was prevented by BBG, U0126, and SP600125. U0126 and SP600125 prevented BzATP-induced up-regulation of osteogenic-related genes expression and down-regulation of adipogenic-related genes expression. These data suggest that BzATP activates the differentiation of BMSCs into osteoblasts but not adipocytes by stimulating ERK1/2 and JNK signaling pathways in a P2 × 7R-dependent way.
Collapse
Affiliation(s)
- Wenkai Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China
| | - Guizhen Li
- Department of Orthopedics, Enshi Center Hospital, Enshi 445000, China
| | - Yingchi Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China
| | - Sheng Wei
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China
| | - Mingyu Song
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China
| | - Wei Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China
| | - Xuefeng Yuan
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China.
| | - Yong Yang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China.
| |
Collapse
|
35
|
Jiao K, Niu LN, Li QH, Chen FM, Zhao W, Li JJ, Chen JH, Cutler CW, Pashley DH, Tay FR. Biphasic silica/apatite co-mineralized collagen scaffolds stimulate osteogenesis and inhibit RANKL-mediated osteoclastogenesis. Acta Biomater 2015; 19:23-32. [PMID: 25792280 DOI: 10.1016/j.actbio.2015.03.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Revised: 02/06/2015] [Accepted: 03/05/2015] [Indexed: 01/19/2023]
Abstract
The effects of a biphasic mineralized collagen scaffold (BCS) containing intrafibrillar silica and apatite on osteogenesis of mouse mesenchymal stem cells (mMSCs) and inhibition of receptor activator of nuclear factor κB ligand (RANKL)-mediated osteoclastogenesis were investigated in the present study. mMSCs were cultured by exposing to BCS for 7 days for cell proliferation/viability examination, and stimulated to differentiate in osteogenic medium for 7-21 days for evaluation of alkaline phosphatase activity, secretion of osteogenic deposits and expression of osteoblast lineage-specific phenotypic markers. The effect of BCS-conditioned mMSCs on osteoclastogenesis of RAW 264.7 cells was evaluated by tartrate-resistant acid phosphatase staining and resorption pit analysis. The contributions of mitogen-activated protein kinase (MAPK) and phosphatidylinositol-3 kinase (PI3K) signal transduction pathways to osteogenesis of mMSCs and their osteoprotegerin (OPG) and RANKL expressions were also evaluated. Compared with unmineralized, intrafibrillarly-silicified or intrafibrillarly-calcified collagen scaffolds, BCS enhanced osteogenic differentiation of mMSCs by activation of the extracellular signal regulated kinases (ERK)/MAPK and p38/MAPK signaling pathways. After mMSCs were exposed to BCS, they up-regulated OPG expression and down-regulated RANKL expression through activation of the p38/MAPK and PI3K/protein kinase B (Akt) pathways, resulting in inhibition of the differentiation of RAW 264.7 cells into multinucleated osteoclasts and reduction in osteoclast function. These observations collectively suggest that BCS has the potential to be used in bone tissue engineering when the demand for anabolic activities is higher than catabolic metabolism during the initial stage of wound rehabilitation.
Collapse
Affiliation(s)
- Kai Jiao
- State Key Laboratory of Military Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Li-na Niu
- State Key Laboratory of Military Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Qi-hong Li
- Department of Stomatology, Affiliated Hospital of Academy of Military Medical Science, Beijing, China
| | - Fa-ming Chen
- State Key Laboratory of Military Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Wei Zhao
- Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jun-jie Li
- Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Ji-hua Chen
- State Key Laboratory of Military Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China.
| | | | - David H Pashley
- College of Dental Medicine, Georgia Reagents University, Augusta, GA, USA
| | - Franklin R Tay
- College of Dental Medicine, Georgia Reagents University, Augusta, GA, USA.
| |
Collapse
|
36
|
Jin Y, Chen S, Duan J, Jia G, Zhang J. Europium-doped Gd2O3 nanotubes cause the necrosis of primary mouse bone marrow stromal cells through lysosome and mitochondrion damage. J Inorg Biochem 2015; 146:28-36. [PMID: 25725393 DOI: 10.1016/j.jinorgbio.2015.02.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 02/11/2015] [Accepted: 02/11/2015] [Indexed: 02/07/2023]
Abstract
With the wide applications of europium-doped Gd2O3 nanoparticles (Gd2O3:Eu(3+) NPs) in biomedical fields, it will inevitably increase the chance of human exposure. It was reported that Gd2O3:Eu(3+) NPs could accumulate in bone. However, there have been few reports about the potential effect of Gd2O3:Eu(3+) NPs on bone marrow stromal cells (BMSCs). In this study, the Gd2O3:Eu(3+) nanotubes were prepared and characterized by powder X-ray diffraction (XRD), photoluminescence (PL) excitation and emission spectra, scanning electron microscope (SEM), and transmission electron microscopy (TEM). The cytotoxicity of Gd2O3:Eu(3+) nanotubes on BMSCs and the associated mechanisms were further studied. The results indicated that they could be uptaken into BMSCs by an energy-dependent and macropinocytosis-mediated endocytosis process, and primarily localized in lysosome. Gd2O3:Eu(3+) nanotubes effectively inhibited the viability of BMSCs in concentration and time-dependent manners. A significant increase in the percentage of late apoptotic/necrotic cells, lactate dehydrogenase (LDH) leakage and the number of PI-stained cells was found after BMSCs were treated by 10, 20, and 40μg/mL of Gd2O3:Eu(3+) nanotubes for 12h. No obvious DNA ladders were detected, but a dispersed band was observed. The above results revealed that Gd2O3:Eu(3+) nanotubes could trigger cell death by necrosis instead of apoptosis. Two mechanisms were involved in Gd2O3:Eu(3+) nanotube-induced BMSCs necrosis: lysosomal rupture and release of cathepsins B; and the overproduction of reactive oxygen species (ROS) injury to the mitochondria and DNA. The study provides novel evidence to elucidate the toxicity mechanisms and may be beneficial to more rational applications of these nanomaterials in the future.
Collapse
Affiliation(s)
- Yi Jin
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China; College of Basic Medical Science, Hebei University, Baoding 071000, China
| | - Shizhu Chen
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China
| | - Jianlei Duan
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China
| | - Guang Jia
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China.
| | - Jinchao Zhang
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China.
| |
Collapse
|
37
|
Zhou X, Tao Y, Wang J, Liang C, Wang J, Li H, Chen Q. Roles of FGF-2 and TGF-beta/FGF-2 on differentiation of human mesenchymal stem cells towards nucleus pulposus-like phenotype. Growth Factors 2015; 33:23-30. [PMID: 25270389 DOI: 10.3109/08977194.2014.969420] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Human mesenchymal stem cells (MSCs) are reported to have the capability of differentiating towards nucleus pulposus (NP)-like phenotype under specific culture conditions. So far, the effects of fibroblast growth factor (FGF)-2 and the cocktail effects of transforming growth factor (TGF)-beta and FGF-2 on MSCs remain unclear. Therefore, we designed this study to clarify these effects. MSCs were cultured in conditioned medium containing FGF-2 or TGF-beta/FGF-2, and compared with basal or TGF-beta medium. The groups with FGF-2 showed the increase of cell proliferation. Functional gene markers and novel NP markers decreased in FGF-2 group, together with functional protein expression. Pho-ERK1/2 and pho-Smad3 differed significantly in the two conditioned groups. All these results suggest FGF-2 promotes MSCs' proliferation, synergistically with TGF-beta. However, FGF-2 plays a negative role in cartilage homeostasis. We also demonstrate that FGF-2 has no positive effect in differentiating MSCs into NP-like cells, but hinders the acceleration effect of TGF-beta.
Collapse
Affiliation(s)
- Xiaopeng Zhou
- Department of Orthopedic Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, Zhejiang , People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
38
|
Liu D, Ge K, Sun J, Chen S, Jia G, Zhang J. Lanthanum breaks the balance between osteogenesis and adipogenesis of mesenchymal stem cells through phosphorylation of Smad1/5/8. RSC Adv 2015. [DOI: 10.1039/c5ra02311d] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
La breaks the balance between osteogenesis and adipogenesis of MSCs through phosphorylating Smad1/5/8 to activate the BMP signaling pathway.
Collapse
Affiliation(s)
- Dandan Liu
- College of Chemistry and Environmental Science
- Chemical Biology Key Laboratory of Hebei Province
- Hebei University
- Baoding 071002
- PR China
| | - Kun Ge
- College of Chemistry and Environmental Science
- Chemical Biology Key Laboratory of Hebei Province
- Hebei University
- Baoding 071002
- PR China
| | - Jing Sun
- Affiliated Hospital of Hebei University
- Baoding 071000
- PR China
| | - Shizhu Chen
- College of Chemistry and Environmental Science
- Chemical Biology Key Laboratory of Hebei Province
- Hebei University
- Baoding 071002
- PR China
| | - Guang Jia
- College of Chemistry and Environmental Science
- Chemical Biology Key Laboratory of Hebei Province
- Hebei University
- Baoding 071002
- PR China
| | - Jinchao Zhang
- College of Chemistry and Environmental Science
- Chemical Biology Key Laboratory of Hebei Province
- Hebei University
- Baoding 071002
- PR China
| |
Collapse
|
39
|
Bonde S, Buch-Månson N, Rostgaard KR, Andersen TK, Berthing T, Martinez KL. Exploring arrays of vertical one-dimensional nanostructures for cellular investigations. NANOTECHNOLOGY 2014; 25:362001. [PMID: 25130133 DOI: 10.1088/0957-4484/25/36/362001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The endeavor of exploiting arrays of vertical one-dimensional (1D) nanostructures (NSs) for cellular applications has recently been experiencing a pronounced surge of activity. The interest is rooted in the intrinsic properties of high-aspect-ratio NSs. With a height comparable to a mammalian cell, and a diameter 100-1000 times smaller, NSs should intuitively reach far into a cell and, due to their small diameter, do so without compromising cell health. Single NSs would thus be expedient for measuring and modifying cell response. Further organization of these structures into arrays can provide up-scaled and detailed spatiotemporal information on cell activity, an achievement that would entail a massive leap forward in disease understanding and drug discovery. Numerous proofs-of-principle published recently have expanded the large toolbox that is currently being established in this rapidly advancing field of research. Encouragingly, despite the diversity of NS platforms and experimental conditions used thus far, general trends and conclusions from combining cells with NSs are beginning to crystallize. This review covers the broad spectrum of NS materials and dimensions used; the observed cellular responses with specific focus on adhesion, morphology, viability, proliferation, and migration; compares the different approaches used in the field to provide NSs with the often crucial cytosolic access; covers the progress toward biological applications; and finally, envisions the future of this technology. By maintaining the impressive rate and quality of recent progress, it is conceivable that the use of vertical 1D NSs may soon be established as a superior choice over other current techniques, with all the further benefits that may entail.
Collapse
Affiliation(s)
- Sara Bonde
- Bionanotechnology and Nanomedicine Laboratory, Department of Chemistry and Nano-science Center, University of Copenhagen, Universitetsparken 5, DK-2100, Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|