1
|
Chen Z, Gan L, Chen X, Zheng J, Shi S, Wu L, Cao Y. LncRNA HOTAIRM1 promotes dental follicle stem cell-mediated bone regeneration by regulating HIF-1α/KDM6/EZH2/H3K27me3 axis. J Cell Physiol 2023. [PMID: 37120836 DOI: 10.1002/jcp.31028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 03/21/2023] [Accepted: 04/12/2023] [Indexed: 05/02/2023]
Abstract
Large bone defect reconstruction undergoes hypoxia and remains a major practical challenge. Bone tissue engineering with a more promising stem cell source facilitates the development of better therapeutic outcomes. Human dental follicle stem cells (hDFSCs) with superior multipotency, osteogenic capacity, and accessibility have been proven a promising cell source for bone regeneration. We previously identified a novel long noncoding RNA (lncRNA), HOTAIRM1, to be highly expressed in hDFSCs. Here we found that HOTAIRM1 overexpressed hDFSCs promoted bone regeneration in rat critical-size calvarial defect model. Mechanically, HOTAIRM1 was induced in hDFSCs under hypoxic conditions and activated HIF-1α. RNA-sequencing analysis indicated that HOTAIRM1 upregulated oxygen-sensing histone demethylases KDM6A/B and suppressed methyltransferase EZH2 via targeting HIF-1α. The osteogenic differentiation of hDFSCs was accompanied with demethylation of H3K27, and HOTAIRM1 overexpression decreased the distribution of H3K27me3 in osteogenic genes, including ALP, M-CSF, Wnt-3a, Wnt-5a, Wnt-7a, and β-catenin, thus promoted their transcription. Our study provided evidence that HOTAIRM1 upregulated KDM6A/B and inhibited EZH2 in a HIF-1α dependent manner to enhance the osteogenesis of hDFSCs. HOTAIRM1-mediated hDFSCs may serve as a promising therapeutic approach to promote bone regeneration in clinical practice.
Collapse
Affiliation(s)
- Zhengyuan Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liyi Gan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xin Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jinxuan Zheng
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Songtao Shi
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liping Wu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yang Cao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Costimulatory Effect of Rough Calcium Phosphate Coating and Blood Mononuclear Cells on Adipose-Derived Mesenchymal Stem Cells In Vitro as a Model of In Vivo Tissue Repair. MATERIALS 2020; 13:ma13194398. [PMID: 33023124 PMCID: PMC7579197 DOI: 10.3390/ma13194398] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/26/2020] [Accepted: 09/29/2020] [Indexed: 02/07/2023]
Abstract
Calcium phosphate (CaP) materials do not always induce ectopic vascularization and bone formation; the reasons remain unclear, and there are active discussions of potential roles for post-implantation hematoma, circulating immune and stem cells, and pericytes, but studies on adipose-derived stem cells (AMSCs) in this context are lacking. The rough (average surface roughness Ra = 2-5 µm) scaffold-like CaP coating deposited on pure titanium plates by the microarc oxidation method was used to investigate its subcutaneous vascularization in CBA/CaLac mice and in vitro effect on cellular and molecular crosstalk between human blood mononuclear cells (hBMNCs) and AMSCs (hAMSCs). Postoperative hematoma development on the CaP surface lasting 1-3 weeks may play a key role in the microvessel elongation and invasion into the CaP relief at the end of the 3rd week of injury and BMNC migration required for enhanced wound healing in mice. Satisfactory osteogenic and chondrogenic differentiation but poor adipogenic differentiation of hAMSCs on the rough CaP surface were detected in vitro by differential cell staining. The fractions of CD73+ (62%), CD90+ (0.24%), and CD105+ (0.41%) BMNCs may be a source of autologous circulating stem/progenitor cells for the subcutis reparation, but allogenic hBMNC participation is mainly related to the effects of CD4+ T cells co-stimulated with CaP coating on the in vitro recruitment of hAMSCs, their secretion of angiogenic and osteomodulatory molecules, and the increase in osteogenic features within the period of in vivo vascularization. Cellular and molecular crosstalk between BMNCs and AMSCs is a model of effective subcutis repair. Rough CaP surface enhanced angio- and osteogenic signaling between cells. We believe that preconditioning and/or co-transplantation of hAMSCs with hBMNCs may broaden their potential in applications related to post-implantation tissue repair and bone bioengineering caused by microarc CaP coating.
Collapse
|
3
|
Hivernaud V, Grimaud F, Guicheux J, Portron S, Pace R, Pilet P, Sourice S, Wuillem S, Bertin H, Roche R, Espitalier F, Weiss P, Corre P. Comparing “intra operative” tissue engineering strategies for the repair of craniofacial bone defects. JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2019; 120:432-442. [DOI: 10.1016/j.jormas.2019.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/23/2018] [Accepted: 01/03/2019] [Indexed: 01/02/2023]
|
4
|
Hayashi K, Kishida R, Tsuchiya A, Ishikawa K. Honeycomb blocks composed of carbonate apatite, β-tricalcium phosphate, and hydroxyapatite for bone regeneration: effects of composition on biological responses. Mater Today Bio 2019; 4:100031. [PMID: 32159156 PMCID: PMC7061555 DOI: 10.1016/j.mtbio.2019.100031] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/02/2019] [Accepted: 09/14/2019] [Indexed: 10/26/2022] Open
Abstract
Synthetic scaffolds exhibiting bone repair ability equal to that of autogenous bone are required in the fields of orthopedics and dentistry. A suitable synthetic bone graft substitute should induce osteogenic differentiation of mesenchymal stem cells, osteogenesis, and angiogenesis. In this study, three types of honeycomb blocks (HCBs), composed of hydroxyapatite (HAp), β-tricalcium phosphate (TCP), and carbonate apatite (CO3Ap), were fabricated, and the effects of HCB composition on bone formation and maturation were investigated. The HC structure was selected to promote cell penetration and tissue ingrowth. HAp and β-TCP HCBs were fabricated by extrusion molding followed by sintering. The CO3Ap HCBs were fabricated by extrusion molding followed by sintering and dissolution-precipitation reactions. These HCBs had similar macroporous structures: all harbored uniformly distributed macropores (∼160 μm) that were regularly arrayed and penetrated the blocks unidirectionally. Moreover, the volumes of macropores were nearly equal (∼0.15 cm3/g). The compressive strengths of CO3Ap, HAp, and β-TCP HCBs were 22.8 ± 3.5, 34.2 ± 3.3, and 24.4 ± 2.4 MPa, respectively. Owing to the honeycomb-type macroporous structure, the compressive strengths of these HCBs were higher than those of commercial scaffolds with intricate three-dimensional or unidirectional macroporous structure. Notably, bone maturation was markedly faster in CO3Ap HCB grafting than in β-TCP and HAp HCB grafting, and the mature bone area percentages for CO3Ap HCBs at postsurgery weeks 4 and 12 were 14.3- and 4.3-fold higher and 7.5- and 1.4-fold higher than those for HAp and β-TCP HCBs, respectively. The differences in bone maturation and formation were probably caused by the disparity in concentrations of calcium ions surrounding the HCBs, which were dictated by the inherent material resorption behavior and mechanism; generally, CO3Ap is resorbed only by osteoclastic resorption, HAp is not resorbed, and β-TCP is rapidly dissolved even in the absence of osteoclasts. Besides the composition, the microporous structure of HC struts, inevitably generated during the formation of HCBs of various compositions, may contribute to the differences in bone maturation and formation.
Collapse
Key Words
- Bone regeneration
- Bone-graft substitute
- Fourier transform infrared, FTIR
- Osteogenesis
- Osteogenic differentiation
- Scaffold
- blood vessels, BV
- calcium phosphate, CaP
- carbonate apatite, CO3Ap
- hematoxylin-eosin, HE
- honeycomb blocks, HCBs
- honeycomb, HC
- hydroxyapatite, HAp
- mesenchymal stem cells, MSCs
- osteoblast, OB
- osteoclasts, OCs
- postoperative week, POW
- tricalcium phosphate, TCP
Collapse
Affiliation(s)
- K. Hayashi
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | | | | | | |
Collapse
|
5
|
Sato T, Anada T, Hamai R, Shiwaku Y, Tsuchiya K, Sakai S, Baba K, Sasaki K, Suzuki O. Culture of hybrid spheroids composed of calcium phosphate materials and mesenchymal stem cells on an oxygen-permeable culture device to predict in vivo bone forming capability. Acta Biomater 2019; 88:477-490. [PMID: 30844570 DOI: 10.1016/j.actbio.2019.03.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/28/2019] [Accepted: 03/03/2019] [Indexed: 12/30/2022]
Abstract
Three-dimensional (3-D) cell culture can better mimic physiological conditions in which cells interact with adjacent cells and the extracellular matrix than monolayer culture. We have developed a 3-D cell culture device, the Oxy chip, which can be used to generate and supply oxygen to cell spheroids to prevent hypoxia. Here, we used the Oxy chip to generate hybrid spheroids comprising calcium phosphate (CaP) particles (hydroxyapatite (HA), β-tricalcium phosphate (β-TCP) or octacalcium phosphate (OCP)) and mesenchymal stem cells (MSCs, C3H10T1/2 cells or D1 cells) that can be used to analyze cell differentiation mechanisms. We showed that the 3-D cell-cell and cell-material interactions and oxygenation offered by the Oxy chip promoted osteoblastic differentiation of MSCs. We also used histomorphometric analysis of hematoxylin and eosin staining, quality analyses by μCT and collagen orientation observation with picrosirius red staining in bone regeneration following implantation of three CaPs in a critical-sized defect in mouse calvaria. The in vivo bone formation capacity of the three tested CaP materials was OCP ≥ β-TCP > HA: the newly formed bone by OCP had a structure relatively close to that of the calvaria intact bone. When MSCs were 3-D cultured with the CaP materials using the Oxy chip, the in vitro osteogenic capacity of these materials was highly similar to trends observed in vivo. The in vitro alkaline phosphatase activity of D1 cells had the highest correlation with in vivo bone volume (R = 0.900). Chemical and FTIR spectroscopic analyses confirmed that differentiation of D1 cells could be associated with amorphous calcium phosphate (ACP) precipitation concomitant with OCP hydrolysis. Taken together, hybrid spheroid cultures using the Oxy chip can be used to screen and predict bone forming potential of bone substitute materials. STATEMENT OF SIGNIFICANCE: An oxygen permeable-culture chip (Oxy chip) can be used to induce formation of cell spheroids by mesenchymal stem cells (MSCs). Use of the Oxy chip avoids hypoxia in the spheroid core and enhances MSC osteoblastic differentiation relative to conventional spheroid culture methods. The present study showed that the Oxy chip mimics the in vivo environment associated with bone formation and can be used to generate hybrid spheroids consisting of calcium phosphates and MSCs that are useful for analyzing cell differentiation mechanisms. Bone formation analysis following implantation of calcium phosphate materials in mouse calvaria defects showed positive correlation with the in vitro results. We propose that hybrid spheroids cultured on the Oxy chip can be used to screen and predict the bone forming potential of bone substitute materials.
Collapse
|
6
|
Hiew VV, Simat SFB, Teoh PL. The Advancement of Biomaterials in Regulating Stem Cell Fate. Stem Cell Rev Rep 2018; 14:43-57. [PMID: 28884292 DOI: 10.1007/s12015-017-9764-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Stem cells are well-known to have prominent roles in tissue engineering applications. Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) can differentiate into every cell type in the body while adult stem cells such as mesenchymal stem cells (MSCs) can be isolated from various sources. Nevertheless, an utmost limitation in harnessing stem cells for tissue engineering is the supply of cells. The advances in biomaterial technology allows the establishment of ex vivo expansion systems to overcome this bottleneck. The progress of various scaffold fabrication could direct stem cell fate decisions including cell proliferation and differentiation into specific lineages in vitro. Stem cell biology and biomaterial technology promote synergistic effect on stem cell-based regenerative therapies. Therefore, understanding the interaction of stem cell and biomaterials would allow the designation of new biomaterials for future clinical therapeutic applications for tissue regeneration. This review focuses mainly on the advances of natural and synthetic biomaterials in regulating stem cell fate decisions. We have also briefly discussed how biological and biophysical properties of biomaterials including wettability, chemical functionality, biodegradability and stiffness play their roles.
Collapse
Affiliation(s)
- Vun Vun Hiew
- Biotechonology Research Institute, Universiti Malaysia Sabah, Jalan UMS, 88400, Kota Kinabalu, Sabah, Malaysia
| | - Siti Fatimah Binti Simat
- C/o Biotechonology Research Institute, Universiti Malaysia Sabah, Jalan UMS, 88400, Kota Kinabalu, Sabah, Malaysia
| | - Peik Lin Teoh
- Biotechonology Research Institute, Universiti Malaysia Sabah, Jalan UMS, 88400, Kota Kinabalu, Sabah, Malaysia.
| |
Collapse
|
7
|
Darzi S, Deane JA, Nold CA, Edwards SE, Gough DJ, Mukherjee S, Gurung S, Tan KS, Vashi AV, Werkmeister JA, Gargett CE. Endometrial Mesenchymal Stem/Stromal Cells Modulate the Macrophage Response to Implanted Polyamide/Gelatin Composite Mesh in Immunocompromised and Immunocompetent Mice. Sci Rep 2018; 8:6554. [PMID: 29700360 PMCID: PMC5919927 DOI: 10.1038/s41598-018-24919-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 03/28/2018] [Indexed: 01/09/2023] Open
Abstract
The immunomodulatory properties of human endometrial mesenchymal stem cells (eMSC) have not been well characterised. Initial studies showed that eMSC modulated the chronic inflammatory response to a non-degradable polyamide/gelatin mesh in a xenogeneic rat skin wound repair model, but the mechanism remains unclear. In this study, we investigated the immunomodulatory effect of eMSC on the macrophage response to polyamide/gelatin composite mesh in an abdominal subcutaneous wound repair model in C57BL6 immunocompetent and NSG (NOD-Scid-IL2Rgammanull) immunocompromised mice to determine whether responses differed in the absence of an adaptive immune system and NK cells. mCherry lentivirus-labelled eMSC persisted longer in NSG mice, inducing longer term paracrine effects. Inclusion of eMSC in the mesh reduced inflammatory cytokine (Il-1β, Tnfα) secretion, and in C57BL6 mice reduced CCR7+ M1 macrophages surrounding the mesh on day 3 and increased M2 macrophage marker mRNA (Arg1, Mrc1, Il10) expression at days 3 and 7. In NSG mice, these effects were delayed and only observed at days 7 and 30 in comparison with controls implanted with mesh alone. These results show that the differences in the immune status in the two animals directly affect the survival of xenogeneic eMSC which leads to differences in the short-term and long-term macrophage responses to implanted meshes.
Collapse
Affiliation(s)
- S Darzi
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, 3168, Australia
| | - J A Deane
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, 3168, Australia
| | - C A Nold
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, 3168, Australia
| | - S E Edwards
- CSIRO Manufacturing, Bayview Avenue, Clayton, Victoria, 3169, Australia
| | - D J Gough
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, 3168, Australia
| | - S Mukherjee
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, 3168, Australia
| | - S Gurung
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, 3168, Australia
| | - K S Tan
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, 3168, Australia
| | - A V Vashi
- CSIRO Manufacturing, Bayview Avenue, Clayton, Victoria, 3169, Australia
| | - J A Werkmeister
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, 3168, Australia.,CSIRO Manufacturing, Bayview Avenue, Clayton, Victoria, 3169, Australia
| | - C E Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, 3168, Australia. .,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, 3168, Australia.
| |
Collapse
|
8
|
Duan W, Chen C, Haque M, Hayes D, Lopez MJ. Polymer-mineral scaffold augments in vivo equine multipotent stromal cell osteogenesis. Stem Cell Res Ther 2018. [PMID: 29523214 PMCID: PMC5845133 DOI: 10.1186/s13287-018-0790-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Use of bioscaffolds to direct osteogenic differentiation of adult multipotent stromal cells (MSCs) without exogenous proteins is a contemporary approach to bone regeneration. Identification of in vivo osteogenic contributions of exogenous MSCs on bioscaffolds after long-term implantation is vital to understanding cell persistence and effect duration. Methods This study was designed to quantify in vivo equine MSC osteogenesis on synthetic polymer scaffolds with distinct mineral combinations 9 weeks after implantation in a murine model. Cryopreserved, passage (P)1, equine bone marrow-derived MSCs (BMSC) and adipose tissue-derived MSCs (ASC) were culture expanded to P3 and immunophenotyped with flow cytometry. They were then loaded by spinner flask on to scaffolds composed of tricalcium phosphate (TCP)/hydroxyapatite (HA) (40:60; HT), polyethylene glycol (PEG)/poly-l-lactic acid (PLLA) (60:40; GA), or PEG/PLLA/TCP/HA (36:24:24:16; GT). Scaffolds with and without cells were maintained in static culture for up to 21 days or implanted subcutaneously in athymic mice that were radiographed every 3 weeks up to 9 weeks. In vitro cell viability and proliferation were determined. Explant composition (double-stranded (ds)DNA, collagen, sulfated glycosaminoglycan (sGAG), protein), equine and murine osteogenic target gene expression, microcomputed tomography (μCT) mineralization, and light microscopic structure were assessed. Results The ASC and BMSC number increased significantly in HT constructs between 7 and 21 days of culture, and BMSCs increased similarly in GT constructs. Radiographic opacity increased with time in GT-BMSC constructs. Extracellular matrix (ECM) components and dsDNA increased significantly in GT compared to HT constructs. Equine and murine osteogenic gene expression was highest in BMSC constructs with mineral-containing scaffolds. The HT constructs with either cell type had the highest mineral deposition based on μCT. Regardless of composition, scaffolds with cells had more ECM than those without, and osteoid was apparent in all BMSC constructs. Conclusions In this study, both exogenous and host MSCs appear to contribute to in vivo osteogenesis. Addition of mineral to polymer scaffolds enhances equine MSC osteogenesis over polymer alone, but pure mineral scaffold provides superior osteogenic support. These results emphasize the need for bioscaffolds that provide customized osteogenic direction of both exo- and endogenous MSCs for the best regenerative potential.
Collapse
Affiliation(s)
- Wei Duan
- Laboratory for Equine and Comparative Orthopedic Research, Louisiana State University, Baton Rouge, LA, USA
| | - Cong Chen
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
| | - Masudul Haque
- Laboratory for Equine and Comparative Orthopedic Research, Louisiana State University, Baton Rouge, LA, USA
| | - Daniel Hayes
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
| | - Mandi J Lopez
- Laboratory for Equine and Comparative Orthopedic Research, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
9
|
Rh Owen G, Dard M, Larjava H. Hydoxyapatite/beta-tricalcium phosphate biphasic ceramics as regenerative material for the repair of complex bone defects. J Biomed Mater Res B Appl Biomater 2017; 106:2493-2512. [PMID: 29266701 DOI: 10.1002/jbm.b.34049] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 10/27/2017] [Accepted: 10/31/2017] [Indexed: 01/07/2023]
Abstract
Bone is a composite material composed of collagen and calcium phosphate (CaP) mineral. The collagen gives bone its flexibility while the inorganic material gives bone its resilience. The CaP in bone is similar in composition and structure to the mineral hydroxyapatite (HA) and is bioactive, osteoinductive and osteoconductive. Therefore synthetic versions of bone apatite (BA) have been developed to address the demand for autologous bone graft substitutes. Synthetic HA (s-HA) are stiff and strong, but brittle. These lack of physical attributes limit the use of synthetic apatites in situations where no physical loading of the apatite occurs. s-HA chemical properties differ from BA and thus change the physical and mechanical properties of the material. Consequently, s-HA is more chemically stable than BA and thus its resorption rate is slower than the rate of bone regeneration. One solution to this problem is to introduce a faster resorbing CaP, such as β-tricalcium phosphate (β-TCP), when synthesizing the material creating a biphasic (s-HA and β-TCP) formulation of calcium phosphate (BCP). The focus of this review is to introduce the major differences between BCP and biological apatites and how material scientists have overcome the inadequacies of the synthetic counterparts. Examples of BCP performance in vitro and in vivo following structural and chemical modifications are provided as well as novel ultrastructural data. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 2493-2512, 2018.
Collapse
Affiliation(s)
- Gethin Rh Owen
- Department of Oral, Biological & Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver V6T 1Z3, Canada
| | - Michel Dard
- College of Dentistry, New York University, New York, New York
| | - Hannu Larjava
- Department of Oral, Biological & Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver V6T 1Z3, Canada
| |
Collapse
|
10
|
Duan W, Haque M, Kearney MT, Lopez MJ. Collagen and Hydroxyapatite Scaffolds Activate Distinct Osteogenesis Signaling Pathways in Adult Adipose-Derived Multipotent Stromal Cells. Tissue Eng Part C Methods 2017; 23:592-603. [PMID: 28877641 PMCID: PMC5653142 DOI: 10.1089/ten.tec.2017.0078] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 07/19/2017] [Indexed: 12/13/2022] Open
Abstract
Osteogenic cell signaling pathway disruption varies among bone diseases. This investigation was designed to identify adipose-derived multipotent stromal cell (ASC) and bone graft scaffold combinations for local, targeted restoration of gene expression and extracellular matrix (ECM) deposition. Human ASC osteogenesis on bone graft materials was quantified following culture in stromal (S), osteogenic (O), or osteogenic for 48 h followed by stromal medium (OS) to test the two-part hypothesis: (1) identical ASC isolates on distinct bone graft scaffolds demonstrate unique viability, differentiation, ECM production, and gene expression in the same culture conditions; (2) identical ASC-bone graft scaffold combinations have different cell viability, differentiation, ECM production, and gene expression when cultured in S, O, or OS medium. Three commercially available bone graft scaffold materials, type I bovine collagen (C), hydroxyapatite + β-tricalcium phosphate + type I bovine collagen (HT), and β-tricalcium phosphate + type I bovine collagen (CT) were evaluated. Passage 3 ASCs were loaded onto scaffold blocks with a spinner flask bioreactor, and constructs were cultured up to 28 days. Cell viability, gene expression (alkaline phosphatase [ALPL], osteoprotegerin [TNFRSF11B], osteocalcin [BGLAP], cannabinoid receptors type I [CNR1] and II [CNR2], receptor activator of nuclear factor kappa β ligand [TNFSF11]), as well as ECM DNA, collagen, sulfated glycosaminoglycan, and protein content were quantified. Matrix organization was evaluated with scanning electron microscopy. Effects of scaffold, medium, or culture duration on cell viability were minimal. Significantly higher initial ALPL expression decreased with time, while BGLAP expression increased in HT constructs in O medium, and the constructs had the most abundant ECM components and ultrastructural organization. There was a similar, although delayed, pattern of gene expression and greater ECM collagen with less organization in C constructs in O medium. Higher CNR1 expression in C versus higher TNFRSF11B/TNFSF11 expression in HT constructs throughout the study support stimulation of unique osteogenic signaling pathways by identical cell isolates. These results suggest that bone scaffold composition may be used to selectively target specific osteogenic cell signaling pathways in ASC constructs to stimulate ECM deposition based on therapeutic needs.
Collapse
Affiliation(s)
- Wei Duan
- 1 Laboratory for Equine and Comparative Orthopedic Research, School of Veterinary Medicine, Louisiana State University , Baton Rouge, Louisiana
| | - Masudul Haque
- 1 Laboratory for Equine and Comparative Orthopedic Research, School of Veterinary Medicine, Louisiana State University , Baton Rouge, Louisiana
| | - Michael T Kearney
- 2 Department of Pathobiological Sciences, Louisiana State University , Baton Rouge, Louisiana
| | - Mandi J Lopez
- 1 Laboratory for Equine and Comparative Orthopedic Research, School of Veterinary Medicine, Louisiana State University , Baton Rouge, Louisiana
| |
Collapse
|
11
|
Westhauser F, Senger AS, Reible B, Moghaddam A. * In Vivo Models for the Evaluation of the Osteogenic Potency of Bone Substitutes Seeded with Mesenchymal Stem Cells of Human Origin: A Concise Review. Tissue Eng Part C Methods 2017; 23:881-888. [PMID: 28747099 DOI: 10.1089/ten.tec.2017.0164] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Research concerning bone substitutes is one of the most challenging fields in orthopedic research and has a high clinical relevance, especially since the currently available bone substitutes are limited in their osteostimulative capabilities. In vitro models for the evaluation of the properties of bone substitutes allow the use of human mesenchymal stem cells (hMSCs) seeded onto scaffolds, but suffer from the lack of a physiological environment for those cells. Most in vivo models include the use of non-hMSC and are therefore lacking in clinical relevance. To overcome these issues, in vivo models were created that allow the evaluation of hMSC-seeded bone substitutes, combining the advantages of the use of human cells with the physiological conditions of an organism in vivo. In brief, models usually aim for bone formation in immunocompromised rodents. The subcutaneous implantation of scaffolds is most widely performed, showing low complication rates along with good results, but suffering from inferior vascularization of the implants and the absence of the realistic structural and mechanical conditions of bone. Orthotopic implantation, for example in calvarian or long bone defects, provides the most appropriate surrounding for hMSC-seeded scaffolds. However, parallel host-induced bone formation is a major limitation. This review summarizes in vivo models for the evaluation of the osteogenic potency of bone substitutes seeded with mesenchymal stem cells of human origin.
Collapse
Affiliation(s)
- Fabian Westhauser
- 1 HTRG-Heidelberg Trauma Research Group, Center of Orthopedics, Traumatology, and Spinal Cord Injury, Heidelberg University Hospital , Heidelberg, Germany
| | - Anne-Sophie Senger
- 1 HTRG-Heidelberg Trauma Research Group, Center of Orthopedics, Traumatology, and Spinal Cord Injury, Heidelberg University Hospital , Heidelberg, Germany
| | - Bruno Reible
- 1 HTRG-Heidelberg Trauma Research Group, Center of Orthopedics, Traumatology, and Spinal Cord Injury, Heidelberg University Hospital , Heidelberg, Germany
| | - Arash Moghaddam
- 2 Clinic for Orthopedic Surgery, Trauma Surgery, and Hand Surgery, Klinikum Aschaffenburg-Alzenau , Aschaffenburg, Germany
| |
Collapse
|
12
|
Benning L, Gutzweiler L, Tröndle K, Riba J, Zengerle R, Koltay P, Zimmermann S, Stark GB, Finkenzeller G. Cytocompatibility testing of hydrogels toward bioprinting of mesenchymal stem cells. J Biomed Mater Res A 2017; 105:3231-3241. [DOI: 10.1002/jbm.a.36179] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 07/07/2017] [Accepted: 07/28/2017] [Indexed: 01/09/2023]
Affiliation(s)
- Leo Benning
- Department of Plastic and Hand Surgery; Faculty of Medicine, Medical Center-University of Freiburg, Freiburg; Germany
| | - Ludwig Gutzweiler
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering; University of Freiburg, Georges-Koehler-Allee 103; Freiburg 79110 Germany
- Hahn-Schickard, Georges-Koehler-Allee 103; Freiburg 79110 Germany
| | - Kevin Tröndle
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering; University of Freiburg, Georges-Koehler-Allee 103; Freiburg 79110 Germany
| | - Julian Riba
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering; University of Freiburg, Georges-Koehler-Allee 103; Freiburg 79110 Germany
| | - Roland Zengerle
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering; University of Freiburg, Georges-Koehler-Allee 103; Freiburg 79110 Germany
- Hahn-Schickard, Georges-Koehler-Allee 103; Freiburg 79110 Germany
- FIT-Freiburg Centre for Interactive Materials and Bioinspired Technologies; University of Freiburg, Georges-Koehler-Allee 105; Freiburg 79110 Germany
| | - Peter Koltay
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering; University of Freiburg, Georges-Koehler-Allee 103; Freiburg 79110 Germany
| | - Stefan Zimmermann
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering; University of Freiburg, Georges-Koehler-Allee 103; Freiburg 79110 Germany
| | - G. Björn Stark
- Department of Plastic and Hand Surgery; Faculty of Medicine, Medical Center-University of Freiburg, Freiburg; Germany
| | - Günter Finkenzeller
- Department of Plastic and Hand Surgery; Faculty of Medicine, Medical Center-University of Freiburg, Freiburg; Germany
| |
Collapse
|
13
|
Tanikake Y, Akahane M, Furukawa A, Tohma Y, Inagaki Y, Kira T, Tanaka Y. Calcium Concentration in Culture Medium as a Nondestructive and Rapid Marker of Osteogenesis. Cell Transplant 2016; 26:1067-1076. [PMID: 27983908 DOI: 10.3727/096368916x694166] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Artificial bones made of β-tricalcium phosphate (β-TCP) combined with bone marrow-derived mesenchymal stromal cells (BM-MSCs) are used for effective reconstruction of bone defects caused by genetic defects, traumatic injury, or surgical resection of bone tumors. However, the selection of constructs with high osteogenic potential before implantation is challenging. The purpose of this study was to determine whether the calcium concentration in BM-MSC culture medium can be used as a nondestructive and simple osteogenic marker for selecting tissue-engineered grafts constructed using β-TCP and BM-MSCs. We prepared three cell passages of BM-MSCs derived from three 7-week-old, male Fischer 344 rats; the cells were cultured in osteoinductive medium in the presence of β-TCP for 15 days. The medium was replaced with fresh medium on day 1 in culture and subsequently changed every 48 h; it was collected for measurement of osteocalcin secretion and calcium concentration by enzyme-linked immunosorbent assay and X-ray fluorescence spectrometry, respectively. After cultivation, the constructs were implanted subcutaneously into the backs of recipient rats. Four weeks after implantation, the alkaline phosphatase (ALP) activity and osteocalcin content of the constructs were measured. A strong inverse correlation was observed between the calcium concentration in the medium and the ALP activity and osteocalcin content of the constructs, with Pearson's correlation coefficients of 0.92 and 0.90, respectively. These results indicate that tissue-engineered bone with high osteogenic ability can be selected before implantation based on low calcium content of the culture medium, resulting in successful bone formation after implantation. This nondestructive, simple method shows great promise for assessing the osteogenic ability of tissue-engineered bone.
Collapse
|
14
|
Akahane M, Shimizu T, Kira T, Onishi T, Uchihara Y, Imamura T, Tanaka Y. Culturing bone marrow cells with dexamethasone and ascorbic acid improves osteogenic cell sheet structure. Bone Joint Res 2016; 5:569-576. [PMID: 27881440 PMCID: PMC5131089 DOI: 10.1302/2046-3758.511.bjr-2016-0013.r1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 09/09/2016] [Indexed: 02/06/2023] Open
Abstract
Objectives To assess the structure and extracellular matrix molecule expression of osteogenic cell sheets created via culture in medium with both dexamethasone (Dex) and ascorbic acid phosphate (AscP) compared either Dex or AscP alone. Methods Osteogenic cell sheets were prepared by culturing rat bone marrow stromal cells in a minimal essential medium (MEM), MEM with AscP, MEM with Dex, and MEM with Dex and AscP (Dex/AscP). The cell number and messenger (m)RNA expression were assessed in vitro, and the appearance of the cell sheets was observed after mechanical retrieval using a scraper. β-tricalcium phosphate (β-TCP) was then wrapped with the cell sheets from the four different groups and subcutaneously implanted into rats. Results After mechanical retrieval, the osteogenic cell sheets from the MEM, MEM with AscP, and MEM with Dex groups appeared to be fragmented or incomplete structures. The cell sheets cultured with Dex/AscP remained intact after mechanical retrieval, without any identifiable tears. Culture with Dex/AscP increased the mRNA and protein expression of extracellular matrix proteins and cell number compared with those of the other three groups. More bridging bone formation was observed after transplantation of the β-TCP scaffold wrapped with cell sheets cultured with Dex/AscP, than in the other groups. Conclusions These results suggest that culture with Dex/AscP improves the mechanical integrity of the osteogenic cell sheets, allowing retrieval of the confluent cells in a single cell sheet structure. This method may be beneficial when applied in cases of difficult tissue reconstruction, such as nonunion, bone defects, and osteonecrosis. Cite this article: M. Akahane, T. Shimizu, T. Kira, T. Onishi, Y. Uchihara, T. Imamura, Y. Tanaka. Culturing bone marrow cells with dexamethasone and ascorbic acid improves osteogenic cell sheet structure. Bone Joint Res 2016;5:569–576. DOI: 10.1302/2046-3758.511.BJR-2016-0013.R1.
Collapse
Affiliation(s)
- M Akahane
- Department of Public Health, Health Management and Policy, Nara Medical University Faculty of Medicine, Kashihara, Nara, Japan
| | - T Shimizu
- Department of Orthopedic Surgery, Nara Medical University Faculty of Medicine, Kashihara, Nara, Japan
| | - T Kira
- Department of Orthopedic Surgery, Nara Medical University Faculty of Medicine, Kashihara, Nara, Japan
| | - T Onishi
- Department of Orthopedic Surgery, Nara Medical University Faculty of Medicine, Kashihara, Nara, Japan
| | - Y Uchihara
- Department of Orthopedic Surgery, Nara Medical University Faculty of Medicine, Kashihara, Nara, Japan
| | - T Imamura
- Department of Public Health, Health Management and Policy, Nara Medical University Faculty of Medicine, Kashihara, Nara, Japan
| | - Y Tanaka
- Department of Orthopedic Surgery, Nara Medical University Faculty of Medicine, Kashihara, Nara, Japan
| |
Collapse
|
15
|
Takato T. Tissue Engineering in Bone: The Contention of a Hundred Schools of Thought. Artif Organs 2016; 40:331-3. [PMID: 27076203 DOI: 10.1111/aor.12739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Tsuyoshi Takato
- Department of Oral and Maxillofacial Surgery, The University of Tokyo Hospital, Tokyo, Japan.
| |
Collapse
|
16
|
Suenaga H, Furukawa KS, Suzuki Y, Takato T, Ushida T. Bone regeneration in calvarial defects in a rat model by implantation of human bone marrow-derived mesenchymal stromal cell spheroids. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2015; 26:254. [PMID: 26449444 PMCID: PMC4598349 DOI: 10.1007/s10856-015-5591-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 09/26/2015] [Indexed: 05/26/2023]
Abstract
Mesenchymal stem cell (MSC) condensation contributes to membrane ossification by enhancing their osteodifferentiation. We investigated bone regeneration in rats using the human bone marrow-derived MSC-spheroids prepared by rotation culture, without synthetic or exogenous biomaterials. Bilateral calvarial defects (8 mm) were created in nude male rats; the left-sided defects were implanted with MSC-spheroids, β-tricalcium phosphate (β-TCP) granules, or β-TCP granules + MSC-spheroids, while the right-sided defects served as internal controls. Micro-computed tomography and immunohistochemical staining for osteocalcin/osteopontin indicated formation of new, full-thickness bones at the implantation sites, but not at the control sites in the MSC-spheroid group. Raman spectroscopy revealed similarity in the spectral properties of the repaired bone and native calvarial bone. Mechanical performance of the bones in the MSC-implanted group was good (50 and 60% those of native bones, respectively). All tests showed poor bone regeneration in the β-TCP and β-TCP + MSC-spheroid groups. Thus, significant bone regeneration was achieved with MSC-spheroid implantation into bone defects, justifying further investigation.
Collapse
Affiliation(s)
- Hideyuki Suenaga
- Department of Oral-Maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Katsuko S Furukawa
- Biomedical Engineering Laboratory, Department of Bioengineering and Mechanical Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Yukako Suzuki
- Department of Oral-Maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Tsuyoshi Takato
- Department of Oral-Maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Division of Tissue Engineering, The University of Tokyo Hospital, Tokyo, Japan
| | - Takashi Ushida
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
17
|
Denry I, Kuhn LT. Design and characterization of calcium phosphate ceramic scaffolds for bone tissue engineering. Dent Mater 2015; 32:43-53. [PMID: 26423007 DOI: 10.1016/j.dental.2015.09.008] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 09/04/2015] [Accepted: 09/09/2015] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Our goal is to review design strategies for the fabrication of calcium phosphate ceramic scaffolds (CPS), in light of their transient role in bone tissue engineering and associated requirements for effective bone regeneration. METHODS We examine the various design options available to meet mechanical and biological requirements of CPS and later focus on the importance of proper characterization of CPS in terms of architecture, mechanical properties and time-sensitive properties such as biodegradability. Finally, relationships between in vitro versus in vivo testing are addressed, with an attempt to highlight reliable performance predictors. RESULTS A combinatory design strategy should be used with CPS, taking into consideration 3D architecture, adequate surface chemistry and topography, all of which are needed to promote bone formation. CPS represent the media of choice for delivery of osteogenic factors and anti-infectives. Non-osteoblast mediated mineral deposition can confound in vitro osteogenesis testing of CPS and therefore the expression of a variety of proteins or genes including collagen type I, bone sialoprotein and osteocalcin should be confirmed in addition to increased mineral content. CONCLUSIONS CPS are a superior scaffold material for bone regeneration because they actively promote osteogenesis. Biodegradability of CPS via calcium and phosphate release represents a unique asset. Structural control of CPS at the macro, micro and nanoscale and their combination with cells and polymeric materials is likely to lead to significant developments in bone tissue engineering.
Collapse
Affiliation(s)
- Isabelle Denry
- Department of Prosthodontics, University of Iowa College of Dentistry, 801 Newton Road, Iowa City, IA 52242-1010, USA.
| | - Liisa T Kuhn
- Department of Reconstructive Sciences, UConn Health, 263 Farmington Avenue, MC 1615, Farmington, CT 06030-1615, USA
| |
Collapse
|
18
|
Gamie Z, MacFarlane RJ, Tomkinson A, Moniakis A, Tran GT, Gamie Y, Mantalaris A, Tsiridis E. Skeletal tissue engineering using mesenchymal or embryonic stem cells: clinical and experimental data. Expert Opin Biol Ther 2015; 14:1611-39. [PMID: 25303322 DOI: 10.1517/14712598.2014.945414] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Mesenchymal stem cells (MSCs) can be obtained from a wide variety of tissues for bone tissue engineering such as bone marrow, adipose, birth-associated, peripheral blood, periosteum, dental and muscle. MSCs from human fetal bone marrow and embryonic stem cells (ESCs) are also promising cell sources. AREAS COVERED In vitro, in vivo and clinical evidence was collected using MEDLINE® (1950 to January 2014), EMBASE (1980 to January 2014) and Google Scholar (1980 to January 2014) databases. EXPERT OPINION Enhanced results have been found when combining bone marrow-derived mesenchymal stem cells (BMMSCs) with recently developed scaffolds such as glass ceramics and starch-based polymeric scaffolds. Preclinical studies investigating adipose tissue-derived stem cells and umbilical cord tissue-derived stem cells suggest that they are likely to become promising alternatives. Stem cells derived from periosteum and dental tissues such as the periodontal ligament have an osteogenic potential similar to BMMSCs. Stem cells from human fetal bone marrow have demonstrated superior proliferation and osteogenic differentiation than perinatal and postnatal tissues. Despite ethical concerns and potential for teratoma formation, developments have also been made for the use of ESCs in terms of culture and ideal scaffold.
Collapse
Affiliation(s)
- Zakareya Gamie
- Aristotle University Medical School, 'PapaGeorgiou' Hospital, Academic Orthopaedic Unit , Thessaloniki , Greece
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Ueha T, Akahane M, Shimizu T, Uchihara Y, Morita Y, Nitta N, Kido A, Inagaki Y, Kawate K, Tanaka Y. Utility of tricalcium phosphate and osteogenic matrix cell sheet constructs for bone defect reconstruction. World J Stem Cells 2015; 7:873-882. [PMID: 26131318 PMCID: PMC4478634 DOI: 10.4252/wjsc.v7.i5.873] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/08/2014] [Accepted: 03/20/2015] [Indexed: 02/07/2023] Open
Abstract
AIM: To determine the effects of transplanting osteogenic matrix cell sheets and beta-tricalcium phosphate (TCP) constructs on bone formation in bone defects.
METHODS: Osteogenic matrix cell sheets were prepared from bone marrow stromal cells (BMSCs), and a porous TCP ceramic was used as a scaffold. Three experimental groups were prepared, comprised of TCP scaffolds (1) seeded with BMSCs; (2) wrapped with osteogenic matrix cell sheets; or (3) both. Constructs were implanted into a femoral defect model in rats and bone growth was evaluated by radiography, histology, biochemistry, and mechanical testing after 8 wk.
RESULTS: In bone defects, constructs implanted with cell sheets showed callus formation with segmental or continuous bone formation at 8 wk, in contrast to TCP seeded with BMSCs, which resulted in bone non-union. Wrapping TCP constructs with osteogenic matrix cell sheets increased their osteogenic potential and resulting bone formation, compared with conventional bone tissue engineering TCP scaffolds seeded with BMSCs. The compressive stiffness (mean ± SD) values were 225.0 ± 95.7, 30.0 ± 11.5, and 26.3 ± 10.6 MPa for BMSC/TCP/Sheet constructs with continuous bone formation, BMSC/TCP/Sheet constructs with segmental bone formation, and BMSC/TCP constructs, respectively. The compressive stiffness of BMSC/TCP/Sheet constructs with continuous bone formation was significantly higher than those with segmental bone formation and BMSC/TCP constructs.
CONCLUSION: This technique is an improvement over current methods, such as TCP substitution, and is useful for hard tissue reconstruction and inducing earlier bone union in defects.
Collapse
|
20
|
Biomineral/Agarose Composite Gels Enhance Proliferation of Mesenchymal Stem Cells with Osteogenic Capability. Int J Mol Sci 2015; 16:14245-58. [PMID: 26110392 PMCID: PMC4490550 DOI: 10.3390/ijms160614245] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Revised: 06/10/2015] [Accepted: 06/16/2015] [Indexed: 12/02/2022] Open
Abstract
Hydroxyapatite (HA) or calcium carbonate (CaCO3) formed on an organic polymer of agarose gel is a biomaterial that can be used for bone tissue regeneration. However, in critical bone defects, the regeneration capability of these materials is limited. Mesenchymal stem cells (MSCs) are multipotent cells that can differentiate into bone forming osteoblasts. In this study, we loaded MSCs on HA- or CaCO3-formed agarose gel and cultured them with dexamethasone, which triggers the osteogenic differentiation of MSCs. High alkaline phosphatase activity was detected on both the HA- and CaCO3-formed agarose gels; however, basal activity was only detected on bare agarose gel. Bone-specific osteocalcin content was detected on CaCO3-formed agarose gel on Day 14 of culture, and levels subsequently increased over time. Similar osteocalcin content was detected on HA-formed agarose on Day 21 and levels increased on Day 28. In contrast, only small amounts of osteocalcin were found on bare agarose gel. Consequently, osteogenic capability of MSCs was enhanced on CaCO3-formed agarose at an early stage, and both HA- and CaCO3-formed agarose gels well supported the capability at a later stage. Therefore, MSCs loaded on either HA- or CaCO3-formed agarose could potentially be employed for the repair of critical bone defects.
Collapse
|
21
|
Evaluation of the osteoinductive potential of a bio-inspired scaffold mimicking the osteogenic niche for bone augmentation. Biomaterials 2015; 62:128-37. [PMID: 26048479 DOI: 10.1016/j.biomaterials.2015.05.011] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 05/02/2015] [Accepted: 05/14/2015] [Indexed: 01/18/2023]
Abstract
Augmentation of regenerative osteogenesis represents a premier clinical need, as hundreds of thousands of patients are left with insufficient healing of bony defects related to a host of insults ranging from congenital abnormalities to traumatic injury to surgically-induced deficits. A synthetic material that closely mimics the composition and structure of the human osteogenic niche represents great potential to successfully address this high demand. In this study, a magnesium-doped hydroxyapatite/type I collagen scaffold was fabricated through a biologically-inspired mineralization process and designed to mimic human trabecular bone. The composition of the scaffold was fully characterized by XRD, FTIR, ICP and TGA, and compared to human bone. Also, the scaffold microstructure was evaluated by SEM, while its nano-structure and nano-mechanical properties were evaluated by AFM. Human bone marrow-derived mesenchymal stem cells were used to test the in vitro capability of the scaffold to promote osteogenic differentiation. The cell/scaffold constructs were cultured up to 7 days and the adhesion, organization and proliferation of the cells were evaluated. The ability of the scaffold to induce osteogenic differentiation of the cells was assessed over 3 weeks and the correlate gene expression for classic genes of osteogenesis was assessed. Finally, when tested in an ectopic model in rabbit, the scaffold produced a large volume of trabecular bone in only two weeks, that subsequently underwent maturation over time as expected, with increased mature cortical bone formation, supporting its ability to promote bone regeneration in clinically-relevant scenarios. Altogether, these results confirm a high level of structural mimicry by the scaffold to the composition and structure of human osteogenic niche that translated to faster and more efficient osteoinduction in vivo--features that suggest such a biomaterial may have great utility in future clinical applications where bone regeneration is required.
Collapse
|
22
|
Response of stem cells from different origins to biphasic calcium phosphate bioceramics. Cell Tissue Res 2015; 361:477-95. [PMID: 25676006 PMCID: PMC4529461 DOI: 10.1007/s00441-015-2116-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 01/05/2015] [Indexed: 12/21/2022]
Abstract
Biphasic calcium phosphate (BCP) bioceramics have been successfully applied in a broad variety of presentation forms and with different ratios of hydroxyapatite (HA) and β-tricalcium phosphate (β-TCP). BCPs have been loaded with stem cells from different origins for bone tissue engineering purposes, but evidence of stem cell behavior on different compositions (various HA/β-TCP ratios) and physical features of BCPs is limited. We compared the adhesion, proliferation, viability and osteogenic potential of human mesenchymal stem cells (MSCs) on granular BCPs with equal HA/β-TCP ratio of diverse particle sizes and on porous blocks which had different chemical compositions. In addition, the osteogenic differentiation of MSCs was compared to adipose-derived (ADSC) and dental pulp (DPSC) stem cells, as well as to pre-osteoblasts on a particulate BCP. MSCs growing on granular BCPs demonstrated increased number as compared to MSCs growing on blocks. Cells proliferated to a greater extent on small granular BCPs, while large granular BCPs and blocks promoted cell differentiation. Surprisingly, the expression of genes involved in osteogenesis was upregulated in MSCs on bioceramics in basal medium which indicates that BCPs may have osteoinductive potential. This was confirmed with the upregulation of osteochondrogenic markers, at different time points, when stem cells from various tissues were grown on the BCP. This study demonstrates that BCPs, depending on their physical features and chemical composition, modulate stem cell behavior, and that stem cells from different origins are inherently distinct in their gene expression profile and can be triggered toward osteochondrogenic fate by BCPs.
Collapse
|
23
|
Lenze U, Pohlig F, Seitz S, Ern C, Milz S, Docheva D, Schieker M. Influence of osteogenic stimulation and VEGF treatment on in vivo bone formation in hMSC-seeded cancellous bone scaffolds. BMC Musculoskelet Disord 2014; 15:350. [PMID: 25323565 PMCID: PMC4216837 DOI: 10.1186/1471-2474-15-350] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 09/23/2014] [Indexed: 01/24/2023] Open
Abstract
Background Tissue engineering approaches for reconstruction of large bone defects are still technically immature, especially in regard to sufficient blood supply. Therefore, the aim of the present study was to investigate the influence of osteogenic stimulation and treatment with VEGF on new bone formation and neovascularization in hMSC-loaded cancellous bone scaffolds in vivo. Methods Cubic scaffolds were seeded with hMSC and either cultured in stem cell medium or osteogenic stimulation medium. One osteogenically stimulated group was additionally treated with 0.8 μg VEGF prior to subcutaneous implantation in athymic mice. After 2 and 12 weeks in vivo, constructs and selected organs were harvested for histological and molecular analysis. Results Histological analysis revealed similar vascularization of the constructs with and without VEGF treatment and absence of new bone formation in any group. Human DNA was detected in all inoculated scaffolds, but a significant decrease in cells was observed after 2 weeks with no further decrease after 12 weeks in vivo. Conclusion Under the chosen conditions, osteogenic stimulation and treatment with VEGF does not have any influence on the new bone formation and neovascularization in hMSC-seeded cancellous bone scaffolds. Electronic supplementary material The online version of this article (doi:10.1186/1471-2474-15-350) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | - Denitsa Docheva
- Laboratory of Experimental Surgery and Regenerative Medicine, Department of Surgery, University of Munich (LMU), Munich, Germany.
| | | |
Collapse
|
24
|
Akahane M, Ueha T, Shimizu T, Shigematsu H, Kido A, Omokawa S, Kawate K, Imamura T, Tanaka Y. Cell sheet injection as a technique of osteogenic supply. Int J Stem Cells 2014; 3:138-43. [PMID: 24855551 DOI: 10.15283/ijsc.2010.3.2.138] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2010] [Indexed: 12/26/2022] Open
Abstract
We previously reported a new cell transplantation method utilizing injections of mesenchymal stem cell (MSC) sheets that have osteogenic potential. After subcutaneous transplantation without any scaffold, the sheet demonstrated in vivo bone formation. In the present study, we transplanted such sheets by injection into implanted ceramics and assessed whether the injectable MSC sheets could stimulate osteogenic integration of the ceramics. To fabricate MSC sheets, bone marrow cells cultured from femur shafts of 7-week-old rats were subcultured in regular 10-cm dishes containing dexamethasone and ascorbic acid phosphate until confluent. Each cell sheet was then lifted using a scraper. Porous β-tricalcium phosphate (β-TCP) disks (5 mm Φ×2 mm) were transplanted subcutaneously into the backs of the rats. Immediately following implantation, the sheets were injected around the disks via a 16G needle (immediate group). Cell sheets were also injected into the remaining implanted disks 1 week after disk implantation (1-wk group). Four weeks following sheet injection, radiography and histology revealed calcification and bone tissue around the harvested disks of the immediate group (eight disks exhibited bone formation/eight implanted disks), whereas calcification and bone tissue were observed in 50% of the samples in the 1-wk group (four disks exhibited bone formation/eight implanted disks). The present study indicates that injected cell sheets can supply osteogenic potential to implanted ceramics. Owing to the usage of a needle for cell sheet transplantation, such an injection method can be applied as a minimally invasive technique of osteogenic supply to implanted ceramics.
Collapse
Affiliation(s)
- M Akahane
- Department of Public Health, Health Management and Policy, Nara Medical University School of Medicine, Nara, Japan
| | - T Ueha
- Department of Orthopedic Surgery, Nara Medical University School of Medicine, Nara, Japan
| | - T Shimizu
- Department of Orthopedic Surgery, Nara Medical University School of Medicine, Nara, Japan
| | - H Shigematsu
- Department of Orthopedic Surgery, Nara Medical University School of Medicine, Nara, Japan
| | - A Kido
- Department of Orthopedic Surgery, Nara Medical University School of Medicine, Nara, Japan
| | - S Omokawa
- Department of Orthopedic Surgery, Nara Medical University School of Medicine, Nara, Japan
| | - K Kawate
- Department of Orthopedic Surgery, Nara Medical University School of Medicine, Nara, Japan
| | - T Imamura
- Department of Public Health, Health Management and Policy, Nara Medical University School of Medicine, Nara, Japan
| | - Y Tanaka
- Department of Orthopedic Surgery, Nara Medical University School of Medicine, Nara, Japan
| |
Collapse
|
25
|
Ma J, Yang F, Both SK, Prins HJ, Helder MN, Pan J, Cui FZ, Jansen JA, van den Beucken JJ. Bone forming capacity of cell- and growth factor-based constructs at different ectopic implantation sites. J Biomed Mater Res A 2014; 103:439-50. [DOI: 10.1002/jbm.a.35192] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 03/14/2014] [Accepted: 04/04/2014] [Indexed: 12/29/2022]
Affiliation(s)
- Jinling Ma
- Department of Biomaterials; Radboud University Medical Centre; Nijmegen the Netherlands
- Department of VIP service; Beijing Stomatological Hospital, Capital Medical University; Beijing 100050 China
| | - Fang Yang
- Department of Biomaterials; Radboud University Medical Centre; Nijmegen the Netherlands
| | - Sanne K. Both
- Department of Biomaterials; Radboud University Medical Centre; Nijmegen the Netherlands
| | - Henk-Jan Prins
- Department of Oral Cell Biology; Academic Centre for Dentistry Amsterdam; University of Amsterdam and VU University Amsterdam; Amsterdam the Netherlands
- Department of Oral and Maxillofacial Surgery; VU University Medical Centre/ACTA; Amsterdam the Netherlands
| | - Marco N. Helder
- Department of Orthopedic Surgery; VU University Medical Centre; Amsterdam the Netherlands
| | - Juli Pan
- Department of VIP service; Beijing Stomatological Hospital, Capital Medical University; Beijing 100050 China
- Department of Oral and Maxillofacial Surgery; Beijing Stomatological Hospital, Capital Medical University; Beijing 100050 China
| | - Fu-Zhai Cui
- Department of Materials Science and Engineering; State Key Laboratory of New Ceramics and Fine Processing; Tsinghua University; Beijing 100084 China
| | - John A. Jansen
- Department of Biomaterials; Radboud University Medical Centre; Nijmegen the Netherlands
| | | |
Collapse
|
26
|
Ohgushi H. Osteogenically differentiated mesenchymal stem cells and ceramics for bone tissue engineering. Expert Opin Biol Ther 2013; 14:197-208. [PMID: 24308323 DOI: 10.1517/14712598.2014.866086] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION In the human body, cells having self-renewal and multi-differentiation capabilities reside in many tissues and are called adult stem cells. In bone marrow tissue, two types of stem cells are well known: hematopoietic stem cells and mesenchymal stem cells (MSCs). Though the number of MSCs in bone marrow tissue is very low, it can be increased by in vitro culture of the marrow, and culture-expanded MSCs are available for various tissue regeneration. AREAS COVERED The culture-expanded MSCs can further differentiate into osteogenic cells such as bone forming osteoblasts by culturing the MSCs in an osteogenic medium. This paper discusses osteogenically differentiated MSCs derived from the bone marrow of patients. Importantly, the differentiation can be achieved on ceramic surfaces which demonstrate mineralized bone matrix formation as well as appearance of osteogenic cells. The cell/matrix/ceramic constructs could show immediate in vivo bone formation and are available for bone reconstruction surgery. EXPERT OPINION Currently, MSCs are clinically available for the regeneration of various tissues due to their high proliferation/differentiation capabilities. However, the capabilities are still limited and thus technologies to improve or recover the inherent capabilities of MSCs are needed.
Collapse
Affiliation(s)
- Hajime Ohgushi
- Department Head, Ookuma Hospital, Department of Orthopedics , 2-17-13 Kuise-honmachi, Amagasaki City, Hyogo 660-0814 , Japan +81-6-6481-1667 ; +81-6-6481-4234
| |
Collapse
|
27
|
Wagner-Ecker M, Voltz P, Egermann M, Richter W. The collagen component of biological bone graft substitutes promotes ectopic bone formation by human mesenchymal stem cells. Acta Biomater 2013; 9:7298-307. [PMID: 23542556 DOI: 10.1016/j.actbio.2013.03.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 03/06/2013] [Accepted: 03/23/2013] [Indexed: 12/13/2022]
Abstract
Synthetic bone substitutes are attractive materials for repairing a variety of bone defects. They are readily available in unlimited quantities, have a defined composition without batch variability and bear no risk of disease transmission. When combined with mesenchymal stem cells (MSCs), bone healing can be further enhanced due to the osteogenic potential of these cells. However, human MSCs showed considerable donor variability in ectopic bone formation assays on synthetic bone substitutes, which may limit clinical success. This study addresses whether bone formation variability of MSCs is cell-intrinsic or biomaterial-dependent and may be improved using biological bone substitutes with and without collagen. Ectopic bone formation of MSCs from nine donors was tested in immune-deficient mice on biological bone substitutes of bovine and equine origin, containing collagen (bHA-C; eHA-C) or not (bHA; eHA). Synthetic β-TCP was used for comparison. Histology of 8-week explants demonstrated a significant influence of the bone graft substitute (BGS) on donor variability of ectopic bone formation with best results seen for eHA-C (15/17) and β-TCP (16/18). Bone was of human origin in all groups according to species-specific in situ hybridization, but MSCs from one donor formed no bone with any bone substitute. According to histomorphometry, most neo-bone was formed on eHA-C with significant differences to bHA, eHA and β-TCP (p<0.001). Collagen-free biological BGSs were inferior to biological BGSs with collagen (p<0.001), while species-origin was of little influence. In conclusion, BGS composition had a strong influence on ectopic bone formation ability of MSCs, and biological BGSs with a collagen component seem most promising to display the strong osteogenic potential of MSCs.
Collapse
|
28
|
Rathbone CR, Guda T, Singleton BM, Oh DS, Appleford MR, Ong JL, Wenke JC. Effect of cell-seeded hydroxyapatite scaffolds on rabbit radius bone regeneration. J Biomed Mater Res A 2013; 102:1458-66. [DOI: 10.1002/jbm.a.34834] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 05/31/2013] [Accepted: 06/05/2013] [Indexed: 01/18/2023]
Affiliation(s)
- C. R. Rathbone
- Extremity Trauma and Regenerative Medicine, U.S. Army Institute of Surgical Research; Ft. Sam Houston Texas
| | - T. Guda
- Extremity Trauma and Regenerative Medicine, U.S. Army Institute of Surgical Research; Ft. Sam Houston Texas
- Department of Biomedical Engineering, The University of Texas at San Antonio; San Antonio Texas
| | - B. M. Singleton
- Department of Biomedical Engineering, The University of Texas at San Antonio; San Antonio Texas
| | - D. S. Oh
- Department of Biomedical Engineering, The University of Texas at San Antonio; San Antonio Texas
- Department of Orthopaedic Surgery, Columbia University Medical Center; New York New York
| | - M. R. Appleford
- Department of Biomedical Engineering, The University of Texas at San Antonio; San Antonio Texas
| | - J. L. Ong
- Department of Biomedical Engineering, The University of Texas at San Antonio; San Antonio Texas
| | - J. C. Wenke
- Extremity Trauma and Regenerative Medicine, U.S. Army Institute of Surgical Research; Ft. Sam Houston Texas
| |
Collapse
|
29
|
Barradas AMC, Monticone V, Hulsman M, Danoux C, Fernandes H, Tahmasebi Birgani Z, Barrère-de Groot F, Yuan H, Reinders M, Habibovic P, van Blitterswijk C, de Boer J. Molecular mechanisms of biomaterial-driven osteogenic differentiation in human mesenchymal stromal cells. Integr Biol (Camb) 2013; 5:920-31. [PMID: 23752904 DOI: 10.1039/c3ib40027a] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Calcium phosphate (CaP) based ceramics are used as bone graft substitutes in the treatment of bone defects. The physico-chemical properties of these materials determine their bioactivity, meaning that molecular and cellular responses in the body will be tuned accordingly. In a previous study, we compared two porous CaP ceramics, hydroxyapatite (HA) and β-tricalcium phosphate (TCP), which, among other properties, differ in their degradation behaviour in vitro and in vivo, and we demonstrated that the more degradable β-TCP induced more bone formation in a heterotopic model in sheep. This is correlated to in vitro data, where human bone marrow derived mesenchymal stromal cells (MSC) exhibited higher expression of osteogenic differentiation markers, such as osteopontin, osteocalcin and bone sialoprotein, when cultured in β-TCP than in HA. More recently, we also showed that this effect could be mimicked in vitro by exposure of MSC to high concentrations of calcium ions (Ca(2+)). To further correlate surface physico-chemical dynamics of HA and β-TCP ceramics with the molecular response of MSC, we followed Ca(2+) release and surface changes in time as well as cell attachment and osteogenic differentiation of MSC on these ceramics. Within 24 hours, we observed differences in cell morphology, with MSC cultured in β-TCP displaying more pronounced attachment and spreading than cells cultured in HA. In the same time frame, β-TCP induced expression of G-protein coupled receptor (GPCR) 5A and regulator of G-protein signaling 2, revealed by DNA microarray analysis. These genes, associated with the protein kinase A and GPCR signaling pathways, may herald the earliest response of MSC to bone-inducing ceramics.
Collapse
Affiliation(s)
- Ana M C Barradas
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Shimizu T, Akahane M, Ueha T, Kido A, Omokawa S, Kobata Y, Murata K, Kawate K, Tanaka Y. Osteogenesis of cryopreserved osteogenic matrix cell sheets. Cryobiology 2013; 66:326-32. [PMID: 23562780 DOI: 10.1016/j.cryobiol.2013.03.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 03/18/2013] [Accepted: 03/18/2013] [Indexed: 12/25/2022]
Abstract
Cryopreservation of tissue engineered bone (TEB), whilst maintaining its osteogenic ability, is imperative for large-scale clinical application. We previously reported a novel cell transplantation method, in which bone-marrow-derived mesenchymal stem cells (BMSCs) were cultured to confluence and differentiated down the osteogenic lineage to form osteogenic matrix cell sheets (OMCS). OMCS have high alkaline phosphatase (ALP) activity and osteocalcin (OC) contents and can be easily used for producing TEB. The aim of the present study was to investigate whether TEB produced by cryopreserved OMCS maintains sufficient osteogenic potential in vivo. OMCS were prepared and divided into three groups according to storage period of cryopreservation (fresh (no cryopreservation), 4 week and 12 week cryopreservation groups). OMCS were cryopreserved by storage in freezing medium (Cell Banker 1®) at -80 °C. Cryopreserved OMCSs were rapidly thawed at room temperature and wrapped around Hydroxyapatite (HA) scaffolds prior to implantation into subcutaneous sites in rats, to determine their in vivo bone-forming capability. The constructs were harvested 4 weeks after transplantation and examined histologically and biochemically. Histological analysis of the constructs showed extensive bone formation in the HA pores with high ALP activity and OC content detected in the cryopreservation groups. The present study clearly indicates that cryopreserved/thawed OMCS are still capable of producing mineralized matrix on scaffolds, resulting in bone formation. This cryopreservation technique could be applied for hard tissue reconstruction to ease the cell preparation method prior to time of use.
Collapse
Affiliation(s)
- Takamasa Shimizu
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara 634-8522, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Farré-Guasch E, Prins HJ, Overman JR, ten Bruggenkate CM, Schulten EA, Helder MN, Klein-Nulend J. Human Maxillary Sinus Floor Elevation as a Model for Bone Regeneration Enabling the Application of One-Step Surgical Procedures. TISSUE ENGINEERING PART B-REVIEWS 2013; 19:69-82. [DOI: 10.1089/ten.teb.2012.0404] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Elisabet Farré-Guasch
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam, Research Institute MOVE, University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Henk-Jan Prins
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam, Research Institute MOVE, University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
- Department of Oral and Maxillofacial Surgery, Research Institute MOVE, VU University Medical Center/Academic Centre for Dentistry Amsterdam, Amsterdam, The Netherlands
| | - Janice R. Overman
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam, Research Institute MOVE, University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
- Department of Oral and Maxillofacial Surgery, Research Institute MOVE, VU University Medical Center/Academic Centre for Dentistry Amsterdam, Amsterdam, The Netherlands
| | - Christiaan M. ten Bruggenkate
- Department of Oral and Maxillofacial Surgery, Research Institute MOVE, VU University Medical Center/Academic Centre for Dentistry Amsterdam, Amsterdam, The Netherlands
| | - Engelbert A.J.M. Schulten
- Department of Oral and Maxillofacial Surgery, Research Institute MOVE, VU University Medical Center/Academic Centre for Dentistry Amsterdam, Amsterdam, The Netherlands
| | - Marco N. Helder
- Department of Orthopaedics, Research Institute MOVE, VU University Medical Center, Amsterdam, The Netherlands
| | - Jenneke Klein-Nulend
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam, Research Institute MOVE, University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
32
|
Overman JR, Farré-Guasch E, Helder MN, ten Bruggenkate CM, Schulten EAJM, Klein-Nulend J. Short (15 minutes) bone morphogenetic protein-2 treatment stimulates osteogenic differentiation of human adipose stem cells seeded on calcium phosphate scaffolds in vitro. Tissue Eng Part A 2012; 19:571-81. [PMID: 23075297 DOI: 10.1089/ten.tea.2012.0133] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
A one-step concept for bone regeneration has been postulated in which human adipose stem cells (hASCs) are harvested, triggered to differentiate, seeded on carriers, and implanted in the same operative procedure. Toward this goal it was investigated whether short (minutes) incubation with bone morphogenetic protein-2 (BMP-2) suffices to trigger osteogenic differentiation of hASCs seeded on calcium phosphate carriers. hASCs were treated with or without BMP-2 (10 ng/mL) for 15 min, and seeded on β-tricalcium phosphate granules (β-TCP; sized <0.7 mm or >0.7 mm) or biphasic calcium phosphate (BCP; 60%/40% or 20%/80% hydroxyapatite/β-TCP). Attachment was determined after 10-30 min. Proliferation (DNA content) and osteogenic differentiation (alkaline phosphatase activity, gene expression) were analyzed up to 3 weeks of culture. hASC attachment to the different scaffolds was similar, and unaffected by BMP-2. It stimulated gene expression of the osteogenic markers core binding factor alpha 1, collagen-1, osteonectin, and osteocalcin in hASCs seeded on BCP and β-TCP. Downregulation of osteopontin expression by BMP-2 was seen in BCP-seeded cells only. BMP-2 treatment inhibited expression of the adipogenic marker peroxisome proliferator-activated receptor gamma. In conclusion, 15 min BMP-2 preincubation of hASCs seeded on BCP/β-TCP scaffolds had a long-lasting stimulating effect on osteogenic differentiation in vitro. These results strongly support a one-step clinical concept for bone regeneration.
Collapse
Affiliation(s)
- Janice R Overman
- Department of Oral Cell Biology, Academic Centre of Dentistry Amsterdam, Research Institute MOVE, University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
33
|
Gamie Z, Tran GT, Vyzas G, Korres N, Heliotis M, Mantalaris A, Tsiridis E. Stem cells combined with bone graft substitutes in skeletal tissue engineering. Expert Opin Biol Ther 2012; 12:713-29. [DOI: 10.1517/14712598.2012.679652] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
34
|
Erdman CP, Dosier CR, Olivares-Navarrete R, Baile C, Guldberg RE, Schwartz Z, Boyan BD. Effects of resveratrol on enrichment of adipose-derived stem cells and their differentiation to osteoblasts in two-and three-dimensional cultures. J Tissue Eng Regen Med 2012; 6 Suppl 3:s34-46. [PMID: 22467433 DOI: 10.1002/term.513] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 04/06/2011] [Accepted: 08/27/2011] [Indexed: 01/05/2023]
Abstract
The goal of this study was to develop a method for increasing the yield of multipotent adipose-derived mesenchymal stem cells (ASCs) and osteoprogenitor cells (OPCs) from subcutaneous fat. After removing mature adipocytes and haematopoietic cells from rat inguinal fat, ASCs in the remaining cell population were verified by their attachment to plastic, surface marker profile (CD271(+), CD73(+) and CD45(-)) and ability to differentiate into adipocytes, chondrocytes and osteoblasts. OPCs were defined as E11(+) and OCN(+). Adherent cells were cultured in growth medium (GM) or osteogenic medium (OM) and treated with resveratrol (0, 12.5, and 25 µM) for 7 days; ASCs and OPCs were assessed by flow cytometry. Osteogenic potential was determined in two-dimensional (2D) cultures as a function of alkaline phosphatase-specific activity and osteocalcin production. In addition, cells were seeded onto three-dimensional (3D) poly-ε-caprolactone scaffolds and cultured under dynamic conditions; mineralization was quantified by micro-CT at 4, 8 and 12 weeks. Resveratrol increased the percentage of ASCs in the population (population%) and number of ASCs in both GM and OM, but increased only the number of OPCs in GM. In both media types resveratrol increased alkaline phosphatase activity and osteocalcin levels. In 3D cultures, resveratrol-treated cells significantly increased mineralized matrix volume at early time points. Resveratrol exerted a biphasic effect on adherent cells by enriching the ASC and OPC populations and enhancing osteogenic differentiation. Resveratrol pretreatment induced more mineralization at earlier time points and represents a clinically viable technique for orthopaedic and dental applications for autologous stem cell therapy.
Collapse
Affiliation(s)
- Christopher P Erdman
- Parker H Petit Institute for Bioengineering and Bioscience, Georgia, Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Imamura T, Ishizuka O, Nishizawa O. Autologous Bone Marrow-Derived Cells Regenerate Urethral Sphincters. Low Urin Tract Symptoms 2012; 4 Suppl 1:87-94. [PMID: 26676706 DOI: 10.1111/j.1757-5672.2011.00136.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Regenerative medicine based on tissue engineering and/or stem cell therapy techniques has the potential to improve irreversibly damaged tissues. Surgical injury to the lower urinary tract can occur as a result of radical prostatectomy or bladder neck surgery. Regeneration of urethral sphincters could be an effective treatment for post-surgical intrinsic sphincter deficiency (ISD)-related urinary incontinence. The replacement, enhancement, and/or recovery the urethral sphincter striated and smooth muscles could increase urethral closure pressure to help patients regain continence. Stem cells from muscle-derived satellite or adipose-derived mesenchymal cells provide temporary improvement in urethral closure pressure but do not reconstruct the muscle layer structures. Our strategy to accomplish regeneration of urethral sphincters is the utilization of autologous bone marrow-derived cells. We have developed a freeze injury model of ISD in rabbits. Freezing of the urinary sphincter causes loss of the majority of striated and smooth muscle cells, and causes a significant decrease in leak point pressure. In this review, we show that the autologous bone marrow-derived cells implanted within the freeze-injured sphincters differentiate into striated or smooth muscle cells. These cells then develop to reconstitute muscle layer structures within the sphincter. Furthermore, the leak point pressure of cell-implanted rabbits is significantly higher than that of cell-free injected controls. We conclude that implantation of autologous bone marrow-derived cells could be an effective treatment for human post-surgical ISD-related urinary incontinence.
Collapse
Affiliation(s)
- Tetsuya Imamura
- Department of Lower Urinary Tract Medicine, Shinshu University School of Medicine, Nagano, JapanDepartment of Urology, Shinshu University School of Medicine, Nagano, Japan
| | - Osamu Ishizuka
- Department of Lower Urinary Tract Medicine, Shinshu University School of Medicine, Nagano, JapanDepartment of Urology, Shinshu University School of Medicine, Nagano, Japan
| | - Osamu Nishizawa
- Department of Lower Urinary Tract Medicine, Shinshu University School of Medicine, Nagano, JapanDepartment of Urology, Shinshu University School of Medicine, Nagano, Japan
| |
Collapse
|
36
|
Scott MA, Levi B, Askarinam A, Nguyen A, Rackohn T, Ting K, Soo C, James AW. Brief review of models of ectopic bone formation. Stem Cells Dev 2012; 21:655-67. [PMID: 22085228 DOI: 10.1089/scd.2011.0517] [Citation(s) in RCA: 146] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Ectopic bone formation is a unique biologic entity--distinct from other areas of skeletal biology. Animal research models of ectopic bone formation most often employ rodent models and have unique advantages over orthotopic (bone) environments, including a relative lack of bone cytokine stimulation and cell-to-cell interaction with endogenous (host) bone-forming cells. This allows for relatively controlled in vivo experimental bone formation. A wide variety of ectopic locations have been used for experimentation, including subcutaneous, intramuscular, and kidney capsule transplantation. The method, benefits and detractions of each method are summarized in the following review. Briefly, subcutaneous implantation is the simplest method. However, the most pertinent concern is the relative paucity of bone formation in comparison to other models. Intramuscular implantation is also widely used and relatively simple, however intramuscular implants are exposed to skeletal muscle satellite progenitor cells. Thus, distinguishing host from donor osteogenesis becomes challenging without cell-tracking studies. The kidney capsule (perirenal or renal capsule) method is less widely used and more technically challenging. It allows for supraphysiologic blood and nutrient resource, promoting robust bone growth. In summary, ectopic bone models are extremely useful in the evaluation of bone-forming stem cells, new osteoinductive biomaterials, and growth factors; an appropriate choice of model, however, will greatly increase experimental success.
Collapse
Affiliation(s)
- Michelle A Scott
- Orthodontics and Dentofacial Orthopedics, Roseman University of Health Sciences, Henderson, Nevada, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Shirai Y, Okuda K, Kubota T, Wolff LF, Yoshie H. The comparative effectiveness of granules or blocks of superporous hydroxyapatite for the treatment of intrabony periodontal defects. ACTA ACUST UNITED AC 2012. [DOI: 10.4236/ojst.2012.22015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
38
|
Akahane M, Ueha T, Shimizu T, Inagaki Y, Kido A, Imamura T, Kawate K, Tanaka Y. Increased osteogenesis with hydroxyapatite constructs combined with serially-passaged bone marrow-derived mesenchymal stem cells. ACTA ACUST UNITED AC 2012. [DOI: 10.4236/scd.2012.24018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
39
|
Secretory osteocalcin as a nondestructive osteogenic marker of tissue-engineered bone. J Orthop Sci 2011; 16:622-8. [PMID: 21725671 DOI: 10.1007/s00776-011-0121-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 06/10/2011] [Indexed: 10/18/2022]
Abstract
BACKGROUND AND PURPOSE The constructs of mesenchymal stem cells and ceramics form bone tissue after implantation. Therefore, the constructs can include cultured bone (tissue-engineered bone) as bone grafts. However, the selection of constructs, prior to implantation, with high osteogenic potential is still difficult. We used a rat model to measure the secretory osteocalcin level in culture medium to verify that monitoring osteocalcin levels enables the selection of constructs with high osteogenic potential. METHODS We prepared constructs of rat hydroxyapatite/cells and used different cell passages of P-1 and P-3 as well as different cell numbers: 1 × 10(5) and 1 × 10(6) cells/ml suspension. These constructs were cultured for 14 days under osteoinductive or nonosteoinductive conditions and implanted subcutaneously in the recipient rat. Secretory osteocalcin in the culture medium was measured using an enzyme-linked immunosorbent assay system during the culture period until day 14, and the osteocalcin content of the harvested construct at 4 weeks was also measured. RESULTS AND CONCLUSION All constructs except the hydroxyapatite/P-3 construct showed abundant bone formation by histology and both high secretory osteocalcin level in the medium and high osteocalcin content after implantation. Our study revealed that secretory osteocalcin level in vitro was related to osteocalcin content in vivo. The study clearly showed that measuring secretory osteocalcin is a nondestructive method of assessing the osteogenic potential of tissue-engineered bone. One can choose tissue-engineered bone with high osteogenic potential by integrating secretory osteocalcin measurement into the process of bone-tissue regeneration.
Collapse
|
40
|
Ring A, Tilkorn DJ, Goertz O, Langer S, Schaffran A, Awakowicz P, Hauser J. Surface modification by glow discharge gasplasma treatment improves vascularization of allogenic bone implants. J Orthop Res 2011; 29:1237-44. [PMID: 21381095 DOI: 10.1002/jor.21358] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Accepted: 12/13/2010] [Indexed: 02/04/2023]
Abstract
Sufficient induction of blood vessel ingrowth decisively influence transplant functionality. In this study, microvascular response to transplants of surface modified bone substitutes were assessed in vivo. The surface modification of allogenic bone substitutes (dehydrated human femoral head) was achieved in a double-conductive low-pressure gasplasma reactor (Ar(2) /O(2) , 13.65 MHz, 1,000 W, 5 Pa). The modified bone substitutes (n = 10) as well as untreated bone substitutes serving as controls (n = 10) were placed into the dorsal skinfold chamber of female balb/c mice (n = 10). Dynamic assessment of microcirculatory parameters was performed using intravital fluorescence microscopy during an implantation period of 10 days. The angiogenic response was found markedly accelerated in gasplasma-treated bone. Compared to untreated implants, the gasplasma-activated bone substitutes showed significantly higher microvascular density on days 5 and 10. The quantification of the microvascular diameters, red blood cell velocity, and microvascular permeability displayed stable perfusion and vascular integrity of the newly developed blood vessels throughout the 10-day observation period. The surface activation via cold low-pressure glow discharge gasplasma supports the vascular integration of allogenic bone by earlier induction of the angiogenesis.
Collapse
Affiliation(s)
- Andrej Ring
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany.
| | | | | | | | | | | | | |
Collapse
|
41
|
Low-intensity pulsed ultrasound enhances posterior spinal fusion implanted with mesenchymal stem cells-calcium phosphate composite without bone grafting. Spine (Phila Pa 1976) 2011; 36:1010-6. [PMID: 21325987 DOI: 10.1097/brs.0b013e318205c5f5] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
STUDY DESIGN Experimental study on the effect of low-intensity pulsed ultrasound (LIPUS) on rabbit spinal fusion with mesenchymal stem cell (MSC)-derived osteogenic cells and bioceramic composite. OBJECTIVE To investigate the efficacy of LIPUS in enhancing fusion rate and bone formation with porous tricalcium phosphate (TCP) bioceramic scaffold impregnated with MSCs without any bone grafts. SUMMARY OF BACKGROUND DATA The goal of spinal fusion in the corrective spinal surgery for spinal deformities is to achieve solid bony fusion between selected vertebral segments. Previous studies with bone morphogenetic proteins and genetically manipulated materials revealed significant difficulties in actual clinical application. Alternative such as LIPUS has been shown to be effective in enhancing healing of fracture and nonunion clinically. Its potential for enhancing spinal fusion warrants further in-depth study. METHODS Posterolateral intertransverse processes spinal fusion at the L5 and L6 levels were evaluated in New Zealand white rabbit model. The animals were divided into three groups with (A) TCP alone, (B) TCP with differentiated MSCs, and (C) TCP with differentiated MSCs and LIPUS treatment. At week 7 postoperation, manual palpation, peripheral quantitative computed tomography, and histomorphometric assessments were performed. RESULTS At week 7 postoperation, a statistically significant increase in clinical fusion by manual palpation was observed in group C animals treated with LIPUS (86%) in comparing with groups A (0%) and B (14%) without LIPUS. With peripheral quantitative computed tomographic analysis, the bone volume of group C fusion mass was significantly larger than the other two groups. Group C fusion also had better osteointegration length between host bone and implanted composite and more new bone formed in the TCP implants. Importantly, all the group C animals had osteochondral bridging--early stage of bony fusion histologically. Endochondral ossification was observed at the junction between the cartilaginous and osseous tissues at the intertransverse processes area. Quantitative analysis showed that the fusion mass in group C had significantly smaller gap and larger area of cartilaginous tissue between the transverse processes. CONCLUSION The present study showed that the combination of synthetic biomaterials, autologous differentiated MSCs, and LIPUS could promote clinical fusion in rabbit posterior spinal fusion model. The mechanism was likely to be mediated through better osteointegration between the host bone and implanted materials and enhanced endochondral ossification at the fusion site.
Collapse
|
42
|
Tadokoro M, Matsushima A, Kotobuki N, Hirose M, Kimura Y, Tabata Y, Hattori K, Ohgushi H. Bone morphogenetic protein-2 in biodegradable gelatin and β-tricalcium phosphate sponges enhances the in vivo bone-forming capability of bone marrow mesenchymal stem cells. J Tissue Eng Regen Med 2011; 6:253-60. [DOI: 10.1002/term.427] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Accepted: 03/13/2011] [Indexed: 11/08/2022]
|
43
|
Imamura T, Ishizuka O, Kinebuchi Y, Kurizaki Y, Nakayama T, Ishikawa M, Nishizawa O. Implantation of autologous bone-marrow-derived cells reconstructs functional urethral sphincters in rabbits. Tissue Eng Part A 2011; 17:1069-81. [PMID: 21091339 DOI: 10.1089/ten.tea.2010.0478] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The purpose of this study was to determine if implantation of autologous bone-marrow-derived cells has the potential to treat stress urinary incontinence caused by intrinsic sphincter deficiency. Bone marrow cells harvested from femurs of New Zealand White rabbits were cultured for 10 days. Seven days before implantation, the urethral sphincters located at the internal urethral orifice were cryo-injured by spraying liquid nitrogen for 15 s. The cultured autologous bone-marrow-derived cells were implanted 7 days after cryo-injury. For controls, cell-free solutions were injected. At 7 and 14 days after implantation, leak point pressures were determined and the urethral sphincters were examined by immunohistochemistry. At 7 and 14 days, the cell-implanted regions contained numerous striated and smooth muscle-like cells expressing myoglobin and smooth muscle actin, respectively. The proportions of myoglobin- and smooth muscle actin-expressing areas in both the 7- and 14-day cell-implanted regions were significantly higher than in controls. By 14 days, these differentiated cells formed contacts with similar cells, creating layered muscle structures. At that time, the leak point pressure of the cell-implanted rabbits was significantly higher than that of the controls. In conclusion, autologous bone-marrow-derived cells can reconstruct functional urethral sphincters.
Collapse
Affiliation(s)
- Tetsuya Imamura
- Department of Urology, Shinshu University School of Medicine, Matsumoto, Japan.
| | | | | | | | | | | | | |
Collapse
|
44
|
Matsumoto T, Hattori K, Matsushima A, Tadokoro M, Yagyuu T, Kodama M, Sato J, Ohgushi H. Osteogenic Potential of Mesenchymal Stem Cells on Expanded Polytetrafluoroethylene Coated with Both a Poly-Amino-Acid Urethane Copolymer and Collagen. Tissue Eng Part A 2011; 17:171-80. [DOI: 10.1089/ten.tea.2010.0077] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Tomohiro Matsumoto
- First Department of Oral and Maxillofacial Surgery, Tsurumi University, Yokohama, Kanagawa, Japan
- Division of Oral and Maxillofacial Implantology, Tsurumi University, Yokohama, Kanagawa, Japan
- Health Research Institute, National Institute of Advanced Industrial Science and Technology, Amagasaki, Hyogo, Japan
| | - Koji Hattori
- Health Research Institute, National Institute of Advanced Industrial Science and Technology, Amagasaki, Hyogo, Japan
| | - Asako Matsushima
- Health Research Institute, National Institute of Advanced Industrial Science and Technology, Amagasaki, Hyogo, Japan
| | - Mika Tadokoro
- Health Research Institute, National Institute of Advanced Industrial Science and Technology, Amagasaki, Hyogo, Japan
| | - Takahiro Yagyuu
- Health Research Institute, National Institute of Advanced Industrial Science and Technology, Amagasaki, Hyogo, Japan
- Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Nara, Japan
| | | | - Junichi Sato
- First Department of Oral and Maxillofacial Surgery, Tsurumi University, Yokohama, Kanagawa, Japan
- Division of Oral and Maxillofacial Implantology, Tsurumi University, Yokohama, Kanagawa, Japan
| | - Hajime Ohgushi
- Health Research Institute, National Institute of Advanced Industrial Science and Technology, Amagasaki, Hyogo, Japan
| |
Collapse
|
45
|
Bernhardt A, Lode A, Peters F, Gelinsky M. Novel ceramic bone replacement material Osbone® in a comparative in vitro study with osteoblasts. Clin Oral Implants Res 2010; 22:651-7. [DOI: 10.1111/j.1600-0501.2010.02015.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
46
|
Lu Z, Zreiqat H. The Osteoconductivity of Biomaterials Is Regulated by Bone Morphogenetic Protein 2 Autocrine Loop Involving α2β1 Integrin and Mitogen-Activated Protein Kinase/Extracellular Related Kinase Signaling Pathways. Tissue Eng Part A 2010; 16:3075-84. [DOI: 10.1089/ten.tea.2010.0204] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- ZuFu Lu
- Biomaterials and Tissue Engineering Research Unit, School of AMME, The University of Sydney, Sydney, Australia
| | - Hala Zreiqat
- Biomaterials and Tissue Engineering Research Unit, School of AMME, The University of Sydney, Sydney, Australia
| |
Collapse
|
47
|
|
48
|
Kawase T, Okuda K, Kogami H, Nakayama H, Nagata M, Sato T, Wolff LF, Yoshie H. Human periosteum-derived cells combined with superporous hydroxyapatite blocks used as an osteogenic bone substitute for periodontal regenerative therapy: an animal implantation study using nude mice. J Periodontol 2010; 81:420-7. [PMID: 20192869 DOI: 10.1902/jop.2009.090523] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND A superporous (85%) hydroxyapatite (HA) block was recently developed to improve osteoconductivity, but it was often not clinically successful when used to treat periodontal osseous defects. The primary purpose of this study is to develop a clinically applicable tissue-engineered bone substitute using this HA block and human alveolar periosteum-derived cells. METHODS Commercially available superporous HA blocks were acid treated and subjected to a three-dimensional (3D) culture for periosteal cell cultivation. Cells in the pore regions of the treated HA block were observed on the fracture surface by scanning electron microscopy. After osteogenic induction, the cell-HA complexes were implanted subcutaneously in nude mice. Osteoid formation was histologically evaluated. RESULTS Acid treatment enlarged the interconnections among pores, resulting in the deep penetration of periosteal cells. Under these conditions, cells were maintained for >2 weeks without appreciable cell death in the deep pore regions of the HA block. The cell-HA complexes that received in vitro osteogenic induction formed osteoids in pore regions of the treated HA blocks in vivo. In contrast, most pore regions in the non-pretreated, cell-free HA blocks that were evaluated in vivo remained cell free. CONCLUSIONS Our findings suggest that an acid-treated HA block could function as a better scaffold for the 3D high-density culture of human periosteal cells in vitro, and this cell-HA complex had significant osteogenic potential at the site of implantation in vivo. Compared with the cell-free HA block, our cell-HA complex using periosteal cells, which are the most accessible for clinical periodontists, showed promising results as a bone substitute in periodontal regenerative therapy.
Collapse
Affiliation(s)
- Tomoyuki Kawase
- Division of Oral Bioengineering, Department of Tissue Regeneration and Reconstitution, Institute of Medicine and Dentistry, Niigata University, Niigata, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Sharma AK, Hota PV, Matoka DJ, Fuller NJ, Jandali D, Thaker H, Ameer GA, Cheng EY. Urinary bladder smooth muscle regeneration utilizing bone marrow derived mesenchymal stem cell seeded elastomeric poly(1,8-octanediol-co-citrate) based thin films. Biomaterials 2010; 31:6207-17. [PMID: 20488535 DOI: 10.1016/j.biomaterials.2010.04.054] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Accepted: 04/22/2010] [Indexed: 02/08/2023]
Abstract
Bladder regeneration studies have yielded inconclusive results possibly due to the use of unfavorable cells and primitive scaffold design. We hypothesized that human mesenchymal stem cells seeded onto poly(1,8-octanediol-co-citrate) elastomeric thin films would provide a suitable milieu for partial bladder regeneration. POCfs were created by reacting citric acid with 1,8-octanediol and seeded on opposing faces with human MSCs and urothelial cells; normal bladder smooth muscle cells and UCs, or unseeded POCfs. Partial cystectomized nude rats were augmented with the aforementioned POCfs, enveloped with omentum and sacrificed at 4 and 10 weeks. Isolated bladders were subjected to Trichrome and anti-human gamma-tubulin, calponin, caldesmon, smooth muscle gamma-actin, and elastin stainings. Mechanical testing of POCfs revealed a Young's modulus of 138 kPa with elongation 137% its initial length without permanent deformation demonstrating its high uniaxial elastic potential. Trichrome and immunofluorescent staining of MSC/UC POCf augmented bladders exhibited typical bladder architecture with muscle bundle formation and the expression and retention of bladder smooth muscle contractile proteins of human derivation. Quantitative morphometry of MSC/UC samples revealed muscle/collagen ratios approximately 1.75x greater than SMC/UC controls at 10 weeks. Data demonstrate MSC seeded POCfs support partial regeneration of bladder tissue in vivo.
Collapse
Affiliation(s)
- Arun K Sharma
- Children's Memorial Hospital Chicago, Division of Pediatric Urology, 2300 Children's Plaza, Chicago, IL 60614, USA.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Bahar H, Yaffe A, Boskey A, Binderman I. Influence of bone-derived matrices on generation of bone in an ectopic rat model. J Orthop Res 2010; 28:664-70. [PMID: 19918912 DOI: 10.1002/jor.21017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Most bone regeneration experimental models that test bone-derived matrices take place in conjunction with the native bone. Here, we compared the relative effectiveness of bone matrix components on bone-marrow-directed osteogenesis in an ectopic model. Cortical bone cylinders consisted of diaphysis of DA rat femurs. They were either demineralized (DBM), deproteinized (HABM), or nontreated (MBM). Fresh bone marrow was placed into cylinders and implanted at subcutaneous thoracic sites of 2-month-old DA rats. At designated times the cylinders were surgically removed from the animals. Microradiographs of DBM and histology of DBM and MBM cylinders demonstrated progressive increase in mineralized bone volume and its trabecular configuration. Bone filled the inner volume of DBM and MBM cylinders within 4 weeks, while in HABM cylinders mostly granulation tissue developed. In the DBM cylinders cartilage deposited within 10 days, while in the MBM cylinders bone was directly deposited. As early as day 3 after marrow transplantation, marrow cells interacting with DBM increased significantly the genes that express the cartilage and the bone phenotype. In conclusion, organic components of bone are needed for marrow-directed osteogenesis.
Collapse
Affiliation(s)
- H Bahar
- Department of Oral Biology, The Maurice and Gabriela Goldschleger, School of Dental Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | |
Collapse
|