1
|
Jin X, Lu Y, Fan Z. Exploring NamiRNA networks and time-series gene expression in osteogenic differentiation of adipose-derived stem cells. Ann Med 2025; 57:2478323. [PMID: 40100054 PMCID: PMC11921168 DOI: 10.1080/07853890.2025.2478323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 02/12/2025] [Accepted: 02/28/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Adipose-derived stem cells (ADSCs) are a type of stem cell found in adipose tissue with the capacity to differentiate into multiple lineages, including osteoblasts. The differentiation of ADSCs into osteoblasts underlies osteogenic and pathological cellular basis in osteoporosis, bone damage and repair. METHODS Focused on ADSCs osteogenic differentiation, we conducted mRNA, microRNA expression and bioinformatics analysis, including gene differential expression, time series-based trend analysis, functional enrichment, and generates potential nuclear activating miRNAs (NamiRNA) regulatory network. The screened mRNAs in NamiRNA regulatory network were validated with correlation analysis. RESULTS The NamiRNA Regulatory Network reveals 4 mRNAs (C12orf61, MIR31HG, NFE2L1, and PCYOX1L) significantly downregulated in differentiated group and may be associated with ADSCs stemness. Furthermore, the significantly upregulated 10 genes (ACTA2, TAGLN, LY6E, IFITM3, NGFRAP1, TCEAL4, ATP5C1, CAV1, RPSA, and KDELR3) were significantly enriched in osteogenic-related pathways, and negatively correlated with ADSCs cell stemness in vitro. CONCLUSION These findings uncover potential genes related to ADSCs osteogenic differentiation, and provide theoretical basis for underlying ADSCs osteogenic differentiation and related diseases.
Collapse
Affiliation(s)
- Xin Jin
- Department of Plastic Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yi Lu
- Department of Plastic Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhihong Fan
- Department of Plastic Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Nayak VV, Costello JP, Ehlen QT, Slavin BV, Mirsky NA, Kelly S, Suarez C, Daunert S, Witek L, Coelho PG. A rhPDGF-BB/bovine type I collagen/β-TCP mixture for the treatment of critically sized non-union tibial defects: An in vivo study in rabbits. J Orthop Res 2024; 42:1998-2006. [PMID: 38598203 DOI: 10.1002/jor.25847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/08/2024] [Accepted: 03/20/2024] [Indexed: 04/11/2024]
Abstract
Non-union during healing of bone fractures affects up to ~5% of patients worldwide. Given the success of recombinant human platelet-derived growth factor-B chain homodimer (rhPDGF-BB) in promoting angiogenesis and bone fusion in the hindfoot and ankle, rhPDGF-BB combined with bovine type I collagen/β-TCP matrix (AIBG) could serve as a viable alternative to autografts in the treatment of non-unions. Defects (~2 mm gaps) were surgically induced in tibiae of skeletally mature New Zealand white rabbits. Animals were allocated to one of four groups-(1) negative control (empty defect, healing for 8 weeks), (2 and 3) acute treatment with AIBG (healing for 4 or 8 weeks), and (4) chronic treatment with AIBG (injection 4 weeks post defect creation and then healing for 8 weeks). Bone formation was analyzed qualitatively and semi-quantitatively through histology. Samples were imaged using dual-energy X-ray absorptiometry and computed tomography for defect visualization and volumetric reconstruction, respectively. Delayed healing or non-healing was observed in the negative control group, whereas defects treated with AIBG in an acute setting yielded bone formation as early as 4 weeks with bone growth appearing discontinuous. At 8 weeks (acute setting), substantial remodeling was observed with higher degrees of bone organization characterized by appositional bone growth. The chronic healing, experimental, group yielded bone formation and remodeling, with no indication of non-union after treatment with AIBG. Furthermore, bone growth in the chronic healing group was accompanied by an increased presence of osteons, osteonal canals, and interstitial lamellae. Qualitatively and semiquantitatively, chronic application of AI facilitated complete bridging of the induced non-union defects, while untreated defects or defects treated acutely with AIBG demonstrated a lack of complete bridging at 8 weeks.
Collapse
Affiliation(s)
- Vasudev Vivekanand Nayak
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | - Quinn T Ehlen
- University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Blaire V Slavin
- University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | - Sophie Kelly
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, USA
| | - Camila Suarez
- Trinity College of Arts and Sciences, Duke University, Durham, North Carolina, USA
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Lukasz Witek
- Biomaterials Division, NYU Dentistry, New York, New York, USA
- Department of Biomedical Engineering, NYU Tandon School of Engineering, Brooklyn, New York, USA
- Hansjörg Wyss Department of Plastic Surgery, NYU Grossman School of Medicine, New York, New York, USA
| | - Paulo G Coelho
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida, USA
- DeWitt Daughtry Family Department of Surgery, Division of Plastic Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
3
|
Entezari A, Wu Q, Mirkhalaf M, Lu Z, Roohani I, Li Q, Dunstan CR, Jiang X, Zreiqat H. Unraveling the influence of channel size and shape in 3D printed ceramic scaffolds on osteogenesis. Acta Biomater 2024; 180:115-127. [PMID: 38642786 DOI: 10.1016/j.actbio.2024.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/31/2024] [Accepted: 04/11/2024] [Indexed: 04/22/2024]
Abstract
Bone has the capacity to regenerate itself for relatively small defects; however, this regenerative capacity is diminished in critical-size bone defects. The development of synthetic materials has risen as a distinct strategy to address this challenge. Effective synthetic materials to have emerged in recent years are bioceramic implants, which are biocompatible and highly bioactive. Yet nothing suitable for the repair of large bone defects has made the transition from laboratory to clinic. The clinical success of bioceramics has been shown to depend not only on the scaffold's intrinsic material properties but also on its internal porous geometry. This study aimed to systematically explore the implications of varying channel size, shape, and curvature in tissue scaffolds on in vivo bone regeneration outcomes. 3D printed bioceramic scaffolds with varying channel sizes (0.3 mm to 1.5 mm), shapes (circular vs rectangular), and curvatures (concave vs convex) were implanted in rabbit femoral defects for 8 weeks, followed by histological evaluation. We demonstrated that circular channel sizes of around 0.9 mm diameter significantly enhanced bone formation, compared to channel with diameters of 0.3 mm and 1.5 mm. Interestingly, varying channel shapes (rectangular vs circular) had no significant effect on the volume of newly formed bone. Furthermore, the present study systematically demonstrated the beneficial effect of concave surfaces on bone tissue growth in vivo, reinforcing previous in silico and in vitro findings. This study demonstrates that optimizing architectural configurations within ceramic scaffolds is crucial in enhancing bone regeneration outcomes. STATEMENT OF SIGNIFICANCE: Despite the explosion of work on developing synthetic scaffolds to repair bone defects, the amount of new bone formed by scaffolds in vivo remains suboptimal. Recent studies have illuminated the pivotal role of scaffolds' internal architecture in osteogenesis. However, these investigations have mostly remained confined to in silico and in vitro experiments. Among the in vivo studies conducted, there has been a lack of systematic analysis of individual architectural features. Herein, we utilized bioceramic 3D printing to conduct a systematic exploration of the effects of channel size, shape, and curvature on bone formation in vivo. Our results demonstrate the significant influence of channel size and curvature on in vivo outcomes. These findings provide invaluable insights into the design of more effective bone scaffolds.
Collapse
Affiliation(s)
- Ali Entezari
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, NSW 2007, Australia; Tissue Engineering and Biomaterials Research Unit, School of Biomedical Engineering, The University of Sydney, NSW 2006, Australia
| | - Qianju Wu
- Department of Prosthodontics, Oral Bioengineering, and Regenerative Medicine Lab, Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai 200011, China; Stomatological Hospital of Xiamen Medical College, Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, Fujian, China
| | - Mohammad Mirkhalaf
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, 2 George St Brisbane, QLD 4000, Australia; Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; Centre for Materials Science, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| | - Zufu Lu
- Tissue Engineering and Biomaterials Research Unit, School of Biomedical Engineering, The University of Sydney, NSW 2006, Australia
| | - Iman Roohani
- Tissue Engineering and Biomaterials Research Unit, School of Biomedical Engineering, The University of Sydney, NSW 2006, Australia
| | - Qing Li
- School of Aerospace, Mechanical and Mechatronic Engineering, The University of Sydney, Sydney, NSW 2006, Australia
| | - Colin R Dunstan
- Tissue Engineering and Biomaterials Research Unit, School of Biomedical Engineering, The University of Sydney, NSW 2006, Australia
| | - Xinquan Jiang
- Department of Prosthodontics, Oral Bioengineering, and Regenerative Medicine Lab, Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai 200011, China.
| | - Hala Zreiqat
- Tissue Engineering and Biomaterials Research Unit, School of Biomedical Engineering, The University of Sydney, NSW 2006, Australia.
| |
Collapse
|
4
|
Liu Z, Liu J, Li J, Li Y, Sun J, Deng Y, Zhou H. Substrate stiffness can affect the crosstalk between adipose derived mesenchymal stem cells and macrophages in bone tissue engineering. Front Bioeng Biotechnol 2023; 11:1133547. [PMID: 37576988 PMCID: PMC10415109 DOI: 10.3389/fbioe.2023.1133547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 06/15/2023] [Indexed: 08/15/2023] Open
Abstract
Purpose: This study aimed to explore the effect of biomaterials with different stiffness on Adipose Derived Mesenchymal Stem Cells (ADSC)-macrophage crosstalk in bone tissue engineering and its role in bone repair. Methods: Biomaterials with Young's modulus of 64 and 0.2 kPa were selected, and the crosstalk between ADSCs and macrophages was investigated by means of conditioned medium treatment and cell co-culture, respectively. Polymerase chain reaction (PCR) and flow cytometry were used to evaluate the polarization of macrophages. Alkaline phosphatase (ALP) and alizarin red staining (ARS) solutions were used to evaluate the osteogenic differentiation of ADSCs. Transwell assay was used to evaluate the chemotaxis of ADSCs and macrophages. Moreover, mass spectrometry proteomics was used to analyze the secreted protein profile of ADSCs of different substrates and macrophages in different polarization states. Results: On exploring the influence of biomaterials on macrophages from ADSCs on different substrates, we found that CD163 and CD206 expression levels in macrophages were significantly higher in the 64-kPa group than in the 0.2-kPa group in conditioned medium treatment and cell co-culture. Flow cytometry showed that more cells became CD163+ or CD206+ cells in the 64-kPa group under conditioned medium treatment or cell co-culture. The Transwell assay showed that more macrophages migrated to the lower chamber in the 64-kPa group. The proteomic analysis found that ADSCs in the 64-kPa group secreted more immunomodulatory proteins, such as LBP and RBP4, to improve the repair microenvironment. On exploring the influence of biomaterials on ADSCs from macrophages in different polarization states, we found that ALP and ARS levels in ADSCs were significantly higher in the M2 group than in the other three groups (NC, M0, and M1 groups) in both conditioned medium treatment and cell co-culture. The Transwell assay showed that more ADSCs migrated to the lower chamber in the M2 group. The proteomic analysis found that M2 macrophages secreted more extracellular remodeling proteins, such as LRP1, to promote bone repair. Conclusion: In bone tissue engineering, the stiffness of repair biomaterials can affect the crosstalk between ADSCs and macrophages, thereby regulating local repair immunity and affecting bone repair.
Collapse
Affiliation(s)
- Zeyang Liu
- Department of Ophthalmology, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jin Liu
- Department of Ophthalmology, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jipeng Li
- Department of Ophthalmology, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yinwei Li
- Department of Ophthalmology, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Sun
- Department of Ophthalmology, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuan Deng
- Department of Ophthalmology, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huifang Zhou
- Department of Ophthalmology, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
5
|
Xie X, Miao B, Yao J, Chen Z. Silk fibroin-hydroxyapatite scaffolds promote the proliferation of adipose-derived mesenchymal stem cells by activating the ERK signal. J Biomater Appl 2023; 37:1767-1775. [PMID: 37001507 DOI: 10.1177/08853282231168730] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Adipose-derived mesenchymal stem cell (Ad-MSC) with capacities of releasing trophic factors and chondrogenic differentiation was a promising candidate for tracheal reconstruction. Silk fibroin (SF)- hydroxyapatite (HA) scaffolds were fabricated by the freeze-drying method. And Ad-MSCs were co-cultured on the scaffolds for 14 days in vitro. The role of the SF-HA scaffold in regulating the adhesion, growth, and proliferation of Ad-MSCs, and its potential mechanisms were investigated. The identity of Ad-MSCs was confirmed by cell morphology, surface markers, and differentiation characteristics. Cell proliferation, viability, and morphology were observed via CCK-8, live/dead assay, and scanning electron microscopy (SEM). Gene mRNA and protein levels were examined using quantitative real-time polymerase chain reaction and western blotting, respectively. SF-HA scaffolds showed excellent properties of promoting Ad-MSCs adhesion, growth, and proliferation for at least 14 days. In the CCK-8 assay, the relative OD value of Ad-MSCs cultured on SF-HA scaffolds increased (p < 0.001). Furthermore, live/dead staining showed that the fluorescent coverage increased with time (p < 0.05). SEM also showed that 3 days after inoculation, the coverage of Ad-MSCs on the SF-HA scaffolds was 78.15%, increased to 92.91% on day 7, and reached a peak of 94.38% on day 14. Extracellular signal-regulated kinase (ERK) mRNA and phosphorylated ERK (pERK) protein expression increased at day 3 (p < 0.05), followed by a significant decline at day 7 (p < 0.05). And ERK mRNA expression was positively correlated with Ad-MSCs proliferation (p < 0.05). In summary, the SF-HA scaffold co-cultured with Ad-MSCs is a promising biomaterial for tracheal repair by activating the ERK signal pathway.
Collapse
Affiliation(s)
- Xingqiao Xie
- Department of Otorhinolaryngology-Head and Neck Surgery, Huashan Hospital Fudan University, Shanghai, China
| | - Bianliang Miao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science and the Laboratory of Advanced Materials, Fudan University, Shanghai, China
| | - Jinrong Yao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science and the Laboratory of Advanced Materials, Fudan University, Shanghai, China
| | - Zhongchun Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Huashan Hospital Fudan University, Shanghai, China
| |
Collapse
|
6
|
Shineh G, Patel K, Mobaraki M, Tayebi L. Functional Approaches in Promoting Vascularization and Angiogenesis in Bone Critical-Sized Defects via Delivery of Cells, Growth Factors, Drugs, and Particles. J Funct Biomater 2023; 14:99. [PMID: 36826899 PMCID: PMC9960138 DOI: 10.3390/jfb14020099] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Critical-sized bone defects, or CSDs, are defined as bone defects that cannot be regenerated by themselves and require surgical intervention via employing specific biomaterials and a certain regenerative strategy. Although a variety of approaches can be used to treat CSDs, poor angiogenesis and vascularization remain an obstacle in these methods. The complex biological healing of bone defects depends directly on the function of blood flow to provide sufficient oxygen and nutrients and the removal of waste products from the defect site. The absence of vascularization can lead to non-union and delayed-union defect development. To overcome this challenge, angiogenic agents can be delivered to the site of injury to stimulate vessel formation. This review begins by introducing the treatment methods for CSDs. The importance of vascularization in CSDs is subsequently highlighted. Delivering angiogenesis agents, including relevant growth factors, cells, drugs, particles, cell secretion substances, their combination, and co-delivery to CSDs are fully explored. Moreover, the effects of such agents on new bone formation, followed by vessel formation in defect areas, are evaluated.
Collapse
Affiliation(s)
- Ghazal Shineh
- School of Biomedical Engineering, University of Sydney, Sydney, NSW 2006, Australia
| | - Kishan Patel
- School of Dentistry, Marquette University, Milwaukee, WI 53207, USA
| | - Mohammadmahdi Mobaraki
- Biomaterial Group, Faculty of Biomedical Engineering, Amirkabir University of Technology, Tehran 15916-34311, Iran
| | - Lobat Tayebi
- School of Dentistry, Marquette University, Milwaukee, WI 53207, USA
| |
Collapse
|
7
|
Wang Z, Liu J, Huang Y, Liu Q, Chen M, Ji C, Feng J, Ma Y. Pituitary Adenylate Cyclase-activating Polypeptide (PACAP) -derived Peptide MPAPO Stimulates Adipogenic Differentiation by Regulating the Early Stage of Adipogenesis and ERK Signaling Pathway. Stem Cell Rev Rep 2023; 19:516-530. [PMID: 36112309 DOI: 10.1007/s12015-022-10415-y] [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] [Accepted: 06/15/2022] [Indexed: 02/07/2023]
Abstract
Regenerative medicine and tissue engineering have delivered new healing possibilities to the treatment of soft tissue defects, but the selection of seed cells is critical for treatment. Adipose-derived stem cells have perpetually been a preferred candidate for seed cells due to their wealthy sources, simple access, high plasticity, and powerful value-added capabilities. How to improve the efficiency of adipogenic differentiation is the key to the treatment. Pituitary adenylate cyclase-activating peptide, as a biologically active peptide secreted by the pituitary, is widely involved in regulating the body's sugar metabolism and lipid metabolism. However, the effects of MPAPO in ADSCs adipogenic differentiation remain unknown. Our results reveal that MPAPO treatment improves the adipogenic differentiation efficiency of ADSCs, including promoting the accumulation of lipid droplets and triglycerides, and the expression of adipocyte protein biomarkers PPARγ and C/EBPa. Additionally, the mechanism studies showed that the effective window of MPAPO-induced adipogenesis was the first 3 days during ADSCs differentiation. MPAPO selectively binds to the PAC1 receptor and promotes adipogenic differentiation of ADSCs by activating the ERK signaling pathway and elevating cell proliferation during postconfluent mitosis stage. Altogether, we demonstrate that MPAPO plays a crucial role in ADSCs adipogenesis, providing experimental basis and data for exploring therapeutic options in tissue defect repair.
Collapse
Affiliation(s)
- Zixian Wang
- Department of Cellular Biology, Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, China
| | - Jianmin Liu
- Department of Cellular Biology, Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, China
| | - Yongmei Huang
- Department of Cellular Biology, Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, China
| | - Qian Liu
- Department of Cellular Biology, Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, China
| | - Meng Chen
- Department of Cellular Biology, Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, China
| | - Chunyan Ji
- Department of Cellular Biology, Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, China
| | - Jia Feng
- Department of Cellular Biology, Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, China
| | - Yi Ma
- Department of Cellular Biology, Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, China.
- Department of Cellular Biology, Institute of Biomedicine, Jinan University, 601 Huangpu Avenue West, 510632, Guangzhou, China.
| |
Collapse
|
8
|
Li J, Chen X, Ren L, Chen X, Wu T, Wang Y, Ren X, Cheng B, Xia J. Type H vessel/platelet-derived growth factor receptor β + perivascular cell disintegration is involved in vascular injury and bone loss in radiation-induced bone damage. Cell Prolif 2023:e13406. [PMID: 36694343 DOI: 10.1111/cpr.13406] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 01/26/2023] Open
Abstract
Collapse of the microvascular system is a prerequisite for radiation-induced bone loss. Since type H vessels, a specific bone vessel subtype surrounded by platelet-derived growth factor receptor β+ (PDGFRβ+ ) perivascular cells (PVCs), has been recently identified to couple angiogenesis and osteogenesis, we hypothesize that type H vessel injury initiates PDGFRβ+ PVC dysfunction, which contributes to the abnormal angiogenesis and osteogenesis after irradiation. In this study, we found that radiation led to the decrease of both type H endothelial cell (EC) and PDGFRβ+ PVC numbers. Remarkably, results from lineage tracing showed that PDGFRβ+ PVCs detached from microvessels and converted the lineage commitment from osteoblasts to adipocytes, leading to vascular injury and bone loss after irradiation. These phenotype transitions above were further verified to be associated with the decrease in hypoxia-inducible factor-1α (HIF-1α)/PDGF-BB/PDGFRβ signalling between type H ECs and PDGFRβ+ PVCs. Pharmacological blockade of HIF-1α/PDGF-BB/PDGFRβ signalling induced a phenotype similar to radiation-induced bone damage, while the rescue of this signalling significantly alleviated radiation-induced bone injury. Our findings show that the decrease in HIF-1α/PDGF-BB/PDGFRβ signalling between type H ECs and PDGFRβ+ PVCs after irradiation affects the homeostasis of EC-PVC coupling and plays a part in vascular damage and bone loss, which has broad implications for effective translational therapies.
Collapse
Affiliation(s)
- Jiayan Li
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xiaodan Chen
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Lin Ren
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xijuan Chen
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Tong Wu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yun Wang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xianyue Ren
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Bin Cheng
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Juan Xia
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
9
|
Zhang HJ, Li FS, Wang F, Wang H, He TC, Reid RR, He BC, Xia Q. Transgenic PDGF-BB sericin hydrogel potentiates bone regeneration of BMP9-stimulated mesenchymal stem cells through a crosstalk of the Smad-STAT pathways. Regen Biomater 2022; 10:rbac095. [PMID: 36683747 PMCID: PMC9847547 DOI: 10.1093/rb/rbac095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/08/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
Silk as a natural biomaterial is considered as a promising bone substitute in tissue regeneration. Sericin and fibroin are the main components of silk and display unique features for their programmable mechanical properties, biocompatibility, biodegradability and morphological plasticity. It has been reported that sericin recombinant growth factors (GFs) can support cell proliferation and induce stem cell differentiation through cross-talk of signaling pathways during tissue regeneration. The transgenic technology allows the productions of bioactive heterologous GFs as fusion proteins with sericin, which are then fabricated into solid matrix or hydrogel format. Herein, using an injectable hydrogel derived from transgenic platelet-derived GF (PDGF)-BB silk sericin, we demonstrated that the PDGF-BB sericin hydrogel effectively augmented osteogenesis induced by bone morphogenetic protein (BMP9)-stimulated mesenchymal stem cells (MSCs) in vivo and in vitro, while inhibiting adipogenic differentiation. Further gene expression and protein-protein interactions studies demonstrated that BMP9 and PDGF-BB synergistically induced osteogenic differentiation through the cross-talk between Smad and Stat3 pathways in MSCs. Thus, our results provide a novel strategy to encapsulate osteogenic factors and osteoblastic progenitors in transgenic sericin-based hydrogel for robust bone tissue engineering.
Collapse
Affiliation(s)
- Hui-Jie Zhang
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
| | - Fu-Shu Li
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Department of Pharmacy, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing 400014, China
| | - Feng Wang
- Biological Science Research Center, Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing 400715, China
| | - Han Wang
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Department of Pharmacy, Panzhou People’s Hospital, Guizhou 553599, China
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Russell R Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Laboratory of Craniofacial Biology and Development, Department of Surgery Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Bai-Cheng He
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Qingyou Xia
- Biological Science Research Center, Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing 400715, China
| |
Collapse
|
10
|
Strontium-incorporated bioceramic scaffolds for enhanced osteoporosis bone regeneration. Bone Res 2022; 10:55. [PMID: 35999199 PMCID: PMC9399250 DOI: 10.1038/s41413-022-00224-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/15/2022] [Accepted: 05/29/2022] [Indexed: 11/23/2022] Open
Abstract
The restoration of bone defects caused by osteoporosis remains a challenge for surgeons. Strontium ranelate has been applied in preventative treatment approaches due to the biological functions of the trace element strontium (Sr). In this study, we aimed to fabricate bioactive scaffolds through Sr incorporation based on our previously developed modified amino-functional mesoporous bioactive glass (MBG) and to systematically investigate the bioactivity of the resulting scaffold in vitro and in vivo in an osteoporotic rat model. The results suggested that Sr-incorporated amino-functional MBG scaffolds possessed favorable biocompatibility. Moreover, with the incorporation of Sr, osteogenic and angiogenic capacities were upregulated in vitro. The in vivo results showed that the Sr-incorporated amino-functional MBG scaffolds achieved better bone regeneration and vessel formation. Furthermore, bioinformatics analysis indicated that the Sr-incorporated amino-functional MBG scaffolds could reduce reactive oxygen species levels in bone marrow mesenchymal stem cells in the osteoporotic model by activating the cAMP/PKA signaling pathway, thus playing an anti-osteoporosis role while promoting osteogenesis. This study demonstrated the feasibility of incorporating trace elements into scaffolds and provided new insights into biomaterial design for facilitating bone regeneration in the treatment of osteoporosis.
Collapse
|
11
|
Cao L, Su H, Si M, Xu J, Chang X, Lv J, Zhai Y. Tissue Engineering in Stomatology: A Review of Potential Approaches for Oral Disease Treatments. Front Bioeng Biotechnol 2021; 9:662418. [PMID: 34820359 PMCID: PMC8606749 DOI: 10.3389/fbioe.2021.662418] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 10/01/2021] [Indexed: 01/09/2023] Open
Abstract
Tissue engineering is an emerging discipline that combines engineering and life sciences. It can construct functional biological structures in vivo or in vitro to replace native tissues or organs and minimize serious shortages of donor organs during tissue and organ reconstruction or transplantation. Organ transplantation has achieved success by using the tissue-engineered heart, liver, kidney, and other artificial organs, and the emergence of tissue-engineered bone also provides a new approach for the healing of human bone defects. In recent years, tissue engineering technology has gradually become an important technical method for dentistry research, and its application in stomatology-related research has also obtained impressive achievements. The purpose of this review is to summarize the research advances of tissue engineering and its application in stomatology. These aspects include tooth, periodontal, dental implant, cleft palate, oral and maxillofacial skin or mucosa, and oral and maxillofacial bone tissue engineering. In addition, this article also summarizes the commonly used cells, scaffolds, and growth factors in stomatology and discusses the limitations of tissue engineering in stomatology from the perspective of cells, scaffolds, and clinical applications.
Collapse
Affiliation(s)
- Lilan Cao
- School of Stomatology, Henan University, Kaifeng, China
| | - Huiying Su
- School of Stomatology, Henan University, Kaifeng, China
| | - Mengying Si
- School of Stomatology, Henan University, Kaifeng, China
| | - Jing Xu
- School of Stomatology, Henan University, Kaifeng, China
| | - Xin Chang
- School of Stomatology, Henan University, Kaifeng, China
| | - Jiajia Lv
- School of Stomatology, Henan University, Kaifeng, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Kaifeng, China
| | - Yuankun Zhai
- School of Stomatology, Henan University, Kaifeng, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Kaifeng, China
| |
Collapse
|
12
|
Vadaye Kheiry E, Fazly Bazzaz BS, Kerachian MA. Implantation of stem cells on synthetic or biological scaffolds: an overview of bone regeneration. Biotechnol Genet Eng Rev 2021; 37:238-268. [PMID: 34789069 DOI: 10.1080/02648725.2021.2003590] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Humans are exposed to a wide range of bone tissue injuries. In severe cases, bone damages could be only treated with transplantation of autologous or allogeneic grafting.In recent years, tissue engineering has become a promising strategy for repairing damaged organs and tissues, providing a great opportunity to cure several diseases. Bone tissue engineering consists of three components: scaffold, cells, and growth factors. Current bone tissue engineering strategies combine the use of stem cells with biologically active materials and gene therapy to mimic the natural microenvironment of bone. The combination of the scaffold with growth factors and extracellular matrix protein molecules can promote cell attachment, proliferation, and induce osteogenesis, which could provide signals for cell migration to begin the healing process during repair and bone formation.This article reviews the principles of bone regeneration and the most current developments of bone tissue engineering related to bone growth factors, the biologically active materials, such as bacterial cellulose, and stem cells.
Collapse
Affiliation(s)
- Elahe Vadaye Kheiry
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Bibi Sedigheh Fazly Bazzaz
- Biotechnology Research Center, Pharmaceutical Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Amin Kerachian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
13
|
Madecassic acid protects human periodontal ligament fibroblasts against hydrogen peroxide-induced cell damage by maintaining mitochondrial membrane potential. Mol Cell Toxicol 2021. [DOI: 10.1007/s13273-021-00174-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
14
|
The platelet derived growth factor BB promotes osteogenic differentiation of periodontal ligament stem cells via the Wnt/β-catenin signaling pathway. Arch Oral Biol 2021; 129:105162. [PMID: 34087681 DOI: 10.1016/j.archoralbio.2021.105162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/18/2021] [Accepted: 05/18/2021] [Indexed: 11/23/2022]
Abstract
OBJECTIVE To determine the role of platelet derived growth factor BB in the regulation of cell cycle, migration and differentiation of stem cells. DESIGN The gene was overexpressed in periodontal ligament stem cells using lentiviral vectors. Normal stem cells and empty lentiviral vectors-transfected were used as controls. Real time-PCR, western blotting, Cell Counting Kit-8 assay, flow cytometry, cell scratch test, Alkaline phosphatase activity assay, cell cycle analyses were conducted to assess the biological properties of stem cells. In addition, the effect of platelet derived growth factor BB on the Wnt/β-catenin signaling pathway were assessed by western blotting and immunofluorescent staining. RESULTS The gene was successfully overexpressed in periodontal ligament stem cells. The Cell Counting Kit-8, scratch test and cell cycle experiments proved that platelet derived growth factor BB promoted stem cells proliferation, migration and cell cycle progression. The Real time-PCR results showed that the Osterix (OSX) and Bone Morphogenetic Protein 2 (BMP2) genes in the overexpression group were significantly higher than those in the control group, but the Peroxisome Proliferators-activated Receptors (PPARγ) and Glycogen Synthase Kinase-3β (GSK-3β) gene were lower than that in the control group. Western blotting results also indicated that the Collagen Type 1 (COL-1), BMP2, Wnt1 and β-catenin proteins were increased in the overexpression group. In addition, the expression level of β-catenin protein in the cell nuclei was higher than that in the control group. CONCLUSIONS In conclusion, overexpression of platelet derived growth factor BB promoted cell proliferation, migration, cell cycle progression and decreased adipogenic differentiation. Furthermore, platelet derived growth factor BB regulated osteogenic differentiation of stem cells through the Wnt/β-catenin signaling pathway.
Collapse
|
15
|
Contreras O, Córdova-Casanova A, Brandan E. PDGF-PDGFR network differentially regulates the fate, migration, proliferation, and cell cycle progression of myogenic cells. Cell Signal 2021; 84:110036. [PMID: 33971280 DOI: 10.1016/j.cellsig.2021.110036] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/22/2022]
Abstract
Platelet-derived growth factors (PDGFs) regulate embryonic development, tissue regeneration, and wound healing through their binding to PDGF receptors, PDGFRα and PDGFRβ. However, the role of PDGF signaling in regulating muscle development and regeneration remains elusive, and the cellular and molecular responses of myogenic cells are understudied. Here, we explore the PDGF-PDGFR gene expression changes and their involvement in skeletal muscle myogenesis and myogenic fate. By surveying bulk RNA sequencing and single-cell profiling data of skeletal muscle stem cells, we show that myogenic progenitors and muscle stem cells differentially express PDGF ligands and PDGF receptors during myogenesis. Quiescent adult muscle stem cells and myoblasts preferentially express PDGFRβ over PDGFRα. Remarkably, cell culture- and injury-induced muscle stem cell activation altered PDGF family gene expression. In myoblasts, PDGF-AB and PDGF-BB treatments activate two pro-chemotactic and pro-mitogenic downstream transducers, RAS-ERK1/2 and PI3K-AKT. PDGFRs inhibitor AG1296 inhibited ERK1/2 and AKT activation, myoblast migration, proliferation, and cell cycle progression induced by PDGF-AB and PDGF-BB. We also found that AG1296 causes myoblast G0/G1 cell cycle arrest. Remarkably, PDGF-AA did not promote a noticeable ERK1/2 or AKT activation, myoblast migration, or expansion. Also, myogenic differentiation reduced the expression of both PDGFRα and PDGFRβ, whereas forced PDGFRα expression impaired myogenesis. Thus, our data highlight PDGF signaling pathway to stimulate satellite cell proliferation aiming to enhance skeletal muscle regeneration and provide a deeper understanding of the role of PDGF signaling in non-fibroblastic cells.
Collapse
Affiliation(s)
- Osvaldo Contreras
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Kensington 2052, Australia; Departamento de Biología Celular y Molecular and Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile.
| | - Adriana Córdova-Casanova
- Departamento de Biología Celular y Molecular and Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - Enrique Brandan
- Departamento de Biología Celular y Molecular and Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile; Fundación Ciencia & Vida, 7780272 Santiago, Chile
| |
Collapse
|
16
|
Dziedzic DSM, Francisco JC, Mogharbel BF, Irioda AC, Stricker PEF, Floriano J, de Noronha L, Abdelwahid E, Franco CRC, de Carvalho KAT. Combined Biomaterials: Amniotic Membrane and Adipose Tissue to Restore Injured Bone as Promoter of Calcification in Bone Regeneration: Preclinical Model. Calcif Tissue Int 2021; 108:667-679. [PMID: 33420810 PMCID: PMC8064990 DOI: 10.1007/s00223-020-00793-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023]
Abstract
Discarded tissues, like human amniotic membranes and adipose tissue, were investigated for the application of Decellularized Human Amniotic Membrane (DAM) as a viable scaffold for transplantation of Adipose-derived stromal cells (ASCs) in bone regeneration of non-healing calvarial defects in rats. Amniotic membrane was decellularized to provide a scaffold for male Wistar rats ASCs expansion and transplantation. ASCs osteoinduction in vitro promoted the deposition of a mineralized bone-like matrix by ASCs, as calcified globular accretions associated with the cells on the DAM surface and inside the collagenous matrix. Non-healing calvarial defects on male Wistar rats were randomly divided in control without treatment, treatment with four layers of DAM, or four layers of DAM associated with ASCs. After 12 weeks, tissue blocks were examined by micro-computed tomography and histology. DAM promoted osteoconduction by increasing the collagenous matrix on both DAM treatments. DAM with ASCs stimulated bone deposition, demonstrated by a higher percentage of bone volume and trabecular bone number, compared to control. Besides the osteogenic capacity in vitro, ASCs stimulated the healing of calvarial defects with significant DAM graft incorporation concomitant with higher host bone deposition. The enhanced in vivo bone regeneration by undifferentiated ASCs loaded onto DAM confirmed the potential of an easily collected autologous cell source associated with a broadly available collagenous matrix in tissue engineering.
Collapse
Affiliation(s)
- Dilcele Silva Moreira Dziedzic
- Cell Therapy and Biotechnology in Regenerative Medicine Department, The Pelé Pequeno Príncipe Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Ave. Silva Jardim, no. 1632, Box 80240-020, Curitiba, Paraná Brazil
| | - Júlio César Francisco
- Positivo University, St.Professor Pedro Viriato Parigot de Souza, Box 80710-570, Curitiba, Paraná 5300 Brazil
| | - Bassam Felipe Mogharbel
- Cell Therapy and Biotechnology in Regenerative Medicine Department, The Pelé Pequeno Príncipe Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Ave. Silva Jardim, no. 1632, Box 80240-020, Curitiba, Paraná Brazil
| | - Ana Carolina Irioda
- Cell Therapy and Biotechnology in Regenerative Medicine Department, The Pelé Pequeno Príncipe Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Ave. Silva Jardim, no. 1632, Box 80240-020, Curitiba, Paraná Brazil
| | - Priscila Elias Ferreira Stricker
- Cell Therapy and Biotechnology in Regenerative Medicine Department, The Pelé Pequeno Príncipe Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Ave. Silva Jardim, no. 1632, Box 80240-020, Curitiba, Paraná Brazil
| | - Juliana Floriano
- Physics Department, São Paulo State University (UNESP), Ave. Eng. Luís Edmundo Carrijo Coube, 2085 - Núcleo Res. Pres. Geisel, Box 17033-360, Bauru, São Paulo Brazil
| | - Lúcia de Noronha
- Pathology Department, The Institute of Biological and Health Sciences of the Pontifical Catholic University, Ave. Imaculada Conceição, 1155, Box 80215-901, Curitiba, Brazil
| | - Eltyeb Abdelwahid
- Feinberg School of Medicine, Feinberg Cardiovascular Research Institute, Northwestern University, 303 E. Chicago Ave., Tarry 14–725, Chicago, IL 60611 USA
| | - Célia Regina Cavichiolo Franco
- Cell Biology Department, Federal University of Paraná, Ave. Coronel Francisco Heráclito dos Santos 210, Box 81531-970, Curitiba, Paraná Brazil
| | - Katherine Athayde Teixeira de Carvalho
- Cell Therapy and Biotechnology in Regenerative Medicine Department, The Pelé Pequeno Príncipe Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Ave. Silva Jardim, no. 1632, Box 80240-020, Curitiba, Paraná Brazil
| |
Collapse
|
17
|
Paek SC, Min SK, Park JB. Effects of platelet-derived growth factor-BB on cellular morphology and cellular viability of stem cell spheroids composed of bone-marrow-derived stem cells. Biomed Rep 2020; 13:59. [PMID: 33123373 PMCID: PMC7583700 DOI: 10.3892/br.2020.1366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022] Open
Abstract
Platelet-derived growth factor-BB (PDGF-BB) is a potent mitogenic, angiogenic and chemoattractant, and is one of the most abundant growth factors in platelet-derived products. The goal of the present study was to examine the effects of PDGF-BB on cellular morphology and cellular viability using 3D stem cell cultures. On day 1, spheroids formed well in silicon-elastomer-based concave microwells. The addition of 10 or 100 ng/ml PDGF-BB did not affect the morphology of the cell spheroids. During longer periods of incubation, the cell spheroids maintained their shape without noticeable alterations. The majority of cells in the spheroids exhibited green fluorescence when analyzed using a live/dead assay, indicative of live cells. On day 1, the Cell Counting Kit-8 (CCK-8) assay values for PDGF-BB at 0, 10 and 100 ng/ml were 0.241±0.003, 0.227±0.001 and 0.241±0.004, respectively; on day 3, the CCK-8 assay values for PDGF-BB were 0.233±0.005, 0.278±0.001 and 0.194±0.003, respectively; and on day 7, they were 0.248±0.014, 0.293±0.031 and 0.346±0.034, respectively. The 100 ng/ml group showed significantly higher values compared with the control group on day 7. Together, the results of the present study showed that the addition of 10 and 100 ng/ml PDGF-BB increased cellular viability, suggesting that PDGF-BB may be usable in cell therapy.
Collapse
Affiliation(s)
- Soung-Chu Paek
- Department of Dental Implantology, Graduate School of Clinical Dental Science, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sae Kyung Min
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jun-Beom Park
- Department of Dental Implantology, Graduate School of Clinical Dental Science, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
18
|
Aimaiti A, Wahafu T, Keremu A, Yicheng L, Li C. Strontium Ameliorates Glucocorticoid Inhibition of Osteogenesis Via the ERK Signaling Pathway. Biol Trace Elem Res 2020; 197:591-598. [PMID: 31832923 DOI: 10.1007/s12011-019-02009-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 12/05/2019] [Indexed: 02/06/2023]
Abstract
Glucocorticoid (GC) has been widely used in clinical work due to its anti-inflammatory and immune-inhibitory properties. However, long-term or high-dose administration is associated with side effects, such as GC-induced osteoporosis (GIOP), which causes great pain for and poses a heavy financial burden on patients. We sought to investigate the potential effects of strontium on GIOP and further explore its underlying mechanisms, including its reversal of the inhibitory effect of GC on osteogenesis of bone marrow-derived mesenchymal stem cells (BMSCs). We incubated BMSCs with Dexamethasone (DEX) in combination with or without strontium and then measured osteogenic and adipogenic gene expression levels by RT-qPCR and Western blot. We added a specific ERK signaling pathway inhibitor, U0126, to evaluate the involvement of that pathway. Strontium promoted osteogenic differentiation and matrix mineralization in DEX-treated BMSCs, accompanied by upregulation of RUNX2, Osx, ALP, BSP, COL1A1, and OCN. DEX blocked the expression of several osteogenesis-related marker genes by activating the ERK signaling pathway. U0126 attenuated the suppression of osteogenesis in DEX-treated BMSCs. These results suggested that strontium could enhance osteogenic differentiation and matrix mineralization by counteracting DEX's inhibitory effect on osteogenesis via the ERK signaling pathway. Therefore, strontium might be a promising therapeutic agent for GIOP.
Collapse
Affiliation(s)
- Abudousaimi Aimaiti
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, 137 South LiYuShan Road, Urumqi, 830054, Xinjiang, China
| | - Tuerhongjiang Wahafu
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, 137 South LiYuShan Road, Urumqi, 830054, Xinjiang, China
| | - Ajimu Keremu
- Orthopedic Center, First People's Hospital of Kashgar, Kashgar, 844000, Xinjiang, China
| | - Li Yicheng
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, 137 South LiYuShan Road, Urumqi, 830054, Xinjiang, China
| | - Cao Li
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, 137 South LiYuShan Road, Urumqi, 830054, Xinjiang, China.
| |
Collapse
|
19
|
Jin Y, Ding L, Ding Z, Fu Y, Song Y, Jing Y, Li Q, Zhang J, Ni Y, Hu Q. Tensile force-induced PDGF-BB/PDGFRβ signals in periodontal ligament fibroblasts activate JAK2/STAT3 for orthodontic tooth movement. Sci Rep 2020; 10:11269. [PMID: 32647179 PMCID: PMC7347599 DOI: 10.1038/s41598-020-68068-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022] Open
Abstract
Orthodontic force-induced osteogenic differentiation and bone formation at tension side play a pivotal role in orthodontic tooth movement (OTM). Platelet-derived growth factor-BB (PDGF-BB) is a clinically proven growth factor during bone regeneration process with unclear mechanisms. Fibroblasts in periodontal ligament (PDL) are considered to be mechanosensitive under orthodontic force. Thus, we established OTM model to investigate the correlation between PDGF-BB and fibroblasts during bone regeneration at tension side. We confirmed that tensile force stimulated PDL cells to induce osteogenic differentiation via Runx-2, OCN up-regulation, and to accelerate new bone deposition along the periodontium and the alveolar bone interface. Interestingly, PDGF-BB level was remarkably enhanced at tension side during OTM in parallel with up-regulated PDGFRβ+/α-SMA+ fibroblasts in PDL by immunohistochemistry. Moreover, orthodontic force-treated primary fibroblasts from PDL were isolated and, cultured in vitro, which showed similar morphology and phenotype with control fibroblasts without OTM treatment. PDGFRβ expression was confirmed to be increased in orthodontic force-treated fibroblasts by immunofluorescence and flow cytometry. Bioinformatics analysis identified that PDGF-BB/PDGFRβ signals were relevant to the activation of JAK/STAT3 signals. The protein expression of JAK2 and STAT3 was elevated in PDL of tension side. Importantly, in vivo, the treatment of the inhibitors (imatinib and AG490) for PDGFRβ and JAK-STAT signals were capable of attenuating the tooth movement. The osteogenic differentiation and bone regeneration in tension side were down-regulated upon the treatment of inhibitors during OTM. Meanwhile, the expressions of PDGFRβ, JAK2 and STAT3 were inhibited by imatinib and AG490. Thus, we concluded that tensile force-induced PDGF-BB activated JAK2/STAT3 signals in PDGFRβ+ fibroblasts in bone formation during OTM.
Collapse
Affiliation(s)
- Yuqin Jin
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Liang Ding
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, No. 30 Zhongyang Road, Nanjing, 210008, China
| | - Zhuang Ding
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, No. 30 Zhongyang Road, Nanjing, 210008, China
| | - Yong Fu
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, No. 30 Zhongyang Road, Nanjing, 210008, China
| | - Yuxian Song
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, No. 30 Zhongyang Road, Nanjing, 210008, China
| | - Yue Jing
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, No. 30 Zhongyang Road, Nanjing, 210008, China
| | - Qiang Li
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, No. 30 Zhongyang Road, Nanjing, 210008, China
| | - Jianyun Zhang
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yanhong Ni
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, No. 30 Zhongyang Road, Nanjing, 210008, China.
| | - Qingang Hu
- Department of Oral and Maxillofacial Surgery, Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, No. 30 Zhongyang Road, Nanjing, 210008, China.
| |
Collapse
|
20
|
Ibrahim A, Rodriguez-Florez N, Gardner OFW, Zucchelli E, New SEP, Borghi A, Dunaway D, Bulstrode NW, Ferretti P. Three-dimensional environment and vascularization induce osteogenic maturation of human adipose-derived stem cells comparable to that of bone-derived progenitors. Stem Cells Transl Med 2020; 9:1651-1666. [PMID: 32639692 PMCID: PMC7695642 DOI: 10.1002/sctm.19-0207] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 05/26/2020] [Accepted: 06/10/2020] [Indexed: 12/12/2022] Open
Abstract
While human adipose‐derived stem cells (hADSCs) are known to possess osteogenic differentiation potential, the bone tissues formed are generally considered rudimentary and immature compared with those made by bone‐derived precursor cells such as human bone marrow‐derived mesenchymal stem cells (hBMSCs) and less commonly studied human calvarium osteoprogenitor cells (hOPs). Traditional differentiation protocols have tended to focus on osteoinduction of hADSCs through the addition of osteogenic differentiation media or use of stimulatory bioactive scaffolds which have not resulted in mature bone formation. Here, we tested the hypothesis that by reproducing the physical as well as biochemical bone microenvironment through the use of three‐dimensional (3D) culture and vascularization we could enhance osteogenic maturation in hADSCs. In addition to biomolecular characterization, we performed structural analysis through extracellular collagen alignment and mineral density in our bone tissue engineered samples to evaluate osteogenic maturation. We further compared bone formed by hADSCs, hBMSCs, and hOPs against mature human pediatric calvarial bone, yet not extensively investigated. Although bone generated by all three cell types was still less mature than native pediatric bone, a fibrin‐based 3D microenvironment together with vascularization boosted osteogenic maturation of hADSC making it similar to that of bone‐derived osteoprogenitors. This demonstrates the important role of vascularization and 3D culture in driving osteogenic maturation of cells easily available but constitutively less committed to this lineage and suggests a crucial avenue for recreating the bone microenvironment for tissue engineering of mature craniofacial bone tissues from pediatric hADSCs, as well as hBMSCs and hOPs.
Collapse
Affiliation(s)
- Amel Ibrahim
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK.,Department of Plastic Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Naiara Rodriguez-Florez
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK.,Department of Plastic Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.,TECNUN Escuela de Ingenieros, Universidad de Navarra, San Sebastian, Spain
| | - Oliver F W Gardner
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Eleonora Zucchelli
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Sophie E P New
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Alessandro Borghi
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK.,Department of Plastic Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - David Dunaway
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK.,Department of Plastic Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Neil W Bulstrode
- Department of Plastic Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Patrizia Ferretti
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
21
|
Zhong YC, Wang SC, Han YH, Wen Y. Recent Advance in Source, Property, Differentiation, and Applications of Infrapatellar Fat Pad Adipose-Derived Stem Cells. Stem Cells Int 2020; 2020:2560174. [PMID: 32215015 PMCID: PMC7081037 DOI: 10.1155/2020/2560174] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/12/2020] [Accepted: 02/20/2020] [Indexed: 12/18/2022] Open
Abstract
Infrapatellar fat pad (IPFP) can be easily obtained during knee surgery, which avoids the damage to patients for obtaining IPFP. Infrapatellar fat pad adipose-derived stem cells (IPFP-ASCs) are also called infrapatellar fat pad mesenchymal stem cells (IPFP-MSCs) because the morphology of IPFP-ASCs is similar to that of bone marrow mesenchymal stem cells (BM-MSCs). IPFP-ASCs are attracting more and more attention due to their characteristics suitable to regenerative medicine such as strong proliferation and differentiation, anti-inflammation, antiaging, secreting cytokines, multipotential capacity, and 3D culture. IPFP-ASCs can repair articular cartilage and relieve the pain caused by osteoarthritis, so most of IPFP-related review articles focus on osteoarthritis. This article reviews the anatomy and function of IPFP, as well as the discovery, amplification, multipotential capacity, and application of IPFP-ASCs in order to explain why IPFP-ASC is a superior stem cell source in regenerative medicine.
Collapse
Affiliation(s)
- Yu-chen Zhong
- Department of Histology and Embryology, College of Basic Medical Sciences, China Medical University, Shenyang 110122, China
- Class 4, Phase 102, China Medical University, Shenyang 110122, China
| | - Shi-chun Wang
- Department of Histology and Embryology, College of Basic Medical Sciences, China Medical University, Shenyang 110122, China
- Class 4, Phase 102, China Medical University, Shenyang 110122, China
| | - Yin-he Han
- Department of Histology and Embryology, College of Basic Medical Sciences, China Medical University, Shenyang 110122, China
| | - Yu Wen
- Department of Histology and Embryology, College of Basic Medical Sciences, China Medical University, Shenyang 110122, China
| |
Collapse
|
22
|
Deng J, Pan J, Han X, Yu L, Chen J, Zhang W, Zhu L, Huang W, Liu S, You Z, Liu Y. PDGFBB-modified stem cells from apical papilla and thermosensitive hydrogel scaffolds induced bone regeneration. Chem Biol Interact 2019; 316:108931. [PMID: 31874163 DOI: 10.1016/j.cbi.2019.108931] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 11/30/2019] [Accepted: 12/17/2019] [Indexed: 12/17/2022]
Abstract
Bone defects caused by cancer surgery or trauma have a strong negative impact on human health. Treatment with cell and material-based complexes provides an alternative strategy for the regeneration of damaged bone tissue. The good physical properties and suitable biodegradability of a thermosensitive hydrogel has been shown to act as a valuable scaffold. Platelet derived growth factor BB (PDGFBB) is mainly secreted by platelets and promotes the migration and angiogenesis of mesenchymal stem cells (MSCs). Although PDGFBB is known to indirectly enhance bone repair in vivo, the effects of PDGFBB on stem cells from apical papilla (SCAPs) require further investigation. In our study, the proliferation of cells was investigated by the cell counting kit-8 and live/dead staining methods. The results indicated that PDGFBB promoted the proliferation of SCAPs. Real-time polymerase chain reaction and Western blot experiments were used to detect osteogenic genes and proteins. Moreover, calvarial defects were created in Sprague-Dawley rats and different complexes implanted. The results shown by micro-CT and hematoxylin and eosin analysis demonstrated that the hydrogel combined with lentiviral supernatant-green fluorescent protein-PDGFBB significantly improved new bone formation and mineralization compared with the other three groups. In summary, our research showed that a thermosensitive hydrogel can be used as a scaffold for 3D cell culture, and PDGFBB gene-modified SCAPs can improve bone formation in calvarial defects.
Collapse
Affiliation(s)
- Jiajia Deng
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, PR China; Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, PR China
| | - Jie Pan
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, PR China; Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, PR China
| | - Xinxin Han
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, PR China
| | - Liming Yu
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, PR China; Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, PR China
| | - Jing Chen
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, PR China; Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, PR China
| | - Weihua Zhang
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, PR China; Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, PR China
| | - Luying Zhu
- Xiangya School of Stomatology, Xiangya Stomatology Hospital, Central South University, Changsha, China
| | - Wei Huang
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, PR China
| | - Shangfeng Liu
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, PR China
| | - Zhengwei You
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Belt and Road Joint Laboratory of Advanced Fiber and Low-dimension Materials, Donghua University, Shanghai, 201620, China
| | - Yuehua Liu
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, PR China; Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, PR China.
| |
Collapse
|
23
|
Functionalization of Silk Fibers by PDGF and Bioceramics for Bone Tissue Regeneration. COATINGS 2019. [DOI: 10.3390/coatings10010008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Bone regeneration is a complex, well-organized physiological process of bone formation observed during normal fracture healing and involved in continuous remodeling throughout adult life. An ideal medical device for bone regeneration requires interconnected pores within the device to allow for penetration of blood vessels and cells, enabling material biodegradation and bone ingrowth. Additional mandatory characteristics include an excellent resorption rate, a 3D structure similar to natural bone, biocompatibility, and customizability to multiple patient-specific geometries combined with adequate mechanical strength. Therefore, endless silk fibers were spun from native silk solution isolated from silkworm larvae and functionalized with osteoconductive bioceramic materials. In addition, transgenic silkworms were generated to functionalize silk proteins with human platelet-derived growth factor (hPDGF). Both, PDGF-silk and bioceramic modified silk were then assembled into 3D textile implants using an additive manufacturing approach. Textile implants were characterized in terms of porosity, compressive strength, and cyclic load. In addition, osteogenic differentiation of mesenchymal stem cells was evaluated. Silk fiber-based 3D textile implants showed good cytocompatibility and stem cells cultured on bioceramic material functionalized silk implants were differentiating into bone cells. Thus, functionalized 3D interconnected porous textile scaffolds were shown to be promising biomaterials for bone regeneration.
Collapse
|
24
|
Deng B, Zhu W, Duan Y, Hu Y, Chen X, Song S, Yi Z, Song Y. Exendin‑4 promotes osteogenic differentiation of adipose‑derived stem cells and facilitates bone repair. Mol Med Rep 2019; 20:4933-4942. [PMID: 31661134 PMCID: PMC6854547 DOI: 10.3892/mmr.2019.10764] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/10/2019] [Indexed: 02/07/2023] Open
Abstract
Inflammation-related bone defects pose a heavy burden on patients and orthopedic surgeons. Although stem-cell-based bone repair has developed rapidly, it is of great significance to characterize bio-active molecules that facilitate bone regeneration. It is reported that a glucagon-like peptide 1 receptor agonist, exendin-4, promoted bone regeneration mediated by the transplantation of adipose-derived stem cells in a metaphyseal defect mouse model of femur injury. However, the underlying mechanism is unclear. Bone imaging, immunohistochemistry real-time PCR and western blot analysis were used in the present study, and the results revealed that exendin-4 increased the transcription of the osteogenic differentiation-related genes and induced osteogenic differentiation in situ. Furthermore, the present data obtained from sorted adipose-derived stem cells revealed that exendin-4 promoted osteogenic differentiation and inhibited adipogenic differentiation in vitro. These findings indicated that exendin-4 facilitates osteogenic differentiation of transplanted adipose-derived stem cells for bone repair and illuminated clinical prospects of both adipose-derived stem cells and exendin-4 in stem-cell-based bone defect repair.
Collapse
Affiliation(s)
- Banglian Deng
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of Stomatology, Department of Oral Implantation, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Wenzhong Zhu
- Department of Stomatology, Shaanxi Province Geriatric Hospital, Xi'an, Shaanxi 710005, P.R. China
| | - Yansheng Duan
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of Stomatology, Department of Oral Implantation, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yuqian Hu
- Department of Stomatology, The Faculty of Medicine, Eastern University of Liaoning, Shenyang, Liaoning 110000, P.R. China
| | - Xuefeng Chen
- Xuefeng Dental Care Huaian, Huaian, Jiangsu 223000, P.R. China
| | - Shuang Song
- Health Science Center, Peking University, Beijing 100000, P.R. China
| | - Zian Yi
- Department of Stomatology, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Yingliang Song
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of Stomatology, Department of Oral Implantation, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
25
|
Donos N, Dereka X, Calciolari E. The use of bioactive factors to enhance bone regeneration: A narrative review. J Clin Periodontol 2019; 46 Suppl 21:124-161. [DOI: 10.1111/jcpe.13048] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/08/2018] [Accepted: 12/20/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Nikos Donos
- Centre for Oral Immunobiology & Regenerative Medicine & Centre for Oral Clinical Research (COCR); Institute of Dentistry, Barts & The London School of Medicine & Dentistry; Queen Mary University of London (QMUL); London UK
| | - Xanthippi Dereka
- Centre for Oral Immunobiology & Regenerative Medicine & Centre for Oral Clinical Research (COCR); Institute of Dentistry, Barts & The London School of Medicine & Dentistry; Queen Mary University of London (QMUL); London UK
- Department of Periodontology; School of Dentistry; National and Kapodistrian University of Athens; Athens Greece
| | - Elena Calciolari
- Centre for Oral Immunobiology & Regenerative Medicine & Centre for Oral Clinical Research (COCR); Institute of Dentistry, Barts & The London School of Medicine & Dentistry; Queen Mary University of London (QMUL); London UK
| |
Collapse
|
26
|
The Effects of Platelet-Derived Growth Factor-BB on Bone Marrow Stromal Cell-Mediated Vascularized Bone Regeneration. Stem Cells Int 2018; 2018:3272098. [PMID: 30515221 PMCID: PMC6234453 DOI: 10.1155/2018/3272098] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 08/09/2018] [Accepted: 08/16/2018] [Indexed: 12/20/2022] Open
Abstract
Regenerative medicine for bone tissue mainly depends on efficient recruitment of endogenous or transplanted stem cells to guide bone regeneration. Platelet-derived growth factor (PDGF) is a functional factor that has been widely used in tissue regeneration and repair. However, the short half-life of PDGF limits its efficacy, and the mechanism by which PDGF regulates stem cell-based bone regeneration still needs to be elucidated. In this study, we established genetically modified PDGF-B-overexpressing bone marrow stromal cells (BMSCs) using a lentiviral vector and then explored the mechanism by which PDGF-BB regulates BMSC-based vascularized bone regeneration. Our results demonstrated that PDGF-BB increased osteogenic differentiation but inhibited adipogenic differentiation of BMSCs via the extracellular signal-related kinase 1/2 (ERK1/2) signaling pathway. In addition, secreted PDGF-BB significantly enhanced human umbilical vein endothelial cell (HUVEC) migration and angiogenesis via the phosphatidylinositol 3 kinase (PI3K)/AKT and ERK1/2 signaling pathways. We evaluated the effect of PDGF-B-modified BMSCs on bone regeneration using a critical-sized rat calvarial defect model. Radiography, micro-CT, and histological analyses revealed that PDGF-BB overexpression improved BMSC-mediated angiogenesis and osteogenesis during bone regeneration. These results suggest that PDGF-BB facilitates BMSC-based bone regeneration by enhancing the osteogenic and angiogenic abilities of BMSCs.
Collapse
|
27
|
Du J, Xie P, Lin S, Wu Y, Zeng D, Li Y, Jiang X. Time-Phase Sequential Utilization of Adipose-Derived Mesenchymal Stem Cells on Mesoporous Bioactive Glass for Restoration of Critical Size Bone Defects. ACS APPLIED MATERIALS & INTERFACES 2018; 10:28340-28350. [PMID: 30080385 DOI: 10.1021/acsami.8b08563] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The effective transportation of oxygen, nutrients, and metabolic wastes through new blood vessel networks is key to the survival of engineered constructs in large bone defects. Adipose-derived mesenchymal stem cells (ADSCs), which are regarded as excellent candidates for both bone and blood vessel engineering, are the preferred option for the restoration of massive bone defects. Therefore, we propose to induce ADSCs into osteogenic and endothelial cells differently. A modified hierarchical mesoporous bioactive glass (MBG) scaffold with an enhanced compressive strength was constructed and prevascularized by seeding with endothelial-induced ADSCs (EI-ADSCs). The prevascularized scaffolds were combined with osteogenically induced ADSCs (OI-ADSCs) to repair critical-size bone defects. To validate the angiogenesis of the prevascularized MBG scaffolds in vivo, green fluorescent protein (GFP) was used to label EI-ADSCs. The labeled EI-ADSCs were demonstrated to survive and participate in vascularization at day 7 after subcutaneous implantation in nude mice by double immunofluorescence staining of CD31 and GFP. Regarding the restoration of critical size bone defects, early angiogenesis of rat femur plug defects was evaluated by perfusion of Microfil after 3 weeks. Compared to nonvascularized MBG carrying OI-ADSCs (MBG/OI-ADSCs) and non-cell-seeded MBG scaffolds, the prevascularized MBG carrying OI-ADSCs (Pv-MBG/OI-ADSCs) showed enhanced angiogenesis on the surface and interior. Through dynamic bone formation analysis with sequential fluorescent labeling and Van Gieson's picro-fuchsin staining, we found that the Pv-MBG/OI-ADSCs exhibited the highest mineral deposition rate after surgery, which may be contributed by rapid vascular anastomosis facilitating increased survival of the seeded OI-ADSCs and by the recruitment function for bone mesenchymal stem cells. Therefore, the strategy of time-phase sequential utilization of ADSCs on MBG scaffolds is a practical design for the repair of massive bone defects.
Collapse
Affiliation(s)
- Jiahui Du
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology , Shanghai Jiao Tong University School of Medicine , 639 Zhizaoju Road , Shanghai 200011 , China
- National Clinical Research Center for Oral Diseases , 639 Zhizaoju Road , Shanghai 200011 , China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology , 639 Zhizaoju Road , Shanghai 200011 , China
| | - Peng Xie
- The State Key Laboratory of Bioreactor Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Center for Biomedical Materials of Ministry of Education , East China University of Science and Technology , Shanghai 200237 , China
| | - Shuxian Lin
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology , Shanghai Jiao Tong University School of Medicine , 639 Zhizaoju Road , Shanghai 200011 , China
- National Clinical Research Center for Oral Diseases , 639 Zhizaoju Road , Shanghai 200011 , China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology , 639 Zhizaoju Road , Shanghai 200011 , China
| | - Yuqiong Wu
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology , Shanghai Jiao Tong University School of Medicine , 639 Zhizaoju Road , Shanghai 200011 , China
- National Clinical Research Center for Oral Diseases , 639 Zhizaoju Road , Shanghai 200011 , China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology , 639 Zhizaoju Road , Shanghai 200011 , China
| | - Deliang Zeng
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology , Shanghai Jiao Tong University School of Medicine , 639 Zhizaoju Road , Shanghai 200011 , China
- National Clinical Research Center for Oral Diseases , 639 Zhizaoju Road , Shanghai 200011 , China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology , 639 Zhizaoju Road , Shanghai 200011 , China
| | - Yulin Li
- The State Key Laboratory of Bioreactor Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Center for Biomedical Materials of Ministry of Education , East China University of Science and Technology , Shanghai 200237 , China
| | - Xinquan Jiang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology , Shanghai Jiao Tong University School of Medicine , 639 Zhizaoju Road , Shanghai 200011 , China
- National Clinical Research Center for Oral Diseases , 639 Zhizaoju Road , Shanghai 200011 , China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology , 639 Zhizaoju Road , Shanghai 200011 , China
| |
Collapse
|
28
|
Leite ÁJ, Gonçalves AI, Rodrigues MT, Gomes ME, Mano JF. Strontium-Doped Bioactive Glass Nanoparticles in Osteogenic Commitment. ACS APPLIED MATERIALS & INTERFACES 2018; 10:23311-23320. [PMID: 29906095 DOI: 10.1021/acsami.8b06154] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The present work has explored bioactive glass nanoparticles (BGNPs) and developed strontium-doped nanoparticles (BGNPsSr), envisioning orthopedic strategies compatible with vascularization. The nanoparticles were synthesized by the sol-gel method, achieving a diameter of 55 nm for BGNPs and 75 nm for BGNPsSr, and the inclusion of strontium caused no structural alteration. The nanoparticles exhibited high cytocompatibility for human umbilical vein endothelial cells (HUVECs) and SaOS-2. Additionally, the incorporation of strontium emphasized the tubule networking behavior of HUVECs. Our results demonstrate that the nanoparticle dissolution products encouraged the osteogenic differentiation of human adipose stem cells as it favored the expression of key genes and proteins associated with osteogenic lineage. This effect was markedly enhanced for BGNPsSr, which could prompt stem cell osteogenic differentiation without the typical osteogenic inducers. This study not only supports the hypothesis that BGNPs might play a significant role in osteogenic commitment but also highlights that the designed BGNPsSr is a valuable tool for stem cell "tune-up" in bone tissue engineering applications.
Collapse
Affiliation(s)
- Álvaro J Leite
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , Avepark-Parque de Ciência e Tecnologia , 4805-017 Barco , Guimarães , Portugal
- ICVS/3B's-PT Government Associate Laboratory , 4710-057 Braga/Guimarães , Portugal
| | - Ana I Gonçalves
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , Avepark-Parque de Ciência e Tecnologia , 4805-017 Barco , Guimarães , Portugal
- ICVS/3B's-PT Government Associate Laboratory , 4710-057 Braga/Guimarães , Portugal
| | - Márcia T Rodrigues
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , Avepark-Parque de Ciência e Tecnologia , 4805-017 Barco , Guimarães , Portugal
- ICVS/3B's-PT Government Associate Laboratory , 4710-057 Braga/Guimarães , Portugal
| | - Manuela E Gomes
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , Avepark-Parque de Ciência e Tecnologia , 4805-017 Barco , Guimarães , Portugal
- ICVS/3B's-PT Government Associate Laboratory , 4710-057 Braga/Guimarães , Portugal
- The Discoveries Centre for Regenerative and Precision Medicine , Headquarters at University of Minho , Avepark , 4805-017 Barco , Guimarães , Portugal
| | - João F Mano
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , Avepark-Parque de Ciência e Tecnologia , 4805-017 Barco , Guimarães , Portugal
- ICVS/3B's-PT Government Associate Laboratory , 4710-057 Braga/Guimarães , Portugal
| |
Collapse
|
29
|
Tsang EJ, Wu B, Zuk P. MAPK signaling has stage-dependent osteogenic effects on human adipose-derived stem cells in vitro. Connect Tissue Res 2018; 59:129-146. [PMID: 28398098 PMCID: PMC6200338 DOI: 10.1080/03008207.2017.1313248] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 03/14/2017] [Indexed: 02/03/2023]
Abstract
OVERVIEW The use of pro-osteogenic growth factors, such as BMP2, in human adipose-derived stem cell (ASC) osteogenesis is well described. Because these growth factors work via signal transduction pathways, such as the mitogen-activated protein kinase (MAPK) cascade, a study of the relationship between MAPK signaling and ASC osteogenesis was conducted. MATERIALS AND METHODS ERK, JNK, and p38MAPK activation were measured in ASCs osteo-induced using either dexamethasone or vitamin D3 and correlated with mineralization. Activation and mineralization were also measured without dexamethasone or using the glucocorticoid, cortisone. The expression of the MAPK phosphatase, MKP1, and its relationship to mineralization was also assessed. The effect of decreasing MAPK activation on mineralization through the use of exogenous inhibitors was examined along with siRNA-knockdown and adenoviral overexpression of ERK1/2. Finally, the effect of ERK1/2 overexpression on ASCs induced on PLGA scaffolds was assessed. RESULTS ASC mineralization in dexamethasone or vitamin D3-induced ASCs correlated with both increased ERK1/2 and JNK1/2 activation. ASCs induced without dexamethasone also mineralized, with JNK1/2 signaling possibly mediating this event. No link between cortisone induction and MAPK signaling could be ascertained. ASCs treated with ERK, JNK, or p38MAPK inhibitors showed decreased osteogenic gene expression and diminished mineralization. Mineralization levels were also affected by viruses designed to inhibit or augment ERK1/2 expression and activity. Finally, ASC mineralization appeared to be a balance between the MAPK kinase activity and MKP1. CONCLUSIONS It is likely that MAPK signaling plays a significant role in ASC osteogenesis, affecting differentiation in kinase- and stage-specific manners.
Collapse
Affiliation(s)
- Eric J. Tsang
- Regenerative Bioengineering and Repair Laboratory, Department of Surgery, David Geffen School of Medicine at UCLA. University of California, Los Angeles, CA, USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences. University of California, Los Angeles, CA, USA
| | - Benjamin Wu
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences. University of California, Los Angeles, CA, USA
| | - Patricia Zuk
- Regenerative Bioengineering and Repair Laboratory, Department of Surgery, David Geffen School of Medicine at UCLA. University of California, Los Angeles, CA, USA
| |
Collapse
|
30
|
Jing X, Yin W, Tian H, Chen M, Yao X, Zhu W, Guo F, Ye Y. Icariin doped bioactive glasses seeded with rat adipose-derived stem cells to promote bone repair via enhanced osteogenic and angiogenic activities. Life Sci 2018; 202:52-60. [PMID: 29471105 DOI: 10.1016/j.lfs.2018.02.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 02/01/2018] [Accepted: 02/17/2018] [Indexed: 12/29/2022]
Abstract
AIMS Cell communication between mesenchymal stem cells and blood vessel cells are crucial for bone repair. We have previously shown that the phyto-molecule icariin significantly promoted osteogenic differentiation of rat adipose-derived stem cells (ASCs). In the present study, we aimed to investigate the relationship between icariin induced osteogenic differentiation of ASCs and angiogenesis of rat endothelial progenitor cells (EPCs). Besides, we used icariin doped 45S5 Bioglass seeded with ASCs to promote bone healing in rat calvarial bone defect models. MAIN METHODS The conditioned medium from undifferentiated ASCs (ASCs-CM) and icariin induced ASCs (Icariin-ASCs-CM) was obtained and the vascular endothelial growth factor (VEGF) protein secretion level was measured. The angiogenic capacity and molecular mechanism of ASC-CM and Icariin-ASCs-CM on rat EPCs was analyzed. Rat calvarial bone defect models were established and treated with scaffolds implantation. Micro-CT imaging, histological and immunohistological staining were performed on the isolated specimens at 12 weeks post-surgery. KEY FINDINGS VEGF protein expression was significantly increased after icariin treatment with the highest expression in the 10-7 M icariin group. Icariin-ASCs-CM obviously increased the angiogenesis of rat EPCs and this capacity was inhibited by a VEGF/VEGF receptor-specific binding inhibitor bevacizumab. Results of the in vivo investigations showed that all scaffolds promoted bone healing compared to the Control group. Icariin significantly improved the healing capacity of 45S5 Bioglass seeded with ASCs. SIGNIFICANCE Implantation of Icariin/45S5 Bioglass seeded with rat ASCs could obviously promote both osteogenesis and angiogenesis and therefore represents an ideal candidate bone substitutes for bone repair and regeneration.
Collapse
Affiliation(s)
- Xingzhi Jing
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Weifeng Yin
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Hongtao Tian
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Mengcun Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xudong Yao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Wentao Zhu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Fengjing Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| | - Yaping Ye
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
31
|
Abstract
The craniofacial complex is composed of fundamental components such as blood vessels and nerves, and also a variety of specialized tissues such as craniofacial bones, cartilages, muscles, ligaments, and the highly specialized and unique organs, the teeth. Together, these structures provide many functions including speech, mastication, and aesthetics of the craniofacial complex. Craniofacial defects not only influence the structure and function of the jaws and face, but may also result in deleterious psychosocial issues, emphasizing the need for rapid and effective, precise, and aesthetic reconstruction of craniofacial tissues. In a broad sense, craniofacial tissue reconstructions share many of the same issues as noncraniofacial tissue reconstructions. Therefore, many concepts and therapies for general tissue engineering can and have been used for craniofacial tissue regeneration. Still, repair of craniofacial defects presents unique challenges, mainly because of their complex and unique 3D geometry.
Collapse
Affiliation(s)
- Weibo Zhang
- Department of Orthodontics, School of Medicine, School of Engineering, Tufts University, Boston, Massachusetts 02111
| | - Pamela Crotty Yelick
- Department of Orthodontics, School of Medicine, School of Engineering, Tufts University, Boston, Massachusetts 02111
| |
Collapse
|
32
|
Mishra R, Sefcik RS, Bishop TJ, Montelone SM, Crouser N, Welter JF, Caplan AI, Dean D. Growth Factor Dose Tuning for Bone Progenitor Cell Proliferation and Differentiation on Resorbable Poly(propylene fumarate) Scaffolds. Tissue Eng Part C Methods 2017; 22:904-13. [PMID: 27558310 DOI: 10.1089/ten.tec.2016.0094] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
One approach to the development of an artificial graft material could rely on uniform coverage of a resorbable biomaterial with bone extracellular matrix (ECM). To achieve this on the surface of poly(propylene fumarate) (PPF) scaffolds, we selected a growth factor regime of basic fibroblast growth factor (FGF-2) (5 ng/mL), platelet-derived growth factor (PDGF-BB) (40 ng/mL), and epidermal growth factor (EGF) (20 ng/mL) to stimulate proliferation of bone marrow-derived human mesenchymal stem cells (BM-hMSCs). Bone morphogenetic protein (BMP) 4 (50 ng/mL), 6 (50 ng/mL), and 7 (27 ng/mL) in the presence of the following osteogenic substances: dexamethasone (10(-7) M), β-glycerophosphate (10 mM), and ascorbic acid (50 μg/mL) were chosen to induce differentiation of BM-hMSCs into ECM-secreting osteoblasts. These growth factors were also studied at 10× concentration to determine dose effect. Proliferation was analyzed by MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay, scanning electron microscopy (SEM), and toluidine blue staining, whereas differentiation was analyzed through alizarin red S staining and assay, alkaline phosphatase (ALP) staining and assay, and SEM. The proliferation study suggests that a combination of EGF, PDGF-BB, and FGF-2 growth factors at optimal concentration over a period of 1 week exhibits significantly (p = 0.001) higher number of cells (116,024 ± 5165) than these cytokines without EGF (91,706 ± 11,965). Increasing the dosage does not show any significant effect. The BM-hMSC differentiation study results show that ALP enzyme production and mineral deposition increase from day 14 to day 21 in all groups containing BMPs and osteogenic medium. However, mineralization is significantly higher in the BMP-7 group. Furthermore, the feasibility of translating the results from two dimensional thin films to three dimensional-printed PPF scaffolds was determined through uniform initial seeding and spreading of BM-hMSCs. Therefore, we have determined the optimum dose of growth factors for proliferation and differentiation of BM-hMSCs on the surface of PPF scaffolds, which can be used to produce ECM-coated implants for the treatment of bone defects.
Collapse
Affiliation(s)
- Ruchi Mishra
- 1 Department of Plastic Surgery, The Ohio State University , Columbus, Ohio
| | - Ryan S Sefcik
- 1 Department of Plastic Surgery, The Ohio State University , Columbus, Ohio
| | - Tyler J Bishop
- 1 Department of Plastic Surgery, The Ohio State University , Columbus, Ohio
| | | | - Nisha Crouser
- 1 Department of Plastic Surgery, The Ohio State University , Columbus, Ohio
| | - Jean F Welter
- 2 Skeletal Research Center, Department of Biology, Case Western Reserve University , Cleveland, Ohio
| | - Arnold I Caplan
- 2 Skeletal Research Center, Department of Biology, Case Western Reserve University , Cleveland, Ohio
| | - David Dean
- 1 Department of Plastic Surgery, The Ohio State University , Columbus, Ohio
| |
Collapse
|
33
|
Zhang C, Yu L, Liu S, Wang Y. Human amnion-derived mesenchymal stem cells promote osteogenic and angiogenic differentiation of human adipose-derived stem cells. PLoS One 2017; 12:e0186253. [PMID: 29020045 PMCID: PMC5636128 DOI: 10.1371/journal.pone.0186253] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/27/2017] [Indexed: 12/22/2022] Open
Abstract
Tissue engineering using suitable mesenchymal stem cells (MSCs) shows great potential to regenerate bone defects. Our previous studies have indicated that human amnion-derived mesenchymal stem cells (HAMSCs) could promote the osteogenic differentiation of human bone marrow mesenchymal stem cells (HBMSCs). Human adipose-derived stem cells (HASCs), obtained from adipose tissue in abundance, are capable of multi-lineage differentiation. In this study, the effects of HAMSCs on osteogenic and angiogenic differentiation of HASCs were systematically investigated. Proliferation levels were measured by flow cytometry. Osteoblastic differentiation and mineralization were investigated using chromogenic alkaline phosphatase activity (ALP) activity substrate assays, Alizarin red S staining, real-time polymerase chain reaction (real-time PCR) analysis of osteogenic marker expression, and Western blotting. We found that HAMSCs increased the proliferation and osteoblastic differentiation of HASCs. Moreover, enzyme-linked immunosorbent assay (ELISA) and human umbilical vein endothelial cells (HUVECs) tube formation suggested HAMSCs enhanced angiogenic potential of HASCs via secretion of increased vascular endothelial growth factor (VEGF). Thus, we conclude that HAMSC might be a valuable therapeutic approach to promote HASCs-involved bone regeneration.
Collapse
Affiliation(s)
- Chunli Zhang
- Department of Clinical Research, Friendship Plastic Surgery Hospital, Nanjing Medical University, Nanjing, Jiangsu, The People’s Republic of China
| | - Lidong Yu
- Department of Plastic Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, The People’s Republic of China
| | - Songjian Liu
- Department of Plastic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, The People’s Republic of China
| | - Yuli Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, The People’s Republic of China
- * E-mail:
| |
Collapse
|
34
|
Wang G, Roohani-Esfahani SI, Zhang W, Lv K, Yang G, Ding X, Zou D, Cui D, Zreiqat H, Jiang X. Effects of Sr-HT-Gahnite on osteogenesis and angiogenesis by adipose derived stem cells for critical-sized calvarial defect repair. Sci Rep 2017; 7:41135. [PMID: 28106165 PMCID: PMC5247715 DOI: 10.1038/srep41135] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 12/15/2016] [Indexed: 01/08/2023] Open
Abstract
Tissue engineering strategies to construct vascularized bone grafts are now attracting much attention. Strontium-hardystonite-Gahnite (Sr-HT-Gahnite) is a strong, highly porous, and biocompatible calcium silicate based bio-ceramic that contains strontium and zinc ions. Adipose derived stem cells (ASCs) have been demonstrated to have the ability in promoting osteogenesis and angiogenesis. In this study, the effects of Sr-HT-Gahnite on cell morphology, cell proliferation, and osteogenic differentiation of ASCs were systematically investigated. The cell proliferation, migration and angiogenic differentiation of human umbilical vein endothelial cell (HUVECs) were studied. Beta-tricalcium phosphate/hydroxyapatite (TCP/HA) bioceramic scaffolds were set as the control biomaterial. Both bio-ceramics exhibited no adverse influence on cell viability. The Sr-HT-Gahnite scaffolds promoted cell attachment and alkaline phosphatase (ALP) activity of ASCs. The Sr-HT-Gahnite dissolution products enhanced ALP activity, matrix mineralization, and angiogenic differentiation of ASCs. They could also improve cell proliferation, migration, and angiogenic differentiation of HUVECs. Levels of in vivo bone formation with Sr-HT Gahnite were significantly higher than that for TCP/HA. The combination of Sr-HT-Gahnite and ASCs promoted both osteogenesis and angiogenesis in vivo study, compared to Sr-HT-Gahnite and TCP/HA bio-ceramics when administered alone, suggesting Sr-HT-Gahnite can act as a carrier for ASCs for construction of vascularized tissue-engineered bone.
Collapse
Affiliation(s)
- Guifang Wang
- Department of Prosthodontics, Ninth People's Hospital affiliated to Shanghai Jiao Tong University, School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
- Oral Bioengineering Lab, Shanghai Research Institute of Stomatology, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai 200011, China
| | - Seyed-Iman Roohani-Esfahani
- Biomaterials and Tissue Engineering Research Unit, School of AMME, The University of Sydney, Sydney, NSW 2006, Australia
| | - Wenjie Zhang
- Department of Prosthodontics, Ninth People's Hospital affiliated to Shanghai Jiao Tong University, School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
- Oral Bioengineering Lab, Shanghai Research Institute of Stomatology, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai 200011, China
| | - Kaige Lv
- Department of Prosthodontics, Ninth People's Hospital affiliated to Shanghai Jiao Tong University, School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
- Oral Bioengineering Lab, Shanghai Research Institute of Stomatology, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai 200011, China
| | - Guangzheng Yang
- Oral Bioengineering Lab, Shanghai Research Institute of Stomatology, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai 200011, China
| | - Xun Ding
- Department of Prosthodontics, Ninth People's Hospital affiliated to Shanghai Jiao Tong University, School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
- Oral Bioengineering Lab, Shanghai Research Institute of Stomatology, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai 200011, China
| | - Derong Zou
- Department of Stomatology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Daxiang Cui
- Institute of Nano Biomedicine and Engineering, Key Laboratory of Thin Film and Microfabrication Technology of Ministry of Education, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Hala Zreiqat
- Biomaterials and Tissue Engineering Research Unit, School of AMME, The University of Sydney, Sydney, NSW 2006, Australia
| | - Xinquan Jiang
- Department of Prosthodontics, Ninth People's Hospital affiliated to Shanghai Jiao Tong University, School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
- Oral Bioengineering Lab, Shanghai Research Institute of Stomatology, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai 200011, China
| |
Collapse
|
35
|
Bone morphogenetic protein 2 promotes osteogenesis of bone marrow stromal cells in type 2 diabetic rats via the Wnt signaling pathway. Int J Biochem Cell Biol 2016; 80:143-153. [DOI: 10.1016/j.biocel.2016.09.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/25/2016] [Accepted: 09/29/2016] [Indexed: 01/04/2023]
|
36
|
Biomimetic whitlockite inorganic nanoparticles-mediated in situ remodeling and rapid bone regeneration. Biomaterials 2016; 112:31-43. [PMID: 27744219 DOI: 10.1016/j.biomaterials.2016.10.009] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/27/2016] [Accepted: 10/07/2016] [Indexed: 11/20/2022]
Abstract
Bone remodeling process relies on complex signaling pathway between osteoblasts and osteoclasts and control mechanisms to achieve homeostasis of their growth and differentiation. Despite previous achievements in understanding complicated signaling pathways between cells and bone extracellular matrices during bone remodeling process, a role of local ionic concentration remains to be elucidated. Here, we demonstrate that synthetic whitlockite (WH: Ca18Mg2(HPO4)2(PO4)12) nanoparticles can recapitulate early-stage of bone regeneration through stimulating osteogenic differentiation, prohibiting osteoclastic activity, and transforming into mechanically enhanced hydroxyapatite (HAP)-neo bone tissues by continuous supply of PO43- and Mg2+ under physiological conditions. In addition, based on their structural analysis, the dynamic phase transformation from WH into HAP contributed as a key factor for rapid bone regeneration with denser hierarchical neo-bone structure. Our findings suggest a groundbreaking concept of 'living bone minerals' that actively communicate with the surrounding system to induce self-healing, while previous notions about bone minerals have been limited to passive products of cellular mineralization.
Collapse
|
37
|
Chen WH, Lin CM, Huang CF, Hsu WC, Lee CH, Ou KL, Dubey NK, Deng WP. Functional Recovery in Osteoarthritic Chondrocytes Through Hyaluronic Acid and Platelet-Rich Plasma-Inhibited Infrapatellar Fat Pad Adipocytes. Am J Sports Med 2016; 44:2696-2705. [PMID: 27400716 DOI: 10.1177/0363546516651822] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Recent studies have shown evidence that higher adiposity in the infrapatellar fat pad (IFP) induces inflammatory phenotypes in the knee joint and thereby contributes to the development and progression of osteoarthritis (OA). In particular, IFP adipocyte-derived inflammatory cytokines participate in pathological events. Our previous research has already addressed the therapeutic efficacy of hyaluronic acid and platelet-rich plasma (HA+PRP), including the promotion of cartilage regeneration and the inhibition of inflammation. The current study aimed to explore the remedial action of coadministered HA+PRP in OA recovery via IFP adipocyte inhibition. HYPOTHESIS HA+PRP repairs OA articular cartilage by inhibiting the release of adipokines from IFP adipocytes. STUDY DESIGN Controlled laboratory study. METHODS IFP adipocytes and articular chondrocytes were obtained from 10 patients with OA, and the effects of releasates containing cytokines and adipokines in IFP adipocyte-derived conditioned medium (IACM) on articular chondrocytes and IFP adipocytes themselves were evaluated. The therapeutic efficacy of exogenous HA+PRP was determined through its administration to cocultured IFP adipocytes and articular chondrocytes and further demonstrated in a 3-dimensional (3D) arthritic neocartilage model. RESULTS The IACM and IFP adipocyte-induced microenvironment could induce dedifferentiated and inflammatory phenotypes in articular chondrocytes. HA+PRP decreased the inflammatory potential of IFP adipocytes through the profound inhibition of cytokines and adipokines. The IACM-mediated and -reduced cartilaginous extracellular matrix could also be recovered through HA+PRP in the 3D arthritic neocartilage model. CONCLUSION IFP adipocyte-derived releasates mediated inflammatory response dedifferentiation in chondrocytes, which was recovered through HA+PRP administration. CLINICAL RELEVANCE Our findings demonstrated that HA+PRP effectively diminished IFP adipocyte-promoted inflammation in articular chondrocytes, indicating that the IFP could be a potential therapeutic target for OA therapy.
Collapse
Affiliation(s)
- Wei-Hong Chen
- Stem Cell Research Center, Taipei Medical University, Taipei, Taiwan Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Chien-Min Lin
- Department of Neurosurgery, Taipei Medical University-Shuang Ho Hospital, Ministry of Health and Welfare, Taipei, Taiwan
| | - Chiung-Fang Huang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wei-Che Hsu
- Stem Cell Research Center, Taipei Medical University, Taipei, Taiwan Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Chian-Her Lee
- Department of Orthopaedics and Traumatology, Orthopaedic Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Keng-Liang Ou
- Research Center for Biomedical Implants and Microsurgery Devices, Taipei, Taiwan Research Center for Biomedical Devices and Prototype Production, Taipei, Taiwan
| | - Navneet Kumar Dubey
- Stem Cell Research Center, Taipei Medical University, Taipei, Taiwan Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Win-Ping Deng
- Stem Cell Research Center, Taipei Medical University, Taipei, Taiwan Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan College of Medicine, Fu-Jen Catholic University, Taipei, Taiwan
| |
Collapse
|
38
|
Wang G, Li J, Lv K, Zhang W, Ding X, Yang G, Liu X, Jiang X. Surface thermal oxidation on titanium implants to enhance osteogenic activity and in vivo osseointegration. Sci Rep 2016; 6:31769. [PMID: 27546196 PMCID: PMC4992888 DOI: 10.1038/srep31769] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 07/27/2016] [Indexed: 02/06/2023] Open
Abstract
Thermal oxidation, which serves as a low-cost, effective and relatively simple/facile method, was used to modify a micro-structured titanium surface in ambient atmosphere at 450 °C for different time periods to improve in vitro and in vivo bioactivity. The surface morphology, crystallinity of the surface layers, chemical composition and chemical states were evaluated by field-emission scanning electron microscopy (FESEM), X-ray diffraction (XRD), and X-ray photoelectron spectroscopy (XPS). Cell behaviours including cell adhesion, attachment, proliferation, and osteogenic differentiation were observed in vitro study. The ability of the titanium surface to promote osseointegration was evaluated in an in vivo animal model. Surface thermal oxidation on titanium implants maintained the microstructure and, thus, both slightly changed the nanoscale structure of titanium and enhanced the crystallinity of the titanium surface layer. Cells cultured on the three oxidized titanium surfaces grew well and exhibited better osteogenic activity than did the control samples. The in vivo bone-implant contact also showed enhanced osseointegration after several hours of oxidization. This heat-treated titanium enhanced the osteogenic differentiation activity of rBMMSCs and improved osseointegration in vivo, suggesting that surface thermal oxidation could potentially be used in clinical applications to improve bone-implant integration.
Collapse
Affiliation(s)
- Guifang Wang
- Department of Prosthodontics, Ninth People's Hospital affiliated to Shanghai Jiao Tong University, School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China.,Oral Bioengineering Lab, Shanghai Research Institute of Stomatology, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai 200011, China
| | - Jinhua Li
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kaige Lv
- Department of Prosthodontics, Ninth People's Hospital affiliated to Shanghai Jiao Tong University, School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China.,Oral Bioengineering Lab, Shanghai Research Institute of Stomatology, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai 200011, China
| | - Wenjie Zhang
- Department of Prosthodontics, Ninth People's Hospital affiliated to Shanghai Jiao Tong University, School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China.,Oral Bioengineering Lab, Shanghai Research Institute of Stomatology, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai 200011, China
| | - Xun Ding
- Department of Prosthodontics, Ninth People's Hospital affiliated to Shanghai Jiao Tong University, School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China.,Oral Bioengineering Lab, Shanghai Research Institute of Stomatology, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai 200011, China
| | - Guangzheng Yang
- Oral Bioengineering Lab, Shanghai Research Institute of Stomatology, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai 200011, China
| | - Xuanyong Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Xinquan Jiang
- Department of Prosthodontics, Ninth People's Hospital affiliated to Shanghai Jiao Tong University, School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China.,Oral Bioengineering Lab, Shanghai Research Institute of Stomatology, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai 200011, China
| |
Collapse
|
39
|
Fan D, Liu S, Jiang S, Li Z, Mo X, Ruan H, Zou GM, Fan C. The use of SHP-2 gene transduced bone marrow mesenchymal stem cells to promote osteogenic differentiation and bone defect repair in rat. J Biomed Mater Res A 2016; 104:1871-81. [PMID: 26999642 DOI: 10.1002/jbm.a.35718] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 02/23/2016] [Accepted: 03/10/2016] [Indexed: 12/30/2022]
Abstract
Bone tissue engineering is a promising approach for bone regeneration, in which growth factors play an important role. The tyrosine phosphatase Src-homology region 2-containing protein tyrosine phosphatase 2 (SHP2), encoded by the PTPN11 gene, is essential for the differentiation, proliferation and metabolism of osteoblasts. However, SHP-2 has never been systematically studied for its effect in osteogenesis. We predicted that overexpression of SHP-2 could promote bone marrow-derived mesenchymal stem cell (BMSC)osteogenic differentiation and SHP-2 transduced BMSCs could enhance new bone formation, determined using the following study groups: (1) BMSCs transduced with SHP-2 and induced with osteoblast-inducing liquid (BMSCs/SHP-2/OL); (2) BMSCs transduced with SHP-2 (BMSCs/-SHP-2); (3) BMSCs induced with osteoblast-inducing liquid (BMSCs/OL) and (4) pure BMSCs. Cells were assessed for osteogenic differentiation by quantitative real-time polymerase chain reaction analysis, western blot analysis, alkaline phosphatase activity and alizarin red S staining. For in vivo assessment, cells were combined with beta-tricalcium phosphate scaffolds and transplanted into rat calvarial defects for 8 weeks. Following euthanasia, skull samples were explanted for osteogenic evaluation, including micro-computed tomography measurement, histology and immunohistochemistry staining. SHP-2 and upregulation of its gene promoted BMSC osteogenic differentiation and therefore represents a potential new therapeutic approach to bone repair. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 1871-1881, 2016.
Collapse
Affiliation(s)
- Dapeng Fan
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, People's Republic of China
| | - Shen Liu
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, People's Republic of China
| | - Shichao Jiang
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong University, No.324 Jingwu Road, Jinan, 250021, Shandong, People's Republic of China
| | - Zhiwei Li
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, People's Republic of China
| | - Xiumei Mo
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China
| | - Hongjiang Ruan
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, People's Republic of China
| | - Gang-Ming Zou
- Hawaii Gangze Inc, 421 Nahua Street, Suite 146, Honolulu, Hawaii, 96815
| | - Cunyi Fan
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, People's Republic of China
| |
Collapse
|
40
|
Zhang X, Zeng D, Li N, Jiang X, Liu C, Li Y. Large-pore mesoporous Ca–Si-based bioceramics with high in vitro bioactivity and protein adsorption capability for bone tissue regeneration. J Mater Chem B 2016; 4:3916-3924. [DOI: 10.1039/c6tb00454g] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
A new type of large pore mesoporous Ca–Si-based bioceramics demonstrates high in vitro bioactivity and protein adsorption capability.
Collapse
Affiliation(s)
- Xingdi Zhang
- Lab of Low-Dimensional Materials Chemistry
- Key Laboratory for Ultrafine Materials of Ministry of Education
- School of Materials Science and Engineering
- East China University of Science and Technology
- Shanghai 200237
| | - Deliang Zeng
- Department of Prosthodontics
- Ninth People's Hospital affiliated to Shanghai Jiao Tong University
- School of Medicine
- Shanghai 200011
- China
| | - Nan Li
- Lab of Low-Dimensional Materials Chemistry
- Key Laboratory for Ultrafine Materials of Ministry of Education
- School of Materials Science and Engineering
- East China University of Science and Technology
- Shanghai 200237
| | - Xinquan Jiang
- Department of Prosthodontics
- Ninth People's Hospital affiliated to Shanghai Jiao Tong University
- School of Medicine
- Shanghai 200011
- China
| | - Changsheng Liu
- Engineering Research Center for Biomedical Materials of Ministry of Education
- East China University of Science and Technology
- Shanghai 200237
- China
| | - Yongsheng Li
- Lab of Low-Dimensional Materials Chemistry
- Key Laboratory for Ultrafine Materials of Ministry of Education
- School of Materials Science and Engineering
- East China University of Science and Technology
- Shanghai 200237
| |
Collapse
|
41
|
Silk fibroin/collagen and silk fibroin/chitosan blended three-dimensional scaffolds for tissue engineering. EUROPEAN JOURNAL OF ORTHOPAEDIC SURGERY AND TRAUMATOLOGY 2014; 25:243-9. [DOI: 10.1007/s00590-014-1515-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 07/16/2014] [Indexed: 11/25/2022]
|