1
|
Totoki Y, Mutsuzaki H, Yanagisawa Y, Sogo Y, Yasunaga M, Noguchi H, Matsumoto Y, Koda M, Ito A, Yamazaki M. Do Stainless-Steel Pins Coated with Fibroblast Growth Factor-Calcium Phosphatase Composite Layers Have Anti-Infective Effects? MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1419. [PMID: 39336460 PMCID: PMC11434512 DOI: 10.3390/medicina60091419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024]
Abstract
Background: The most problematic complication of external fixation is infection at the pin insertion site. Technology that improves the adhesion of the external fixation pin to the skin, subcutaneous tissue, and bone may prevent infection at the pin site. The purpose of this study is to formulate a calcium phosphate-fibroblast growth factor (Cp-FGF) coating on a stainless-steel external fixation pin and to verify its effectiveness in reducing infection at the pin site and its possible influence on bone fixation in animal experiments. Methods: We compared stainless-steel screws without coating (SS group; n = 32), those with a calcium phosphate coating (Cp group; n = 30), those with a Cp-FGF coating (FGF group; n = 32), and those with a Cp-FGF coating having enhanced biological activity (FGF+ group; n = 32) in male Japanese white domesticated rabbits. Screws were inserted percutaneously into the bilateral proximal tibial diaphysis of the rabbits and implanted for 4 weeks. Screws and periscrew tissue were observed postoperatively for qualitatively assessing infection. Results: Infection assessment by gross findings after 4 weeks (at screw removal) showed no significant differences between the groups. Histopathological evaluation of soft tissue infection and bone tissue infection showed no significant differences between the groups for either soft tissue or bone tissue. Since neither the FGF+ group nor the FGF group showed anti-infective effects, the biological activity of FGF is not the only determining factor. We compared SEM, XRD, coating detaching test, sustained release test, and bioassay to examine physicochemical properties among the coatings but found no sufficient differences. Conclusions: It is suggested that improving the tissue adhesion to and/or biocompatibility of pins is also important to improve the in vivo performance of Cp-FGF-coated external fixation pins.
Collapse
Affiliation(s)
- Yasukazu Totoki
- Department of Orthopaedic Surgery, University of Tsukuba, Tsukuba 305-8571, Ibaraki, Japan; (Y.T.); (Y.Y.); (H.N.); (Y.M.); (M.K.); (M.Y.)
| | - Hirotaka Mutsuzaki
- Center for Medical Science, Ibaraki Prefectural University of Health Sciences, Ami 300-0394, Ibaraki, Japan
- Department of Orthopedic Surgery, Ibaraki Prefectural University of Health Sciences Hospital, Ami 300-0331, Ibaraki, Japan
| | - Yohei Yanagisawa
- Department of Orthopaedic Surgery, University of Tsukuba, Tsukuba 305-8571, Ibaraki, Japan; (Y.T.); (Y.Y.); (H.N.); (Y.M.); (M.K.); (M.Y.)
| | - Yu Sogo
- National Institute of Advanced Industrial Science and Technology (AIST), Health and Medical Research Institute, Tsukuba 305-8566, Ibaraki, Japan; (Y.S.); (M.Y.); (A.I.)
| | - Mayu Yasunaga
- National Institute of Advanced Industrial Science and Technology (AIST), Health and Medical Research Institute, Tsukuba 305-8566, Ibaraki, Japan; (Y.S.); (M.Y.); (A.I.)
| | - Hiroshi Noguchi
- Department of Orthopaedic Surgery, University of Tsukuba, Tsukuba 305-8571, Ibaraki, Japan; (Y.T.); (Y.Y.); (H.N.); (Y.M.); (M.K.); (M.Y.)
| | - Yukei Matsumoto
- Department of Orthopaedic Surgery, University of Tsukuba, Tsukuba 305-8571, Ibaraki, Japan; (Y.T.); (Y.Y.); (H.N.); (Y.M.); (M.K.); (M.Y.)
| | - Masao Koda
- Department of Orthopaedic Surgery, University of Tsukuba, Tsukuba 305-8571, Ibaraki, Japan; (Y.T.); (Y.Y.); (H.N.); (Y.M.); (M.K.); (M.Y.)
| | - Atsuo Ito
- National Institute of Advanced Industrial Science and Technology (AIST), Health and Medical Research Institute, Tsukuba 305-8566, Ibaraki, Japan; (Y.S.); (M.Y.); (A.I.)
| | - Masashi Yamazaki
- Department of Orthopaedic Surgery, University of Tsukuba, Tsukuba 305-8571, Ibaraki, Japan; (Y.T.); (Y.Y.); (H.N.); (Y.M.); (M.K.); (M.Y.)
- Department of Orthopaedic Surgery, Ichihara Hospital, Tsukuba 300-3295, Ibaraki, Japan
| |
Collapse
|
2
|
An YZ, Strauss FJ, Park JY, Shen YQ, Thoma DS, Lee JS. Membrane fixation enhances guided bone regeneration in standardized calvarial defects: A pre-clinical study. J Clin Periodontol 2022; 49:177-187. [PMID: 34866208 DOI: 10.1111/jcpe.13583] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/09/2021] [Accepted: 11/23/2021] [Indexed: 12/21/2022]
Abstract
AIM To determine whether collagen membrane (CM) fixation enhances guided bone regeneration in standardized defects. MATERIALS AND METHODS Four 8-mm-diameter defects were surgically made in eight rabbit calvaria, and randomly allocated into four groups: control (empty), unfixed-CM, fixed-CM, and unfixed-CM with bone graft (BG + CM) (positive control). After 1- and 4-week healing periods, the animals were sacrificed and quantitative reverse transcription polymerase chain reaction, micro-computed tomography, and histological outcomes were assessed. RESULTS At week 1, the expression levels of BMP-2, FGF-2, VEGF, and osteocalcin were significantly higher in the fixed-CM group than in the unfixed-CM and control groups (p < .05). Conversely, cathepsin-K was significantly expressed in the unfixed-CM group. No significant differences in expression markers were observed between the fixed-CM and BG + CM groups (p > .05). At week 4, new bone formation was significantly higher in the fixed-CM group than the unfixed-CM and control groups (p < .05), but similar to the BG + CM group (p > .05). CONCLUSIONS CM fixation enhances the expression of osteogenic factors similar to BG + CM, leading to significantly more new bone formation. This suggests that the osteogenic potential is greater when membranes are fixed, thereby limiting the necessity of membrane-supporting materials to enhance bone formation.
Collapse
Affiliation(s)
- Yin-Zhe An
- Department of Periodontology, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Franz-Josef Strauss
- Clinic of Reconstructive Dentistry, University of Zurich, Zurich, Switzerland
- Department of Conservative Dentistry, Faculty of Dentistry, University of Chile, Santiago, Chile
- Department of Oral Biology, Medical University of Vienna, Vienna, Austria
| | - Jin-Young Park
- Department of Periodontology, Research Institute for Periodontal Regeneration, College of Dentistry, Yonsei University, Seoul, Republic of Korea
| | - Yu Qin Shen
- Department of Periodontology, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Daniel Stefan Thoma
- Clinic of Reconstructive Dentistry, University of Zurich, Zurich, Switzerland
| | - Jung-Seok Lee
- Department of Oral Biology, Medical University of Vienna, Vienna, Austria
- Department of Periodontology, Research Institute for Periodontal Regeneration, College of Dentistry, Yonsei University, Seoul, Republic of Korea
- Innovation Research and Support Center for Dental Science, Yonsei University Dental Hospital, Seoul, Republic of Korea
| |
Collapse
|
3
|
Proposal for a New Bioactive Kinetic Screw in an Implant, Using a Numerical Model. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12020779] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
A new biomechanism, Bioactive Kinetic Screw (BKS) for screws and bone implants created by the first author, is presented using a bone dental implant screw, in which the bone particles, blood, cells, and protein molecules removed during bone drilling are used as a homogeneous autogenous transplant in the same implant site, aiming to obtain primary and secondary bone stability, simplifying the surgical procedure, and improving the healing process. The new BKS is based on complex geometry. In this work, we describe the growth factor (GF) delivery properties and the in situ optimization of the use of the GF in the fixation of bone screws through a dental implant. To describe the drilling process, an explicit dynamic numerical model was created, where the results show a significant impact of the drilling process on the bone material. The simulation demonstrates that the space occupied by the screw causes stress and deformation in the bone during the perforation and removal of the particulate bone, resulting in the accumulation of material removed within the implant screw, filling the limit hole of the drill grooves present on the new BKS.
Collapse
|
4
|
Kuhn LT, Peng T, Gronowicz G, Hurley MM. Endogenous FGF-2 levels impact FGF-2/BMP-2 growth factor delivery dosing in aged murine calvarial bone defects. J Biomed Mater Res A 2021; 109:2545-2555. [PMID: 34173706 PMCID: PMC9943554 DOI: 10.1002/jbm.a.37249] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/20/2021] [Accepted: 06/11/2021] [Indexed: 12/12/2022]
Abstract
Bone repair in elderly mice has been shown to be improved or negatively impacted by supplementing the highly osteogenic bone morphogenetic protein-2 (BMP-2) with fibroblast growth factor-2 (FGF-2). To better predict the outcome of FGF-2 supplementation, we investigated whether endogenous levels of FGF-2 play a role in optimal dosing of FGF-2 for augmenting BMP-2 activity in elderly mice. In vivo calvarial bone defect studies in Fgf2 knockout mice with wildtype controls were conducted with the growth factors delivered in a highly localized manner from a biomimetic calcium phosphate/polyelectrolyte multilayer coating applied to a bone graft substitute. Endogenous FGF-2 levels were measured in old mice versus young and found to decrease with age. Optimal dosing for improving bone defect repair correlated with levels of endogenous FGF-2, with a larger dose of FGF-2 required to have a positive effect on bone healing in the Fgf2 knockout mice. The same dose in wildtype old mice, with higher levels of FGF-2, promoted chondrogenesis and increased osteoclast activity. The results suggest a personalized medicine approach, based on a knowledge of endogenous levels of FGF-2, should guide FGF-2 supplementation in order to avoid provoking excessive bone resorption and cartilage formation, both of which inhibited calvarial bone repair.
Collapse
Affiliation(s)
- Liisa T Kuhn
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Tao Peng
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Gloria Gronowicz
- Department of Surgery, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Marja M Hurley
- Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| |
Collapse
|
5
|
Kauffmann P, Raschke D, Tröltzsch M, Santander P, Brockmeyer P, Schliephake H. The use of rhBMP2 for augmentation of established horizontal/vertical defects may require additional use of rhVEGF to achieve significant bone regeneration: An in vivo experimental study. Clin Oral Implants Res 2021; 32:1228-1240. [PMID: 34352150 DOI: 10.1111/clr.13820] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 02/08/2021] [Accepted: 04/07/2021] [Indexed: 11/28/2022]
Abstract
AIM To test the hypothesis that the use of rhBMP2 in established defects requires additional growth factors such as rhVEGF to accomplish effective bone repair. MATERIALS AND METHODS Horizontal/vertical defects of 2 cm length and 1 cm height were created bilaterally in the alveolar crest of the maxillae of 18 minipigs together with the extraction of all premolar teeth and one molar tooth on both sides. After 3 months of healing, defects were augmented with 0.5 g particulate PDLLA/CaCO3 composite loaded with 400 µg rhBMP2/50 µg rhVEGF165 on one side and 800 µg rhBMP2 on the other in 12 test animals, whereas defects in six control animals were sham operated and left unfilled on one side and augmented with blank carriers on the other. After 4 and 13 weeks, the animals were evaluated each for area of new bone formation (mm²) and bone density (area %). RESULTS Augmentations with carriers loaded with 800 g µrhBMP2 failed to induce significantly more bone than in the augmentations with unloaded carrier after 4 and 13 weeks (p = .1000, p = .381). Augmentations with carriers loaded with 400 µg rhBMP2 and 50 µg erhVEGF165 resulted in significantly increased bone formation after 13 weeks (p = .024) compared to blank carriers. Soft tissue in augmentations with combined rhBMP2/rhVEGF165 loading exhibited numerous microvessels compared to soft tissue in augmentations with rhBMP2. CONCLUSIONS It is concluded that effective bone regeneration in augmentations of established alveolar ridge defects may require the application of rhVEGF additionally to rhBMP2.
Collapse
Affiliation(s)
- Philipp Kauffmann
- Department for Oral & Maxillofacial Surgery, Universitätsmedizin Goettingen, Goettingen, Germany
| | - David Raschke
- Department for Oral & Maxillofacial Surgery, Universitätsmedizin Goettingen, Goettingen, Germany
| | - Markus Tröltzsch
- Private Office Ansbach, Germany & Department for Oral & Maxillofacial Surgery, Universitätsmedizin Goettingen, Goettingen, Germany
| | - Petra Santander
- Department of Orthodontics, Universitätsmedizin Göttingen, Goettingen, Germany
| | - Phillip Brockmeyer
- Department for Oral & Maxillofacial Surgery, Universitätsmedizin Goettingen, Goettingen, Germany
| | - Henning Schliephake
- Department for Oral & Maxillofacial Surgery, Universitätsmedizin Goettingen, Goettingen, Germany
| |
Collapse
|
6
|
Zhao Z, Ma S, Wu C, Li X, Ma X, Hu H, Wu J, Wang Y, Liu Z. Chimeric Peptides Quickly Modify the Surface of Personalized 3D Printing Titanium Implants to Promote Osseointegration. ACS APPLIED MATERIALS & INTERFACES 2021; 13:33981-33994. [PMID: 34260195 DOI: 10.1021/acsami.1c11207] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Titanium (Ti) and titanium alloys have been widely used in the field of biomedicine. However, the unmatched biomechanics and poor bioactivities of conventional Ti implants usually lead to insufficient osseointegration. To tackle these challenges, it is critical to develop a novel Ti implant that meets the bioadaptive requirements for load-bearing critical bone defects. Notably, three-dimensional (3D)-printed Ti implants mimic the microstructure and mechanical properties of natural bones. Additionally, eco-friendly techniques based on inorganic-binding peptides have been applied to modify Ti surfaces. Herein, in our study, Ti surfaces were modified to reinforce osseointegration using chimeric peptides constructed by connecting W9, RP1P, and minTBP-1 directly or via (GP)4, respectively. PR1P is derived from the extracellular VEGF-binding domain of prominin-1, which increases the expression of VEGF and promotes the binding of VEGF to endothelial cells, thereby accelerating angiogenesis. W9 induces osteoblast differentiation in bone marrow mesenchymal stem cells and human mesenchymal stem cells to promote bone formation. Overall, chimeric peptides promote osseointegration by promoting angiogenesis and osteogenesis. Additionally, chimeric peptides with P3&4 were more effective than those with P1&2 in improving osseointegration, which might be ascribed to the capacity of P3&4 to provide a greater range for chimeric peptides to express their activity. This work successfully used chimeric peptides to modify 3D-Ti implant surfaces to improve osseointegration on the implant-bone surface.
Collapse
Affiliation(s)
- Zhezhe Zhao
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Shiqing Ma
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Chenxuan Wu
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Xuewen Li
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Xinying Ma
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Han Hu
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Jie Wu
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Yonglan Wang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Zihao Liu
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, People's Republic of China
| |
Collapse
|
7
|
Burn-induced heterotopic ossification from incidence to therapy: key signaling pathways underlying ectopic bone formation. Cell Mol Biol Lett 2021; 26:34. [PMID: 34315404 PMCID: PMC8313878 DOI: 10.1186/s11658-021-00277-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/20/2021] [Indexed: 01/02/2023] Open
Abstract
Burn injury is one of the potential causes of heterotopic ossification (HO), which is a rare but debilitating condition. The incidence ranges from 3.5 to 5.6 depending on body area. Burns that cover a larger percentage of the total body surface area (TBSA), require skin graft surgeries, or necessitate pulmonary intensive care are well-researched risk factors for HO. Since burns initiate such complex pathophysiological processes with a variety of molecular signal changes, it is essential to focus on HO in the specific context of burn injury to define best practices for its treatment. There are numerous key players in the pathways of burn-induced HO, including neutrophils, monocytes, transforming growth factor-β1-expressing macrophages and the adaptive immune system. The increased inflammation associated with burn injuries is also associated with pathway activation. Neurological and calcium-related contributions are also known. Endothelial-to-mesenchymal transition (EMT) and vascularization are known to play key roles in burn-induced HO, with hypoxia-inducible factor-1 (HIF-1) and vascular endothelial growth factor (VEGF) as potential initiators. Currently, non-steroidal anti-inflammatory drugs (NSAIDs) and radiotherapy are effective prophylaxes for HO. Limited joint motion, ankylosis and intolerable pain caused by burn-induced HO can be effectively tackled via surgery. Effective biomarkers for monitoring burn-induced HO occurrence and bio-prophylactic and bio-therapeutic strategies should be actively developed in the future.
Collapse
|
8
|
Bjelić D, Finšgar M. The Role of Growth Factors in Bioactive Coatings. Pharmaceutics 2021; 13:1083. [PMID: 34371775 PMCID: PMC8309025 DOI: 10.3390/pharmaceutics13071083] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 12/26/2022] Open
Abstract
With increasing obesity and an ageing population, health complications are also on the rise, such as the need to replace a joint with an artificial one. In both humans and animals, the integration of the implant is crucial, and bioactive coatings play an important role in bone tissue engineering. Since bone tissue engineering is about designing an implant that maximally mimics natural bone and is accepted by the tissue, the search for optimal materials and therapeutic agents and their concentrations is increasing. The incorporation of growth factors (GFs) in a bioactive coating represents a novel approach in bone tissue engineering, in which osteoinduction is enhanced in order to create the optimal conditions for the bone healing process, which crucially affects implant fixation. For the application of GFs in coatings and their implementation in clinical practice, factors such as the choice of one or more GFs, their concentration, the coating material, the method of incorporation, and the implant material must be considered to achieve the desired controlled release. Therefore, the avoidance of revision surgery also depends on the success of the design of the most appropriate bioactive coating. This overview considers the integration of the most common GFs that have been investigated in in vitro and in vivo studies, as well as in human clinical trials, with the aim of applying them in bioactive coatings. An overview of the main therapeutic agents that can stimulate cells to express the GFs necessary for bone tissue development is also provided. The main objective is to present the advantages and disadvantages of the GFs that have shown promise for inclusion in bioactive coatings according to the results of numerous studies.
Collapse
Affiliation(s)
| | - Matjaž Finšgar
- Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia;
| |
Collapse
|
9
|
Walsh DP, Raftery RM, Murphy R, Chen G, Heise A, O'Brien FJ, Cryan SA. Gene activated scaffolds incorporating star-shaped polypeptide-pDNA nanomedicines accelerate bone tissue regeneration in vivo. Biomater Sci 2021; 9:4984-4999. [PMID: 34086016 DOI: 10.1039/d1bm00094b] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Increasingly, tissue engineering strategies such as the use of biomaterial scaffolds augmented with specific biological cues are being investigated to accelerate the regenerative process. For example, significant clinical challenges still exist in efficiently healing large bone defects which are above a critical size. Herein, we describe a cell-free, biocompatible and bioresorbable scaffold incorporating a novel star-polypeptide biomaterial as a gene vector. This gene-loaded scaffold can accelerate bone tissue repair in vivo in comparison to a scaffold alone at just four weeks post implantation in a critical sized bone defect. This is achieved via the in situ transfection of autologous host cells which migrate into the implanted collagen-based scaffold via gene-loaded, star-shaped poly(l-lysine) polypeptides (star-PLLs). In vitro, we demonstrate that star-PLL nanomaterials designed with 64 short poly(l-lysine) arms can be used to functionalise a range of collagen based scaffolds with a dual therapeutic cargo (pDual) of the bone-morphogenetic protein-2 plasmid (pBMP-2) and vascular endothelial growth factor plasmid (pVEGF). The versatility of this polymeric vector is highlighted in its ability to transfect Mesenchymal Stem Cells (MSCs) with both osteogenic and angiogenic transgenes in a 3D environment from a range of scaffolds with various macromolecular compositions. In vivo, we demonstrate that a bone-mimetic, collagen-hydroxyapatite scaffold functionalized with star-PLLs containing either 32- or 64- poly(l-lysine) arms can be used to successfully deliver this pDual cargo to autologous host cells. At the very early timepoint of just 4 weeks, we demonstrate the 64-star-PLL-pDual functionalised scaffold as a particularly efficient platform to accelerate bone tissue regeneration, with a 6-fold increase in new bone formation compared to a scaffold alone. Overall, this article describes for the first time the incorporation of novel star-polypeptide biomaterials carrying two therapeutic genes into a cell free scaffold which supports accelerated bone tissue formation in vivo.
Collapse
Affiliation(s)
- David P Walsh
- Drug Delivery & Advanced Materials Team, School of Pharmacy & Biomolecular Sciences, RCSI, Dublin, Ireland and Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, RCSI, Dublin, Ireland and Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin, Ireland and SFI Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Ireland
| | - Rosanne M Raftery
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, RCSI, Dublin, Ireland and Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin, Ireland and SFI Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Ireland
| | | | - Gang Chen
- Centre for the Study of Neurological Disorders, Microsurgical Research and Training Facility (MRTF), RCSI, Dublin, Ireland
| | - Andreas Heise
- SFI Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Ireland and Department of Chemistry, RCSI, Dublin, Ireland and SFI Centre for Research in Medical Devices (CURAM), RCSI, Dublin and National University of Ireland, Galway, Ireland
| | - Fergal J O'Brien
- Drug Delivery & Advanced Materials Team, School of Pharmacy & Biomolecular Sciences, RCSI, Dublin, Ireland and Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, RCSI, Dublin, Ireland and Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin, Ireland and SFI Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Ireland and SFI Centre for Research in Medical Devices (CURAM), RCSI, Dublin and National University of Ireland, Galway, Ireland
| | - Sally-Ann Cryan
- Drug Delivery & Advanced Materials Team, School of Pharmacy & Biomolecular Sciences, RCSI, Dublin, Ireland and Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, RCSI, Dublin, Ireland and Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin, Ireland and SFI Centre for Research in Medical Devices (CURAM), RCSI, Dublin and National University of Ireland, Galway, Ireland
| |
Collapse
|
10
|
Zhu D, Ansari AR, Xiao K, Wang W, Wang L, Qiu W, Zheng X, Song H, Liu H, Zhong J, Peng K. Boron Supplementation Promotes Osteogenesis of Tibia by Regulating the Bone Morphogenetic Protein-2 Expression in African Ostrich Chicks. Biol Trace Elem Res 2021; 199:1544-1555. [PMID: 32676937 DOI: 10.1007/s12011-020-02258-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/17/2020] [Indexed: 12/24/2022]
Abstract
The present study aimed to explore the effects of supplemental boron on osteogenesis of tibia and to investigate the possible relationship between additional boron and the expression of bone morphogenetic protein-2 (BMP-2) in tibia of ostrich chicks. Therefore, forty-eight African ostrich chicks (15 days old) were supplemented with 0 mg/L, 40 mg/L, 80 mg/L, 160 mg/L, 320 mg/L, and 640 mg/L of boron in drinking water for 75 days. The paraffin sections of tibia used to measure histomorphometric parameters by hematoxylin and eosin (HE) staining, Masson's staining, and immunohistochemistry (IHC). Enzyme-linked immunosorbent assay was performed to assess the level of BMP-2, osteocalcin (BGP), glucocorticoids (GCs), osteoprotegerin (OPG), and receptor activator of nuclear factor kappa-B ligand (RANKL) in serum. TUNEL (terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling) technique was performed to detect the cell apoptosis. The results indicated that low dose of supplemental boron (40 mg/L-160 mg/L) in drinking water promotes bone development by increasing the mature ossein. The expression of BMP2 on 45 days was higher than 90 days. Serum level of BMP-2, BGP, and GCs changed significantly in groups with low dosage of boron, and OPG/RANKL ratio was upregulated from 0 to 160 mg/L. Cell apoptosis was least in 40 mg/L and 160 mg/L groups. Taken together, low dose of boron supplemented in drinking water could promote osteogenesis and growth and development of tibia by regulating the expression and secretion of BMP-2 and providing a dynamically balanced environment for tibia growth, development, and reconstruction by regulating the concentrations of BGP, GCs, and OPG/RANKL ratio in serum.
Collapse
Affiliation(s)
- Daiyun Zhu
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Abdur Rahman Ansari
- Section of Anatomy and Histology, Department of Basic Sciences, College of Veterinary and Animal Sciences (CVAS) Jhang, University of Veterinary and Animal Sciences (UVAS), Lahore, Pakistan
| | - Ke Xiao
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Wei Wang
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Lei Wang
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Weiwei Qiu
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Xinting Zheng
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Hui Song
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Huazhen Liu
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Juming Zhong
- College of Veterinary Medicine, Auburn University, Auburn, AL, 36849, USA
| | - Kemei Peng
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
| |
Collapse
|
11
|
BMP-2 and VEGF-A modRNAs in collagen scaffold synergistically drive bone repair through osteogenic and angiogenic pathways. Commun Biol 2021; 4:82. [PMID: 33469143 PMCID: PMC7815925 DOI: 10.1038/s42003-020-01606-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
Bone has a remarkable potential for self-healing and repair, yet several injury types are non-healing even after surgical or non-surgical treatment. Regenerative therapies that induce bone repair or improve the rate of recovery are being intensely investigated. Here, we probed the potential of bone marrow stem cells (BMSCs) engineered with chemically modified mRNAs (modRNA) encoding the hBMP-2 and VEGF-A gene to therapeutically heal bone. Induction of osteogenesis from modRNA-treated BMSCs was confirmed by expression profiles of osteogenic related markers and the presence of mineralization deposits. To test for therapeutic efficacy, a collagen scaffold inoculated with modRNA-treated BMSCs was explored in an in vivo skull defect model. We show that hBMP-2 and VEGF-A modRNAs synergistically drive osteogenic and angiogenic programs resulting in superior healing properties. This study exploits chemically modified mRNAs, together with biomaterials, as a potential approach for the clinical treatment of bone injury and defects.
Collapse
|
12
|
Gromolak S, Krawczenko A, Antończyk A, Buczak K, Kiełbowicz Z, Klimczak A. Biological Characteristics and Osteogenic Differentiation of Ovine Bone Marrow Derived Mesenchymal Stem Cells Stimulated with FGF-2 and BMP-2. Int J Mol Sci 2020; 21:E9726. [PMID: 33419255 PMCID: PMC7766718 DOI: 10.3390/ijms21249726] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/08/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022] Open
Abstract
Cell-based therapies using mesenchymal stem cells (MSCs) are a promising tool in bone tissue engineering. Bone regeneration with MSCs involves a series of molecular processes leading to the activation of the osteoinductive cascade supported by bioactive factors, including fibroblast growth factor-2 (FGF-2) and bone morphogenetic protein-2 (BMP-2). In this study, we examined the biological characteristics and osteogenic differentiation potential of sheep bone marrow MSCs (BM-MSCs) treated with 20 ng/mL of FGF-2 and 100 ng/mL BMP-2 in vitro. The biological properties of osteogenic-induced BM-MSCs were investigated by assessing their morphology, proliferation, phenotype, and cytokine secretory profile. The osteogenic differentiation was characterized by Alizarin Red S staining, immunofluorescent staining of osteocalcin and collagen type I, and expression levels of genetic markers of osteogenesis. The results demonstrated that BM-MSCs treated with FGF-2 and BMP-2 maintained their primary MSC properties and improved their osteogenic differentiation capacity, as confirmed by increased expression of osteocalcin and collagen type I and upregulation of osteogenic-related gene markers BMP-2, Runx2, osterix, collagen type I, osteocalcin, and osteopontin. Furthermore, sheep BM-MSCs produced a variety of bioactive factors involved in osteogenesis, and supplementation of the culture medium with FGF-2 and BMP-2 affected the secretome profile of the cells. The results suggest that sheep osteogenic-induced BM-MSCs may be used as a cellular therapy to study bone repair in the preclinical large animal model.
Collapse
Affiliation(s)
- Sandra Gromolak
- Laboratory of Biology of Stem and Neoplastic Cells, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114 Wroclaw, Poland; (S.G.); (A.K.)
| | - Agnieszka Krawczenko
- Laboratory of Biology of Stem and Neoplastic Cells, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114 Wroclaw, Poland; (S.G.); (A.K.)
| | - Agnieszka Antończyk
- Department of Surgery, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, pl. Grunwaldzki 51, 50-366 Wroclaw, Poland; (A.A.); (K.B.); (Z.K.)
| | - Krzysztof Buczak
- Department of Surgery, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, pl. Grunwaldzki 51, 50-366 Wroclaw, Poland; (A.A.); (K.B.); (Z.K.)
| | - Zdzisław Kiełbowicz
- Department of Surgery, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, pl. Grunwaldzki 51, 50-366 Wroclaw, Poland; (A.A.); (K.B.); (Z.K.)
| | - Aleksandra Klimczak
- Laboratory of Biology of Stem and Neoplastic Cells, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114 Wroclaw, Poland; (S.G.); (A.K.)
| |
Collapse
|
13
|
Dreyer CH, Kjaergaard K, Ding M, Qin L. Vascular endothelial growth factor for in vivo bone formation: A systematic review. J Orthop Translat 2020; 24:46-57. [PMID: 32642428 PMCID: PMC7334443 DOI: 10.1016/j.jot.2020.05.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/29/2020] [Accepted: 05/20/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND To achieve optimal bone formation one of the most influential parameters has been mentioned to be adequate blood supply. Vascular endothelial growth factor (VEGF) is hereby of particular interest in bone regeneration, because of its primary ability to induce neovascularization and chemokine affection for endothelial cells (EC), and is considered to be the main regulator of vascular formation. However, the growth factor has yet to be implemented in a clinical setting in orthopaedic intervention surgery. We hypothesised that the development of VEGF in vivo for bone formation in the last decade had progressed towards clinical application since the latest systematic review from 2008. OBJECTIVE This systematic review recapped the last 13 years of in vivo bone regeneration using vascular endothelial growth factor (VEGF). METHOD A total of 1374 articles were identified using the PubMed search string (vegf or "vascular endothelial growth factor") and (osteogen∗ or "bone formation" or "bone regeneration"). By 3 selection phases 24 published articles were included by the criteria of being in vivo, using only VEGF for bone formation, published after 2007 and written in English. Articles in vitro, written in different languages than English and older than 2007 was excluded. The most recent systematic review on this subject was published in 2008, with the latest included study from 01 to 11-2007. All included studies were classified based on animal, type of defect, scaffold, control group, type of VEGF, release rate, dosage of VEGF, time of evaluation and results. Each study was evaluated for risk of bias by modified CAMARADES quality assessment for the use in experimental animal studies. The score was calculated by peer review journal publication, use of control group, randomisation of groups, justified VEGF dosage, blinding of results, details on animal model, sample size calculation, comply with ethics and no conflict of interest. RESULTS No clinical trials or human application studies were obtained from our search. Experimentally, 11 articles using solely VEGF for bone formation had a group or a timepoint significantly better than the corresponding control group. 18 articles revealed no significant difference of VEGF compared to the control group and 1 article reported a significant decreased bone growth using VEGF compared to control. CONCLUSION Based on these results no clinical studies have yet been performed. However, indications in the best use of VEGF from experimental studies could be made towards that the optimal release is within the first three weeks, in defect models, with the best effect before eight weeks. Future designs should incorporate this with standardised and reproducible models for verification towards clinical practice. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE This systematic review aims to assess the existing literature to focus on methodologies and outcomes that can provide future knowledge regarding the solitary use of VEGF for bone regeneration in a clinical setting.
Collapse
Affiliation(s)
- Chris H. Dreyer
- Orthopaedic Research Laboratory, Department of Orthopaedics & Traumatology, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, 5000, Odense C, Denmark
- Musculoskeletal Research Laboratory, Department of Orthopaedic Surgery & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China
- Acute Medicine, Department of Emergency Medicine, Slagelse Hospital, Slagelse, Denmark
| | - Kristian Kjaergaard
- Orthopaedic Research Laboratory, Department of Orthopaedics & Traumatology, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, 5000, Odense C, Denmark
| | - Ming Ding
- Orthopaedic Research Laboratory, Department of Orthopaedics & Traumatology, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, 5000, Odense C, Denmark
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopaedic Surgery & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| |
Collapse
|
14
|
Palomino-Durand C, Lopez M, Marchandise P, Martel B, Blanchemain N, Chai F. Chitosan/Polycyclodextrin (CHT/PCD)-Based Sponges Delivering VEGF to Enhance Angiogenesis for Bone Regeneration. Pharmaceutics 2020; 12:pharmaceutics12090784. [PMID: 32825081 PMCID: PMC7557476 DOI: 10.3390/pharmaceutics12090784] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/11/2020] [Accepted: 08/15/2020] [Indexed: 02/07/2023] Open
Abstract
Vascularization is one of the main challenges in bone tissue engineering (BTE). In this study, vascular endothelial growth factor (VEGF), known for its angiogenic effect, was delivered by our developed sponge, derived from a polyelectrolyte complexes hydrogel between chitosan (CHT) and anionic cyclodextrin polymer (PCD). This sponge, as a scaffold for growth factor delivery, was formed by freeze-drying a homogeneous CHT/PCD hydrogel, and thereafter stabilized by a thermal treatment. Microstructure, water-uptake, biodegradation, mechanical properties, and cytocompatibility of sponges were assessed. VEGF-delivery following incubation in medium was then evaluated by monitoring the VEGF-release profile and its bioactivity. CHT/PCD sponge showed a porous (open porosity of 87.5%) interconnected microstructure with pores of different sizes (an average pore size of 153 μm), a slow biodegradation (12% till 21 days), a high water-uptake capacity (~600% in 2 h), an elastic property under compression (elastic modulus of compression 256 ± 4 kPa), and a good cytocompatibility in contact with osteoblast and endothelial cells. The kinetic release of VEGF was found to exert a pro-proliferation and a pro-migration effect on endothelial cells, which are two important processes during scaffold vascularization. Hence, CHT/PCD sponges were promising vehicles for the delivery of growth factors in BTE.
Collapse
Affiliation(s)
- Carla Palomino-Durand
- U1008 Controlled Drug Delivery Systems and Biomaterials, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Régional Universitaire de Lille (CHU Lille), University of Lille, 59000 Lille, France; (C.P.-D.); (M.L.); (N.B.)
| | - Marco Lopez
- U1008 Controlled Drug Delivery Systems and Biomaterials, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Régional Universitaire de Lille (CHU Lille), University of Lille, 59000 Lille, France; (C.P.-D.); (M.L.); (N.B.)
| | - Pierre Marchandise
- ULR 4490–MABLab–Adiposité Médullaire et Os, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Régional Universitaire de Lille (CHU Lille), University of Lille, 59000 Lille, France;
- ULR 4490–MABLab–Adiposité Médullaire et Os, Univ. Littoral Côte d’Opale, 62200 Boulogne-sur-Mer, France
| | - Bernard Martel
- UMR 8207, UMET—Unité Matériaux et Transformations, Centre National de la Recherche Scientifique (CNRS), Institut National de la Recherche Agronomique (INRA), Ecole Nationale Supérieure de Chimie de Lille (ENSCL), University of Lille, 59655 Lille, France;
| | - Nicolas Blanchemain
- U1008 Controlled Drug Delivery Systems and Biomaterials, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Régional Universitaire de Lille (CHU Lille), University of Lille, 59000 Lille, France; (C.P.-D.); (M.L.); (N.B.)
| | - Feng Chai
- U1008 Controlled Drug Delivery Systems and Biomaterials, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Régional Universitaire de Lille (CHU Lille), University of Lille, 59000 Lille, France; (C.P.-D.); (M.L.); (N.B.)
- Correspondence: ; Tel.: +33-320-626-997
| |
Collapse
|
15
|
Review of the Pathways Involved in the Osteogenic Differentiation of Adipose-Derived Stem Cells. J Craniofac Surg 2019; 30:703-708. [PMID: 30839467 DOI: 10.1097/scs.0000000000005447] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Grafts and prosthetic materials used for the repair of bone defects are often accompanied by comorbidity and rejection. Therefore, there is an immense need for novel approaches to combating the issues surrounding such defects. Because of their accessibility, substantial proportion, and osteogenic differentiation potential, adipose-derived stem cells (ASCs) make for an ideal source of bone tissue in regenerative medicine. However, efficient induction of ASCs toward an osteoblastic lineage in vivo is met with challenges, and many signaling pathways must come together to secure osteoblastogenesis. Among them are bone morphogenic protein, wingless-related integration site protein, Notch, Hedgehog, fibroblast growth factor, vascular endothelial growth factor, and extracellular regulated-signal kinase. The goal of this literature review is to conglomerate the present research on these pathways to formulate a better understanding of how ASCs are most effectively transformed into bone in the context of tissue engineering.
Collapse
|
16
|
Nejadnik H, Tseng J, Daldrup-Link H. Magnetic resonance imaging of stem cell-macrophage interactions with ferumoxytol and ferumoxytol-derived nanoparticles. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1552. [PMID: 30734542 PMCID: PMC6579657 DOI: 10.1002/wnan.1552] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/13/2018] [Accepted: 12/19/2018] [Indexed: 01/07/2023]
Abstract
"Off the shelf" allogeneic stem cell transplants and stem cell nano-composites are being used for the treatment of degenerative bone diseases. However, major and minor histocompatibility antigens of therapeutic cell transplants can be recognized as foreign and lead to their rejection by the host immune system. If a host immune response is identified within the first week post-transplant, immune modulating therapies could be applied to prevent graft failure and support engraftment. Ferumoxytol (Feraheme™) is an FDA approved iron oxide nanoparticle preparation for the treatment of anemia in patients. Ferumoxytol can be used "off label" as an magnetic resonance (MR) contrast agent, as these nanoparticles provide measurable signal changes on magnetic resonance imaging (MRI). In this focused review article, we will discuss three methods to localize and identify innate immune responses to stem cell transplants using ferumoxytol-enhanced MRI, which are based on tracking stem cells, tracking macrophages or detecting mediators of cell death: (a) monitor MRI signal changes of ferumoxytol-labeled stem cells in the presence or absence of innate immune responses, (b) monitor influx of ferumoxytol-labeled macrophages into stem cell implants, and (c) monitor apoptosis of stem cell implants with caspase-3 activatable nanoparticles. These techniques can detect transplant failure at an early stage, when immune-modulating interventions can potentially preserve the viability of the cell transplants and thereby improve bone and cartilage repair outcomes. Approaches 1 and 2 are immediately translatable to clinical practice. This article is categorized under: Diagnostic Tools > in vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Cells at the Nanoscale Diagnostic Tools > Biosensing.
Collapse
Affiliation(s)
- Hossein Nejadnik
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| | - Jessica Tseng
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| | - Heike Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| |
Collapse
|
17
|
Donos N, Dereka X, Calciolari E. The use of bioactive factors to enhance bone regeneration: A narrative review. J Clin Periodontol 2019; 46 Suppl 21:124-161. [DOI: 10.1111/jcpe.13048] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/08/2018] [Accepted: 12/20/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Nikos Donos
- Centre for Oral Immunobiology & Regenerative Medicine & Centre for Oral Clinical Research (COCR); Institute of Dentistry, Barts & The London School of Medicine & Dentistry; Queen Mary University of London (QMUL); London UK
| | - Xanthippi Dereka
- Centre for Oral Immunobiology & Regenerative Medicine & Centre for Oral Clinical Research (COCR); Institute of Dentistry, Barts & The London School of Medicine & Dentistry; Queen Mary University of London (QMUL); London UK
- Department of Periodontology; School of Dentistry; National and Kapodistrian University of Athens; Athens Greece
| | - Elena Calciolari
- Centre for Oral Immunobiology & Regenerative Medicine & Centre for Oral Clinical Research (COCR); Institute of Dentistry, Barts & The London School of Medicine & Dentistry; Queen Mary University of London (QMUL); London UK
| |
Collapse
|
18
|
Reconstruction of a Calvarial Wound Complicated by Infection: Comparing the Effects of Biopatterned Bone Morphogenetic Protein 2 and Vascular Endothelial Growth Factor. J Craniofac Surg 2019; 30:260-264. [PMID: 30339591 DOI: 10.1097/scs.0000000000004779] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Bone morphogenetic protein 2 (BMP2) bioprinted on biological matrix induces osseous regeneration in large calvarial defects in rabbits, both uncomplicated and scarred. Healing in unfavorable defects scarred from previous infection is decreased due in part to the lack of vascularity. This impedes the access of mesenchymal stem cells, key to osseous regeneration and the efficacy of BMP2, to the wound bed. The authors hypothesized that bioprinted vascular endothelial growth factor (VEGF) would augment the osseous regeneration achieved with low dose biopatterned BMP2 alone. Thirteen New Zealand white rabbits underwent subtotal calvariectomy using a dental cutting burr. Care was taken to preserve the underlying dura. A 15 mm × 15 mm flap of bone was cut away and incubated in a 1 × 108 cfu/mL planktonic solution of S aureus before reimplantation. After 2 weeks of subsequent infection the flap was removed and the surgical wound debrided followed by 10 days of antibiotic treatment. On postoperative day 42 the calvarial defects were treated with acellular dermal matrix bioprinted with nothing (control), VEGF, BMP2, BMP2/VEGF combined. Bone growth was analyzed with serial CT and postmortem histology. Defects treated with BMP2 (BMP2 alone and BMP2/VEGF combination) showed significantly greater healing than control and VEGF treated defect (P < 0.5). Vascular endothelial growth factor treated defect demonstrated less healing than control and VEGF/BMP2 combination treatments achieved less healing than BMP2 alone though these differences were nonsignificant. Low dose BMP2-patterned acellular dermal matrix improves healing of scarred calvarial defects. Vascular endothelial growth factor at the doses applied in this study failed to increase healing.
Collapse
|
19
|
Lee E, Ko JY, Kim J, Park JW, Lee S, Im GI. Osteogenesis and angiogenesis are simultaneously enhanced in BMP2-/VEGF-transfected adipose stem cells through activation of the YAP/TAZ signaling pathway. Biomater Sci 2019; 7:4588-4602. [DOI: 10.1039/c9bm01037h] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
While bone has the capability to heal itself, there is a great difficulty in reconstituting large bone defects created by heavy trauma or the resection of malignant tumors.
Collapse
Affiliation(s)
- Eugene Lee
- Research Institute for Integrative Regenerative Biomedical Engineering
- Dongguk University
- Goyang 10326
- Republic of Korea
- Department of Orthopaedics
| | - Ji-Yun Ko
- Research Institute for Integrative Regenerative Biomedical Engineering
- Dongguk University
- Goyang 10326
- Republic of Korea
| | - Juyoung Kim
- Research Institute for Integrative Regenerative Biomedical Engineering
- Dongguk University
- Goyang 10326
- Republic of Korea
| | - Jeong-Won Park
- Research Institute for Integrative Regenerative Biomedical Engineering
- Dongguk University
- Goyang 10326
- Republic of Korea
| | - Songhee Lee
- Research Institute for Integrative Regenerative Biomedical Engineering
- Dongguk University
- Goyang 10326
- Republic of Korea
| | - Gun-Il Im
- Research Institute for Integrative Regenerative Biomedical Engineering
- Dongguk University
- Goyang 10326
- Republic of Korea
- Department of Orthopaedics
| |
Collapse
|
20
|
Quarto N, Shailendra S, Meyer NP, Menon S, Renda A, Longaker MT. Twist1-Haploinsufficiency Selectively Enhances the Osteoskeletal Capacity of Mesoderm-Derived Parietal Bone Through Downregulation of Fgf23. Front Physiol 2018; 9:1426. [PMID: 30374308 PMCID: PMC6196243 DOI: 10.3389/fphys.2018.01426] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 09/19/2018] [Indexed: 02/05/2023] Open
Abstract
Craniofacial development is a program exquisitely orchestrated by tissue contributions and regulation of genes expression. The basic helix–loop–helix (bHLH) transcription factor Twist1 expressed in the skeletal mesenchyme is a key regulator of craniofacial development playing an important role during osteoskeletogenesis. This study investigates the postnatal impact of Twist1 haploinsufficiency on the osteoskeletal ability and regeneration on two calvarial bones arising from tissues of different embryonic origin: the neural crest-derived frontal and the mesoderm-derived parietal bones. We show that Twist1 haplonsufficiency as well Twist1-sh-mediated silencing selectively enhanced osteogenic and tissue regeneration ability of mesoderm-derived bones. Transcriptomic profiling, gain-and loss-of-function experiments revealed that Twist1 haplonsufficiency triggers its selective activity on mesoderm-derived bone through a sharp downregulation of the bone-derived hormone Fgf23 that is upregulated exclusively in wild-type parietal bone.
Collapse
Affiliation(s)
- Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, CA, United States.,Dipartimento di Scienze Biomediche Avanzate, Universita' degli Studi di Napoli Federico II, Naples, Italy
| | - Siny Shailendra
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, CA, United States
| | - Nathaniel P Meyer
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, CA, United States
| | - Siddharth Menon
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, CA, United States
| | - Andrea Renda
- Dipartimento di Scienze Biomediche Avanzate, Universita' degli Studi di Napoli Federico II, Naples, Italy
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, CA, United States
| |
Collapse
|
21
|
Nguyen MK, Jeon O, Dang PN, Huynh CT, Varghai D, Riazi H, McMillan A, Herberg S, Alsberg E. RNA interfering molecule delivery from in situ forming biodegradable hydrogels for enhancement of bone formation in rat calvarial bone defects. Acta Biomater 2018; 75:105-114. [PMID: 29885529 PMCID: PMC6119505 DOI: 10.1016/j.actbio.2018.06.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 06/01/2018] [Accepted: 06/04/2018] [Indexed: 11/22/2022]
Abstract
RNA interference (RNAi) may be an effective and valuable tool for promoting the growth of functional tissue, as short interfering RNA (siRNA) and microRNA (miRNA) can block the expression of genes that have negative effects on tissue regeneration. Our group has recently reported that the localized and sustained presentation of siRNA against noggin (siNoggin) and miRNA-20a from in situ forming poly(ethylene glycol) (PEG) hydrogels enhanced osteogenic differentiation of encapsulated human bone marrow-derived mesenchymal stem cells (hMSCs). Here, the capacity of the hydrogel system to accelerate bone formation in a rat calvarial bone defect model is presented. After 12 weeks post-implantation, the hydrogels containing encapsulated hMSCs and miRNA-20a resulted in more bone formation in the defects than the hydrogels containing hMSCs without siRNA or with negative control siRNA. This localized and sustained RNA interfering molecule delivery system may provide an excellent platform for healing bony defects and other tissues. STATEMENT OF SIGNIFICANCE Delivery of RNAi molecules may be a valuable strategy to guide cell behavior for tissue engineering applications, but to date there have been no reports of a biomaterial system capable of both encapsulation of cells and controlled delivery of incorporated RNA. Here, we present PEG hydrogels that form in situ via Michael type reaction, and that permit encapsulation of hMSCs and the concomitant controlled delivery of siNoggin and/or miRNA-20a. These RNAs were chosen to suppress noggin, a BMP-2 antagonist, and/or PPAR-γ, a negative regulator of BMP-2-mediated osteogenesis, and therefore promote osteogenic differentiation of hMSCs and subsequent bone repair in critical-sized rat calvarial defects. Simultaneous delivery of hMSCs and miRNA-20a enhanced repair of these defects compared to hydrogels containing hMSCs without siRNA or with negative control siRNA. This in situ forming PEG hydrogel system offers an exciting platform for healing critical-sized bone defects by localized, controlled delivery of RNAi molecules to encapsulated hMSCs and surrounding cells.
Collapse
Affiliation(s)
- Minh K Nguyen
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Oju Jeon
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Phuong N Dang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Cong T Huynh
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Davood Varghai
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Hooman Riazi
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Alexandra McMillan
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Samuel Herberg
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Department of Orthopaedic Surgery, Case Western Reserve University, Cleveland, OH 44106, United States.
| |
Collapse
|
22
|
Abstract
Craniofacial surgery, since its inauguration, has been the culmination of collaborative efforts to solve complex congenital, dysplastic, oncological, and traumatic cranial bone defects. Now, 50 years on from the first craniofacial meeting, the collaborative efforts between surgeons, scientists, and bioengineers are further advancing craniofacial surgery with new discoveries in tissue regeneration. Recent advances in regenerative medicine and stem cell biology have transformed the authors' understanding of bone healing, the role of stem cells governing bone healing, and the effects of the niche environment and extracellular matrix on stem cell fate. This review aims at summarizing the advances within each of these fields.
Collapse
|
23
|
Chen X, Wang L, Zhao K, Wang H. Osteocytogenesis: Roles of Physicochemical Factors, Collagen Cleavage, and Exogenous Molecules. TISSUE ENGINEERING PART B-REVIEWS 2018; 24:215-225. [PMID: 29304315 DOI: 10.1089/ten.teb.2017.0378] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Osteocytes, the most abundant cell type in mammalian bone, are generally considered as the terminally differentiated cells of osteoblasts that are progressively self-buried or passively embedded in bone matrix. Emerging evidence reveals the essential functions of osteocytes in bone homeostasis and mechanotransduction. However, our knowledge on osteocytes, especially their formation, remains scarce. In this regard, the current review mainly focuses on several key factors that drive the osteocytic differentiation of osteoblasts, that is, osteocytogenesis. Available literature has demonstrated the involvement of physicochemical factors such as matrix composition, oxygen tension, and mechanical stress in the osteoblast-to-osteocyte transition. During cell migration and matrix remodeling, the matrix metalloproteinase-dependent collagen cleavage would play an "active" role in maturation and maintenance of the osteocytes. Besides, some in vitro methodologies are also established to induce the transformation of osteoblastic cell lines and primary mesenchymal cells to preosteocytes through cell transfection or addition of exogenous molecules (e.g., fibroblast growth factor-2, retinoic acid), which could potentiate the effort to form functional bone substitutes through elevated osteocytogenesis. Thus, advances of new technologies would enable comprehensive and in-depth understanding of osteocytes and their development, which in turn help promote the research on osteocyte biology and osteopathology.
Collapse
Affiliation(s)
- Xuening Chen
- 1 National Engineering Research Center for Biomaterials, Sichuan University , Chengdu, China
| | - Lichen Wang
- 2 Department of Biomedical Engineering, Chemistry and Biological Sciences, Stevens Institute of Technology , Hoboken, New Jersey
| | - Kaitao Zhao
- 2 Department of Biomedical Engineering, Chemistry and Biological Sciences, Stevens Institute of Technology , Hoboken, New Jersey
| | - Hongjun Wang
- 2 Department of Biomedical Engineering, Chemistry and Biological Sciences, Stevens Institute of Technology , Hoboken, New Jersey
| |
Collapse
|
24
|
Translating the role of osteogenic-angiogenic coupling in bone formation: Highly efficient chitosan-pDNA activated scaffolds can accelerate bone regeneration in critical-sized bone defects. Biomaterials 2017; 149:116-127. [DOI: 10.1016/j.biomaterials.2017.09.036] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/12/2017] [Accepted: 09/27/2017] [Indexed: 12/31/2022]
|
25
|
Murphy MP, Quarto N, Longaker MT, Wan DC. * Calvarial Defects: Cell-Based Reconstructive Strategies in the Murine Model. Tissue Eng Part C Methods 2017; 23:971-981. [PMID: 28825366 DOI: 10.1089/ten.tec.2017.0230] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Calvarial defects pose a continued clinical dilemma for reconstruction. Advancements within the fields of stem cell biology and tissue engineering have enabled researchers to develop reconstructive strategies using animal models. We review the utility of various animal models and focus on the mouse, which has aided investigators in understanding cranial development and calvarial bone healing. The murine model has also been used to study regenerative approaches to critical-sized calvarial defects, and we discuss the application of stem cells such as bone marrow-derived mesenchymal stromal cells, adipose-derived stromal cells, muscle-derived stem cells, and pluripotent stem cells to address deficient bone in this animal. Finally, we highlight strategies to manipulate stem cells using various growth factors and inhibitors and ultimately how these factors may prove crucial in future advancements within calvarial reconstruction using native skeletal stem cells.
Collapse
Affiliation(s)
- Matthew P Murphy
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University , Stanford, California.,2 Lorry I. Lokey Stem Cell Research Building, Stanford Stem Cell Biology and Regenerative Medicine Institute, Stanford University , Stanford, California
| | - Natalina Quarto
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University , Stanford, California
| | - Michael T Longaker
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University , Stanford, California.,2 Lorry I. Lokey Stem Cell Research Building, Stanford Stem Cell Biology and Regenerative Medicine Institute, Stanford University , Stanford, California
| | - Derrick C Wan
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University , Stanford, California
| |
Collapse
|
26
|
Moser N, Goldstein J, Kauffmann P, Epple M, Schliephake H. Experimental variation of the level and the ratio of angiogenic and osteogenic signaling affects the spatiotemporal expression of bone-specific markers and organization of bone formation in ectopic sites. Clin Oral Investig 2017; 22:1223-1234. [DOI: 10.1007/s00784-017-2202-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 09/04/2017] [Indexed: 01/30/2023]
|
27
|
Ning B, Zhao Y, Buza JA, Li W, Wang W, Jia T. Surgically‑induced mouse models in the study of bone regeneration: Current models and future directions (Review). Mol Med Rep 2017; 15:1017-1023. [PMID: 28138711 PMCID: PMC5367352 DOI: 10.3892/mmr.2017.6155] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 12/13/2016] [Indexed: 01/17/2023] Open
Abstract
Bone regeneration has been extensively studied over the past several decades. The surgically‑induced mouse model is the key animal model for studying bone regeneration, of the various research strategies used. These mouse models mimic the trauma and recovery processes in vivo and serve as carriers for tissue engineering and gene modification to test various therapies or associated genes in bone regeneration. The present review introduces a classification of surgically induced mouse models in bone regeneration, evaluates the application and value of these models and discusses the potential development of further innovations in this field in the future.
Collapse
Affiliation(s)
- Bin Ning
- Department of Orthopedic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Yunpeng Zhao
- Department of Orthopedic Surgery, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - John A Buza
- Department of Orthopedic Surgery, New York University Medical Center, New York, NY 10003, USA
| | - Wei Li
- Department of Orthopedic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Wenzhao Wang
- Department of Orthopedic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Tanghong Jia
- Department of Orthopedic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
| |
Collapse
|
28
|
Fujii K, Ito A, Mutsuzaki H, Murai S, Sogo Y, Hara Y, Yamazaki M. Reducing the risk of impaired bone apposition to titanium screws with the use of fibroblast growth factor-2-apatite composite layer coating. J Orthop Surg Res 2017; 12:1. [PMID: 28057033 PMCID: PMC5217243 DOI: 10.1186/s13018-016-0501-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 12/06/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Loosening of screws is a common problem in orthopedic and maxillofacial surgery. Modifying the implant surface to improve the mechanical strength of screws has been tried and reported. We developed screws coated with fibroblast growth factor-2 (FGF-2)-apatite composite layers. We then showed, in a percutaneous external fixation model, that this composite layer had the ability to hold and release FGF-2 slowly, thereby reducing the risk of pin tract infection of the percutaneous external fixation. The objective of the current study was to clarify the effect of FGF-2-apatite composite layers on titanium screws on bone formation around the screw. METHODS We analyzed samples of previously performed animal experiments. The screws were coated with FGF-2-apatite composite layers by immersing them in supersaturated calcium phosphate solutions containing FGF-2. Then, the uncoated, apatite-coated, and FGF-2-apatite composite layer-coated screws were implanted percutaneously in rabbits. Finally, using inflammation-free histological sections, we histomorphometrically assessed them for the presence of bone formation. Weibull plot analysis was then applied to the data. RESULTS On average, screws coated with FGF-2-apatite composite layers showed a significantly higher bone apposition rate than the uncoated or apatite-coated screws. Although the difference in the average bone apposition rate was small, the FGF-2-apatite composite layers produced a significant, marked reduction in the incidence of impaired bone formation around the screw compared with the incidence in the absence of FGF-2 (uncoated and apatite-coated screws). The probability of resulting in a bone apposition rate equal to or less than 63.75%, for example, is 3.5 × 10-4 for screws coated with the FGF-2-apatite composite layers versus 0.05 for screws in the absence of FGF-2. CONCLUSIONS FGF-2-apatite composite layer coating significantly reduced the risk of impaired bone apposition to the screw. Thus, it is feasible to use titanium screws coated with FGF-2-apatite composite layers as internal fixation screws to decrease the risk of loosening.
Collapse
Affiliation(s)
- Kengo Fujii
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Atsuo Ito
- Health Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan.
| | - Hirotaka Mutsuzaki
- Department of Orthopaedic Surgery, Ibaraki Prefectural University of Health Sciences, 4669-2 Ami Ami-machi, Inashiki-gun, Ibaraki, 300-0394, Japan
| | - Shinji Murai
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Yu Sogo
- Health Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Yuki Hara
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Masashi Yamazaki
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| |
Collapse
|
29
|
Acellular dermal matrix loading with bFGF achieves similar acceleration of bone regeneration to BMP-2 via differential effects on recruitment, proliferation and sustained osteodifferentiation of mesenchymal stem cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 70:62-70. [DOI: 10.1016/j.msec.2016.08.049] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 08/02/2016] [Accepted: 08/20/2016] [Indexed: 11/19/2022]
|
30
|
Senarath-Yapa K, Li S, Walmsley GG, Zielins E, Paik K, Britto JA, Grigoriadis AE, Wan DC, Liu KJ, Longaker MT, Quarto N. Small Molecule Inhibition of Transforming Growth Factor Beta Signaling Enables the Endogenous Regenerative Potential of the Mammalian Calvarium. Tissue Eng Part A 2016; 22:707-20. [PMID: 27036931 DOI: 10.1089/ten.tea.2015.0527] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Current approaches for the treatment of skeletal defects are suboptimal, principally because the ability of bone to repair and regenerate is poor. Although the promise of effective cellular therapies for skeletal repair is encouraging, these approaches are limited by the risks of infection, cellular contamination, and tumorigenicity. Development of a pharmacological approach would therefore help avoid some of these potential risks. This study identifies transforming growth factor beta (TGFβ) signaling as a potential pathway for pharmacological modulation in vivo. We demonstrate that inhibition of TGFβ signaling by the small molecule SB431542 potentiates calvarial skeletal repair through activation of bone morphogenetic protein (BMP) signaling on osteoblasts and dura mater cells participating in healing of calvarial defects. Cells respond to inhibition of TGFβ signaling by producing higher levels of BMP2 that upregulates inhibitory Smad6 expression, thus providing a negative feedback loop to contain excessive BMP signaling. Importantly, study on human osteoblasts indicates that molecular mechanism(s) triggered by SB431542 are conserved. Collectively, these data provide insights into the use of small molecules to modulate key signaling pathways for repairing skeletal defects.
Collapse
Affiliation(s)
- Kshemendra Senarath-Yapa
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine , Stanford, California.,2 Department of Craniofacial Development and Stem Cell Biology, Dental Institute , King's College London, London, United Kingdom .,3 Department of Plastic and Reconstructive Surgery, North Western Deanery , Manchester, United Kingdom
| | - Shuli Li
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Graham G Walmsley
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine , Stanford, California.,4 Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine , Stanford, California
| | - Elizabeth Zielins
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Kevin Paik
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Jonathan A Britto
- 5 Department of Craniofacial Surgery, Great Ormond Street Hospital , London, United Kingdom
| | - Agamemnon E Grigoriadis
- 2 Department of Craniofacial Development and Stem Cell Biology, Dental Institute , King's College London, London, United Kingdom
| | - Derrick C Wan
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Karen J Liu
- 2 Department of Craniofacial Development and Stem Cell Biology, Dental Institute , King's College London, London, United Kingdom
| | - Michael T Longaker
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine , Stanford, California.,4 Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine , Stanford, California
| | - Natalina Quarto
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine , Stanford, California.,6 Dipartimento di Scienze Biomediche Avanzate, Universita' degli Studi di Napoli Federico II , Napoli, Italy
| |
Collapse
|
31
|
Han LH, Conrad B, Chung MT, Deveza L, Jiang X, Wang A, Butte MJ, Longaker MT, Wan D, Yang F. Winner of the Young Investigator Award of the Society for Biomaterials at the 10th World Biomaterials Congress, May 17-22, 2016, Montreal QC, Canada: Microribbon-based hydrogels accelerate stem cell-based bone regeneration in a mouse critical-size cranial defect model. J Biomed Mater Res A 2016; 104:1321-31. [PMID: 26991141 DOI: 10.1002/jbm.a.35715] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Revised: 02/23/2016] [Accepted: 03/09/2016] [Indexed: 12/31/2022]
Abstract
Stem cell-based therapies hold great promise for enhancing tissue regeneration. However, the majority of cells die shortly after transplantation, which greatly diminishes the efficacy of stem cell-based therapies. Poor cell engraftment and survival remain a major bottleneck to fully exploiting the power of stem cells for regenerative medicine. Biomaterials such as hydrogels can serve as artificial matrices to protect cells during delivery and guide desirable cell fates. However, conventional hydrogels often lack macroporosity, which restricts cell proliferation and delays matrix deposition. Here we report the use of injectable, macroporous microribbon (μRB) hydrogels as stem cell carriers for bone repair, which supports direct cell encapsulation into a macroporous scaffold with rapid spreading. When transplanted in a critical-sized, mouse cranial defect model, μRB-based hydrogels significantly enhanced the survival of transplanted adipose-derived stromal cells (ADSCs) (81%) and enabled up to three-fold cell proliferation after 7 days. In contrast, conventional hydrogels only led to 27% cell survival, which continued to decrease over time. MicroCT imaging showed μRBs enhanced and accelerated mineralized bone repair compared to hydrogels (61% vs. 34% by week 6), and stem cells were required for bone repair to occur. These results suggest that paracrine signaling of transplanted stem cells are responsible for the observed bone repair, and enhancing cell survival and proliferation using μRBs further promoted the paracrine-signaling effects of ADSCs for stimulating endogenous bone repair. We envision μRB-based scaffolds can be broadly useful as a novel scaffold for enhancing stem cell survival and regeneration of other tissue types. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 1321-1331, 2016.
Collapse
Affiliation(s)
- Li-Hsin Han
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Dr, Edward Building Room 114, Stanford, California 94305
| | - Bogdan Conrad
- Program of Stem Cell Biology and Regenerative Medicine, Stanford University, 300 Pasteur Dr, Edward Building Room 114, Stanford, California 94305
| | - Michael T Chung
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, 257 Campus Dr, Hagey Building Room GK106, Stanford, California 94305
| | - Lorenzo Deveza
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Dr, Edward Building Room 114, Stanford, California 94305
| | - Xinyi Jiang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Dr, Edward Building Room 114, Stanford, California 94305
| | - Andrew Wang
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Dr, Grant Building Room H307A, Stanford, California 94305
| | - Manish J Butte
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Dr, Grant Building Room H307A, Stanford, California 94305
| | - Michael T Longaker
- Division of Plastic and Reconstructive Surgery, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 257 Campus Dr, Hagey Building Room GK106, Stanford, California 94305
| | - Derrick Wan
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, 257 Campus Dr, Hagey Building Room GK106, Stanford, California 94305
| | - Fan Yang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Dr, Edward Building Room 114, Stanford, California 94305.,Department of Bioengineering, Stanford University, 300 Pasteur Dr, Edward Building Room 114, Stanford, California 94305
| |
Collapse
|
32
|
E. Klontzas M, I. Kenanidis E, J. MacFarlane R, Michail T, E. Potoupnis M, Heliotis M, Mantalaris A, Tsiridis E. Investigational drugs for fracture healing: preclinical & clinical data. Expert Opin Investig Drugs 2016; 25:585-96. [DOI: 10.1517/13543784.2016.1161757] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
33
|
Zhang W, Zhang X, Ling J, Wei X, Jian Y. Osteo-/odontogenic differentiation of BMP2 and VEGF gene-co-transfected human stem cells from apical papilla. Mol Med Rep 2016; 13:3747-54. [PMID: 26986020 PMCID: PMC4838134 DOI: 10.3892/mmr.2016.4993] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 02/04/2016] [Indexed: 12/13/2022] Open
Abstract
Stem cells from apical papilla (SCAP) possess clear osteo-/odontogenic differentiation capabilities, and are regarded as the major cellular source for root dentin development. Bone morphogenetic protein 2 (BMP2) and vascular endothelial growth factor (VEGF) serve pivotal roles in the modulation of tooth development and dentin formation. However, the synergistic effects of BMP2 and VEGF on osteo-/odontogenic differentiation of SCAP remain unclear. The current study aimed to investigate the proliferative and osteo-/odontogenic differentiating capabilities of BMP2 and VEGF gene-co-transfected SCAP (SCAP-BMP2-VEGF) in vitro. The basic characteristics of the isolated SCAP were identified by the induction of multipotent differentiation and by flow cytometry. Lentiviral vector-mediated gene transfection was conducted with SCAP in order to construct blank vector-transfected SCAP (SCAP-green fluorescent protein), BMP2 gene-transfected SCAP (SCAP-BMP2), VEGF gene-transfected SCAP (SCAP-VEGF) and SCAP-BMP2-VEGF. The Cell Counting Kit 8 assay was used to analyze the proliferative capacities of the four groups of cells. The expression of osteo-/odontogenic genes and proteins in the cells were evaluated by reverse transcription-quantitative polymerase chain reaction and western blotting. The mineralized nodules formed by the four group cells were visualized by alkaline phosphatase (ALP) staining. Among the four groups of cells, SCAP-VEGF was demonstrated to exhibit increased proliferation, and SCAP-BMP2-VEGF exhibited reduced proliferation during eight days observation. SCAP-BMP2-VEGF exhibited significantly increased expression levels of ALP, osteocalcin, dentin sialophosphoprotein, dentin matrix acidic phosphoprotein gene 1 and dentin sialoprotein than the other three groups at the majority of the time points. Furthermore, the SCAP-BMP2-VEGF group exhibited a significantly greater number of ALP-positive mineralized nodules than the other groups following 16 days culture in vitro. In conclusion, lentiviral vector-mediated BMP2 and VEGF gene co-transfection significantly activated the osteo-/odontogenic differentiation of human SCAP.
Collapse
Affiliation(s)
- Wen Zhang
- Guangdong Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, Guanghua School and Hospital of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Xiaolei Zhang
- Guangdong Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, Guanghua School and Hospital of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Junqi Ling
- Guangdong Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, Guanghua School and Hospital of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Xi Wei
- Guangdong Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, Guanghua School and Hospital of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Yutao Jian
- Guangdong Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, Guanghua School and Hospital of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| |
Collapse
|
34
|
Mele L, Vitiello PP, Tirino V, Paino F, De Rosa A, Liccardo D, Papaccio G, Desiderio V. Changing Paradigms in Cranio-Facial Regeneration: Current and New Strategies for the Activation of Endogenous Stem Cells. Front Physiol 2016; 7:62. [PMID: 26941656 PMCID: PMC4764712 DOI: 10.3389/fphys.2016.00062] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 02/09/2016] [Indexed: 12/20/2022] Open
Abstract
Craniofacial area represent a unique district of human body characterized by a very high complexity of tissues, innervation and vascularization, and being deputed to many fundamental function such as eating, speech, expression of emotions, delivery of sensations such as taste, sight, and earing. For this reasons, tissue loss in this area following trauma or for example oncologic resection, have a tremendous impact on patients' quality of life. In the last 20 years regenerative medicine has emerged as one of the most promising approach to solve problem related to trauma, tissue loss, organ failure etc. One of the most powerful tools to be used for tissue regeneration is represented by stem cells, which have been successfully implanted in different tissue/organs with exciting results. Nevertheless, both autologous and allogeneic stem cell transplantation raise many practical and ethical concerns that make this approach very difficult to apply in clinical practice. For this reason different cell free approaches have been developed aiming to the mobilization, recruitment, and activation of endogenous stem cells into the injury site avoiding exogenous cells implant but instead stimulating patients' own stem cells to repair the lesion. To this aim many strategies have been used including functionalized bioscaffold, controlled release of stem cell chemoattractants, growth factors, BMPs, Platelet-Rich-Plasma, and other new strategies such as ultrasound wave and laser are just being proposed. Here we review all the current and new strategies used for activation and mobilization of endogenous stem cells in the regeneration of craniofacial tissue.
Collapse
Affiliation(s)
- Luigi Mele
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| | - Pietro Paolo Vitiello
- Medical Oncology, Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale "F. Magrassi e A. Lanzara," Second University of Naples Naples, Italy
| | - Virginia Tirino
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| | - Francesca Paino
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| | - Alfredo De Rosa
- Department of Odontology and Surgery, Second University of Naples Naples, Italy
| | - Davide Liccardo
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| | - Gianpaolo Papaccio
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| | - Vincenzo Desiderio
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| |
Collapse
|
35
|
Lee JH, Lee YJ, Cho HJ, Kim DW, Shin H. The incorporation of bFGF mediated by heparin into PCL/gelatin composite fiber meshes for guided bone regeneration. Drug Deliv Transl Res 2016; 5:146-59. [PMID: 25787740 DOI: 10.1007/s13346-013-0154-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The concept of guided bone regeneration facilitated by barrier membranes has been widely considered to achieve enhanced bone healing in maxillofacial surgery. However, the currently available membranes are limited in their active regulation of cellular activities. In this study, we fabricated polycaprolactone/gelatin composite electrospun nanofibers incorporated with basic fibroblast growth factor (bFGF) to direct bone regeneration. The fibrous morphology was maintained after the crosslinking and subsequent conjugation of heparin. Release of bFGF from electrospun nanofibers without heparin resulted in a spontaneous burst, while the heparin-mediated release of bFGF decreased the burst release in 24 h. The bFGF released from the nanofibers enhanced the proliferation and migration of human mesenchymal stem cells as well as the tubule formation of human umbilical cord blood cells. The subcutaneous implantation of fibers incorporated with bFGF mobilized a large number of cells positive for CD31 and smooth muscle alpha actin within 2 weeks. The effect of the nanofibers incorporated with bFGF on bone regeneration was evaluated on a calvarial critical size defect model. As compared to the mice that received fibers without bFGF, which presented minimal new bone formation (5.36 ± 3.4 % of the defect), those that received implants of heparinized nanofibers incorporated with 50 or 100 ng/mL bFGF significantly enhanced new bone formation (10.82 ± 2.2 and 17.55 ± 6.08 %). Taken together, our results suggest that the electrospun nanofibers incorporating bFGF have the potential to be used as an advanced membrane that actively enhances bone regeneration.
Collapse
Affiliation(s)
- Ji-hye Lee
- Department of Bioengineering, Hanyang University, 17 Haengdang-dong, Seongdong-gu, Seoul, 133-791, South Korea
| | | | | | | | | |
Collapse
|
36
|
|
37
|
Walmsley GG, Senarath-Yapa K, Wearda TL, Menon S, Hu MS, Duscher D, Maan ZN, Tsai JM, Zielins ER, Weissman IL, Gurtner GC, Lorenz HP, Longaker MT. Surveillance of Stem Cell Fate and Function: A System for Assessing Cell Survival and Collagen Expression In Situ. Tissue Eng Part A 2015; 22:31-40. [PMID: 26486617 DOI: 10.1089/ten.tea.2015.0221] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cell-based therapy is an emerging paradigm in skeletal regenerative medicine. However, the primary means by which transplanted cells contribute to bone repair and regeneration remain controversial. To gain an insight into the mechanisms of how both transplanted and endogenous cells mediate skeletal healing, we used a transgenic mouse strain expressing both the topaz variant of green fluorescent protein under the control of the collagen, type I, alpha 1 promoter/enhancer sequence (Col1a1(GFP)) and membrane-bound tomato red fluorescent protein constitutively in all cell types (R26(mTmG)). A comparison of healing in parietal versus frontal calvarial defects in these mice revealed that frontal osteoblasts express Col1a1 to a greater degree than parietal osteoblasts. Furthermore, the scaffold-based application of adipose-derived stromal cells (ASCs), bone marrow-derived mesenchymal stem cells (BM-MSCs), and osteoblasts derived from these mice to critical-sized calvarial defects allowed for investigation of cell survival and function following transplantation. We found that ASCs led to significantly faster rates of bone healing in comparison to BM-MSCs and osteoblasts. ASCs displayed both increased survival and increased Col1a1 expression compared to BM-MSCs and osteoblasts following calvarial defect transplantation, which may explain their superior regenerative capacity in the context of bone healing. Using this novel reporter system, we were able to elucidate how cell-based therapies impact bone healing and identify ASCs as an attractive candidate for cell-based skeletal regenerative therapy. These insights potentially influence stem cell selection in translational clinical trials evaluating cell-based therapeutics for osseous repair and regeneration.
Collapse
Affiliation(s)
- Graham G Walmsley
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California.,2 Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine , Stanford, California
| | - Kshemendra Senarath-Yapa
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Taylor L Wearda
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Siddharth Menon
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Michael S Hu
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California.,2 Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine , Stanford, California
| | - Dominik Duscher
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Zeshaan N Maan
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Jonathan M Tsai
- 2 Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine , Stanford, California
| | - Elizabeth R Zielins
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Irving L Weissman
- 2 Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine , Stanford, California
| | - Geoffrey C Gurtner
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - H Peter Lorenz
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Michael T Longaker
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California.,2 Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine , Stanford, California
| |
Collapse
|
38
|
Quarto N, Senarath-Yapa K, Renda A, Longaker MT. TWIST1 silencing enhances in vitro and in vivo osteogenic differentiation of human adipose-derived stem cells by triggering activation of BMP-ERK/FGF signaling and TAZ upregulation. Stem Cells 2015; 33:833-47. [PMID: 25446627 DOI: 10.1002/stem.1907] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 10/06/2014] [Accepted: 10/15/2014] [Indexed: 01/10/2023]
Abstract
Mesenchymal stem cells (MSCs) show promise for cellular therapy and regenerative medicine. Human adipose tissue-derived stem cells (hASCs) represent an attractive source of seed cells in bone regeneration. How to effectively improve osteogenic differentiation of hASCs in the bone tissue engineering has become a very important question with profound translational implications. Numerous regulatory pathways dominate osteogenic differentiation of hASCs involving transcriptional factors and signaling molecules. However, how these factors combine with each other to regulate hASCs osteogenic differentiation still remains to be illustrated. The highly conserved developmental proteins TWIST play key roles for transcriptional regulation in mesenchymal cell lineages. This study investigates TWIST1 function in hASCs osteogenesis. Our results show that TWIST1 shRNA silencing increased the osteogenic potential of hASCs in vitro and their skeletal regenerative ability when applied in vivo. We demonstrate that the increased osteogenic capacity observed with TWIST1 knockdown in hASCs is mediated through endogenous activation of BMP and ERK/FGF signaling leading, in turn, to upregulation of TAZ, a transcriptional modulator of MSCs differentiation along the osteoblast lineage. Inhibition either of BMP or ERK/FGF signaling suppressed TAZ upregulation and the enhanced osteogenesis in shTWIST1 hASCs. Cosilencing of both TWIST1 and TAZ abrogated the effect elicited by TWIST1 knockdown thus, identifying TAZ as a downstream mediator through which TWIST1 knockdown enhanced osteogenic differentiation in hASCs. Our functional study contributes to a better knowledge of molecular mechanisms governing the osteogenic ability of hASCs, and highlights TWIST1 as a potential target to facilitate in vivo bone healing.
Collapse
Affiliation(s)
- Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, California, USA; Dipartimento di Scienze Biomediche Avanzate, Universita' degli Studi di Napoli Federico II, Napoli, Italy
| | | | | | | |
Collapse
|
39
|
The Effect of Covalently Immobilized FGF-2 on Biphasic Calcium Phosphate Bone Substitute on Enhanced Biological Compatibility and Activity. BIOMED RESEARCH INTERNATIONAL 2015; 2015:742192. [PMID: 26436096 PMCID: PMC4576004 DOI: 10.1155/2015/742192] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 04/13/2015] [Accepted: 04/20/2015] [Indexed: 12/30/2022]
Abstract
The purpose of this research was to covalently graft fibroblast growth factor 2 (FGF-2) onto biphasic calcium phosphate (BCP) via a bifunctional cross-linker technique and to estimate the optimal dose of FGF-2 resulting in the best osteogenic differentiation of human mesenchymal stem cells (hMSCs). SEM observation revealed that the surface of the 100 ng FGF-2 coated BCP was completely covered with the nanoparticles expected to be from the silane coupling agent. XRD, FT-IR, and XPS analysis showed that silane treatment, bifunctional cross-linker coating, and FGF-2 covalent grafts were conducted successfully without deforming the crystalline structure of BCP. An MTT assay demonstrated that FGF-2 coated BCP had good biocompatibility, regardless of the concentration of FGF-2, after 24 or 48 h of incubation. An alkaline phosphatase (ALP) activity assay (14 days of incubation) and the ALP gene expression level of real-time PCR analysis (7 days of incubation) revealed that 50, 100, and 200 ng FGF-2 coated BCP induced the highest activities among all experimental groups and control group (P < 0.05). Thus, low concentrations of FGF-2 facilitated excellent osteogenesis and were effective at enhancing osteogenic potential. Also, the bifunctional cross-linker technique is expected to be a more feasible way to induce osteogenic differentiation while minimizing the risk of FGF-2 overdose.
Collapse
|
40
|
Abstract
Fibroblast growth factor (FGF) signaling pathways are essential regulators of vertebrate skeletal development. FGF signaling regulates development of the limb bud and formation of the mesenchymal condensation and has key roles in regulating chondrogenesis, osteogenesis, and bone and mineral homeostasis. This review updates our review on FGFs in skeletal development published in Genes & Development in 2002, examines progress made on understanding the functions of the FGF signaling pathway during critical stages of skeletogenesis, and explores the mechanisms by which mutations in FGF signaling molecules cause skeletal malformations in humans. Links between FGF signaling pathways and other interacting pathways that are critical for skeletal development and could be exploited to treat genetic diseases and repair bone are also explored.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Pierre J Marie
- UMR-1132, Institut National de la Santé et de la Recherche Médicale, Hopital Lariboisiere, 75475 Paris Cedex 10, France; Université Paris Diderot, Sorbonne Paris Cité, 75475 Paris Cedex 10, France
| |
Collapse
|
41
|
Herrmann M, Verrier S, Alini M. Strategies to Stimulate Mobilization and Homing of Endogenous Stem and Progenitor Cells for Bone Tissue Repair. Front Bioeng Biotechnol 2015; 3:79. [PMID: 26082926 PMCID: PMC4451737 DOI: 10.3389/fbioe.2015.00079] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/16/2015] [Indexed: 12/17/2022] Open
Abstract
The gold standard for the treatment of critical-size bone defects is autologous or allogenic bone graft. This has several limitations including donor site morbidity and the restricted supply of graft material. Cell-based tissue engineering strategies represent an alternative approach. Mesenchymal stem cells (MSCs) have been considered as a source of osteoprogenitor cells. More recently, focus has been placed on the use of endothelial progenitor cells (EPCs), since vascularization is a critical step in bone healing. Although many of these approaches have demonstrated effectiveness for bone regeneration, cell-based therapies require time consuming and cost-expensive in vitro cell expansion procedures. Accordingly, research is becoming increasingly focused on the homing and stimulation of native cells. The stromal cell-derived factor-1 (SDF-1) - CXCR4 axis has been shown to be critical for the recruitment of MSCs and EPCs. Vascular endothelial growth factor (VEGF) is a key factor in angiogenesis and has been targeted in many studies. Here, we present an overview of the different approaches for delivering homing factors to the defect site by absorption or incorporation to biomaterials, gene therapy, or via genetically manipulated cells. We further review strategies focusing on the stimulation of endogenous cells to support bone repair. Finally, we discuss the major challenges in the treatment of critical-size bone defects and fracture non-unions.
Collapse
Affiliation(s)
| | | | - Mauro Alini
- AO Research Institute Davos , Davos , Switzerland
| |
Collapse
|
42
|
Peric M, Dumic-Cule I, Grcevic D, Matijasic M, Verbanac D, Paul R, Grgurevic L, Trkulja V, Bagi CM, Vukicevic S. The rational use of animal models in the evaluation of novel bone regenerative therapies. Bone 2015; 70:73-86. [PMID: 25029375 DOI: 10.1016/j.bone.2014.07.010] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 06/30/2014] [Accepted: 07/05/2014] [Indexed: 12/31/2022]
Abstract
Bone has a high potential for endogenous self-repair. However, due to population aging, human diseases with impaired bone regeneration are on the rise. Current strategies to facilitate bone healing include various biomolecules, cellular therapies, biomaterials and different combinations of these. Animal models for testing novel regenerative therapies remain the gold standard in pre-clinical phases of drug discovery and development. Despite improvements in animal experimentation, excessive poorly designed animal studies with inappropriate endpoints and inaccurate conclusions are being conducted. In this review, we discuss animal models, procedures, methods and technologies used in bone repair studies with the aim to assist investigators in planning and performing scientifically sound experiments that respect the wellbeing of animals. In the process of designing an animal study for bone repair investigators should consider: skeletal characteristics of the selected animal species; a suitable animal model that mimics the intended clinical indication; an appropriate assessment plan with validated methods, markers, timing, endpoints and scoring systems; relevant dosing and statistically pre-justified sample sizes and evaluation methods; synchronization of the study with regulatory requirements and additional evaluations specific to cell-based approaches. This article is part of a Special Issue entitled "Stem Cells and Bone".
Collapse
Affiliation(s)
- Mihaela Peric
- University of Zagreb School of Medicine, Center for Translational and Clinical Research, Department for Intercellular Communication, Salata 2, Zagreb, Croatia.
| | - Ivo Dumic-Cule
- University of Zagreb School of Medicine, Center for Translational and Clinical Research, Laboratory for Mineralized Tissues, Salata 11, Zagreb, Croatia
| | - Danka Grcevic
- University of Zagreb School of Medicine, Department of Physiology and Immunology, Salata 3, Zagreb, Croatia
| | - Mario Matijasic
- University of Zagreb School of Medicine, Center for Translational and Clinical Research, Department for Intercellular Communication, Salata 2, Zagreb, Croatia
| | - Donatella Verbanac
- University of Zagreb School of Medicine, Center for Translational and Clinical Research, Department for Intercellular Communication, Salata 2, Zagreb, Croatia
| | - Ruth Paul
- Paul Regulatory Services Ltd, Fisher Hill Way, Cardiff CF15 8DR, UK
| | - Lovorka Grgurevic
- University of Zagreb School of Medicine, Center for Translational and Clinical Research, Laboratory for Mineralized Tissues, Salata 11, Zagreb, Croatia
| | - Vladimir Trkulja
- University of Zagreb School of Medicine, Department of Pharmacology, Salata 11, Zagreb, Croatia
| | - Cedo M Bagi
- Pfizer Inc., Global Research and Development, Global Science and Technology, 100 Eastern Point Road, Groton, CT 06340, USA
| | - Slobodan Vukicevic
- University of Zagreb School of Medicine, Center for Translational and Clinical Research, Laboratory for Mineralized Tissues, Salata 11, Zagreb, Croatia.
| |
Collapse
|
43
|
Donneys A, Nelson NS, Page EE, Deshpande SS, Felice PA, Tchanque-Fossuo CN, Spiegel JP, Buchman SR. Targeting angiogenesis as a therapeutic means to reinforce osteocyte survival and prevent nonunions in the aftermath of radiotherapy. Head Neck 2014; 37:1261-7. [PMID: 24801669 DOI: 10.1002/hed.23744] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 03/19/2014] [Accepted: 05/03/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Radiotherapy (XRT) exerts detrimental collateral effects on bone tissue through mechanisms of vascular damage and impediments to osteocytes, ultimately predisposing patients to the debilitating problems of late pathologic fractures and nonunions. We posit that angiogenic therapy will reverse these pathologic effects in a rat model of radiated fracture healing. METHODS Three groups of rats underwent mandibular osteotomy. Radiated groups received a fractionated 35-Gy dose before surgery. The deferoxamine (DFO) group received local injections postoperatively. A 40-day healing period was allowed before histology. Analysis of variance (ANOVA; p < .05) was used for group comparisons. RESULTS Radiated fractures revealed a significantly decreased osteocyte count and corresponding increase in empty lacunae when compared to nonradiated fractures (p = .001). With the addition of DFO, these differences were not appreciated. Further, a 42% increase in bony unions was observed after DFO therapy. CONCLUSION Targeting angiogenesis is a useful means for promoting osteocyte survival and preventing bone pathology after XRT.
Collapse
Affiliation(s)
- Alexis Donneys
- Craniofacial Research Laboratory, Plastic Surgery Section, University of Michigan, Ann Arbor, Michigan
| | - Noah S Nelson
- Craniofacial Research Laboratory, Plastic Surgery Section, University of Michigan, Ann Arbor, Michigan
| | - Erin E Page
- Craniofacial Research Laboratory, Plastic Surgery Section, University of Michigan, Ann Arbor, Michigan
| | - Sagar S Deshpande
- Craniofacial Research Laboratory, Plastic Surgery Section, University of Michigan, Ann Arbor, Michigan
| | - Peter A Felice
- Craniofacial Research Laboratory, Plastic Surgery Section, University of Michigan, Ann Arbor, Michigan.,Department of General Surgery, University of South Carolina School of Medicine, Columbia, South Carolina
| | | | - Joshua P Spiegel
- Craniofacial Research Laboratory, Plastic Surgery Section, University of Michigan, Ann Arbor, Michigan
| | - Steven R Buchman
- Craniofacial Research Laboratory, Plastic Surgery Section, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
44
|
|
45
|
Abstract
OBJECTIVE To demonstrate the pro-osteogenic effect of burn injury on heterotopic bone formation using a novel burn ossicle in vivo model. BACKGROUND Heterotopic ossification (HO), or the abnormal formation of bone in soft tissue, is a troubling sequela of burn and trauma injuries. The exact mechanism by which burn injury influences bone formation is unknown. The aim of this study was to develop a mouse model to study the effect of burn injury on heterotopic bone formation. We hypothesized that burn injury would enhance early vascularization and subsequent bone formation of subcutaneously implanted mesenchymal stem cells. METHODS Mouse adipose-derived stem cells were harvested from C57/BL6 mice, transfected with a BMP-2 adenovirus, seeded on collagen scaffolds (ossicles), and implanted subcutaneously in the flank region of 8 adult mice. Burn and sham groups were created with exposure of 30% surface area on the dorsum to 60°C water or 30°C water for 18 seconds, respectively (n = 4/group). Heterotopic bone volume was analyzed in vivo by micro-computed tomography for 3 months. Histological analysis of vasculogenesis was performed with platelet endothelial cell adhesion molecule staining. Osteogenic histological analysis was performed by Safranin O, Picrosirius red, and aniline blue staining. Qualitative analysis of heterotopic bone composition was completed with ex vivo Raman spectroscopy. RESULTS Subcutaneously implanted ossicles formed heterotopic bone. Ossicles from mice with burn injuries developed significantly more bone than sham control mice, analyzed by micro-computed tomography at 1, 2, and 3 months (P < 0.05), and had enhanced early and late endochondral ossification as demonstrated by Safranin O, Picrosirius red, and aniline blue staining. In addition, burn injury enhanced vascularization of the ossicles (P < 0.05). All ossicles demonstrated chemical composition characteristic of bone as demonstrated by Raman spectroscopy. CONCLUSIONS Burn injury increases the predilection to osteogenic differentiation of ectopically implanted ossicles. Early differences in vascularity correlated with later bone development. Understanding the role of burn injury on heterotopic bone formation is an important first step toward the development of treatment strategies aimed to prevent unwanted and detrimental heterotopic bone formation.
Collapse
|
46
|
Senarath-Yapa K, McArdle A, Renda A, Longaker MT, Quarto N. Adipose-derived stem cells: a review of signaling networks governing cell fate and regenerative potential in the context of craniofacial and long bone skeletal repair. Int J Mol Sci 2014; 15:9314-30. [PMID: 24865492 PMCID: PMC4100096 DOI: 10.3390/ijms15069314] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 05/16/2014] [Accepted: 05/20/2014] [Indexed: 02/07/2023] Open
Abstract
Improvements in medical care, nutrition and social care are resulting in a commendable change in world population demographics with an ever increasing skew towards an aging population. As the proportion of the world's population that is considered elderly increases, so does the incidence of osteodegenerative disease and the resultant burden on healthcare. The increasing demand coupled with the limitations of contemporary approaches, have provided the impetus to develop novel tissue regeneration therapies. The use of stem cells, with their potential for self-renewal and differentiation, is one potential solution. Adipose-derived stem cells (ASCs), which are relatively easy to harvest and readily available have emerged as an ideal candidate. In this review, we explore the potential for ASCs to provide tangible therapies for craniofacial and long bone skeletal defects, outline key signaling pathways that direct these cells and describe how the developmental signaling program may provide clues on how to guide these cells in vivo. This review also provides an overview of the importance of establishing an osteogenic microniche using appropriately customized scaffolds and delineates some of the key challenges that still need to be overcome for adult stem cell skeletal regenerative therapy to become a clinical reality.
Collapse
Affiliation(s)
- Kshemendra Senarath-Yapa
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, School of Medicine, Stanford University, Stanford, CA 94305-2200, USA; E-Mails: (K.S.-Y.); (A.M.)
| | - Adrian McArdle
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, School of Medicine, Stanford University, Stanford, CA 94305-2200, USA; E-Mails: (K.S.-Y.); (A.M.)
| | - Andrea Renda
- Dipartimento di Scienze Biomediche Avanzate, Universita’ degli Studi di Napoli Federico II, Napoli 80131, Italy; E-Mail:
| | - Michael T. Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, School of Medicine, Stanford University, Stanford, CA 94305-2200, USA; E-Mails: (K.S.-Y.); (A.M.)
| | - Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, School of Medicine, Stanford University, Stanford, CA 94305-2200, USA; E-Mails: (K.S.-Y.); (A.M.)
- Dipartimento di Scienze Biomediche Avanzate, Universita’ degli Studi di Napoli Federico II, Napoli 80131, Italy; E-Mail:
| |
Collapse
|
47
|
Jin Q, Giannobile WV. SDF-1 enhances wound healing of critical-sized calvarial defects beyond self-repair capacity. PLoS One 2014; 9:e97035. [PMID: 24800841 PMCID: PMC4011888 DOI: 10.1371/journal.pone.0097035] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 04/15/2014] [Indexed: 12/29/2022] Open
Abstract
Host blood circulating stem cells are an important cell source that participates in the repair of damaged tissues. The clinical challenge is how to improve the recruitment of circulating stem cells into the local wound area and enhance tissue regeneration. Stromal-derived factor-1 (SDF-1) has been shown to be a potent chemoattractant of blood circulating stem cells into the local wound microenvironment. In order to investigate effects of SDF-1 on bone development and the repair of a large bone defect beyond host self-repair capacity, the BMP-induced subcutaneous ectopic bone formation and calvarial critical-sized defect murine models were used in this preclinical study. A dose escalation of SDF-1 were loaded into collagen scaffolds containing BMP, VEGF, or PDGF, and implanted into subcutaneous sites at mouse dorsa or calvarial critical-sized bone defects for 2 and 4 weeks. The harvested biopsies were examined by microCT and histology. The results demonstrated that while SDF-1 had no effect in the ectopic bone model in promoting de novo osteogenesis, however, in the orthotopic bone model of the critical-sized defects, SDF-1 enhanced calvarial critical-sized bone defect healing similar to VEGF, and PDGF. These results suggest that SDF-1 plays a role in the repair of large critical-sized defect where more cells are needed while not impacting de novo bone formation, which may be associated with the functions of SDF-1 on circulating stem cell recruitment and angiogenesis.
Collapse
Affiliation(s)
- Qiming Jin
- Department of Cariology, Restorative Sciences and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| | - William V. Giannobile
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
48
|
Controlled release of granulocyte colony-stimulating factor enhances osteoconductive and biodegradable properties of Beta-tricalcium phosphate in a rat calvarial defect model. Int J Biomater 2014; 2014:134521. [PMID: 24829581 PMCID: PMC4009298 DOI: 10.1155/2014/134521] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 11/27/2013] [Accepted: 11/27/2013] [Indexed: 12/12/2022] Open
Abstract
Autologous bone grafts remain the gold standard for the treatment of congenital craniofacial disorders; however, there are potential problems including donor site morbidity and limitations to the amount of bone that can be harvested. Recent studies suggest that granulocyte colony-stimulating factor (G-CSF) promotes fracture healing or osteogenesis. The purpose of the present study was to investigate whether topically applied G-CSF can stimulate the osteoconductive properties of beta-tricalcium phosphate (β-TCP) in a rat calvarial defect model. A total of 27 calvarial defects 5 mm in diameter were randomly divided into nine groups, which were treated with various combinations of a β-TCP disc and G-CSF in solution form or controlled release system using gelatin hydrogel. Histologic and histomorphometric analyses were performed at eight weeks postoperatively. The controlled release of low-dose (1 μg and 5 μg) G-CSF significantly enhanced new bone formation when combined with a β-TCP disc. Moreover, administration of 5 μg G-CSF using a controlled release system significantly promoted the biodegradable properties of β-TCP. In conclusion, the controlled release of 5 μg G-CSF significantly enhanced the osteoconductive and biodegradable properties of β-TCP. The combination of G-CSF slow-release and β-TCP is a novel and promising approach for treating pediatric craniofacial bone defects.
Collapse
|
49
|
Quarto N, Li S, Renda A, Longaker MT. Exogenous activation of BMP-2 signaling overcomes TGFβ-mediated inhibition of osteogenesis in Marfan embryonic stem cells and Marfan patient-specific induced pluripotent stem cells. Stem Cells 2013; 30:2709-19. [PMID: 23037987 DOI: 10.1002/stem.1250] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 08/23/2012] [Indexed: 01/10/2023]
Abstract
Marfan syndrome (MFS) is a hereditary disease caused by mutations in the gene encoding Fibrillin-1 (FBN1) and characterized by a number of skeletal abnormalities, aortic root dilatation, and sometimes ectopia lentis. Although the molecular pathogenesis of MFS was attributed initially to a structural weakness of the fibrillin-rich microfibrils within the extracellular matrix, more recent results have documented that many of the pathogenic abnormalities in MFS are the result of alterations in TGFβ signaling. Mutations in FBN1 are therefore associated with increased activity and bioavailability of TGF-β1, which is suspected to be the basis for phenotypical similarities of FBN1 mutations in MFS and mutations in the receptors for TGFβ in Marfan syndrome-related diseases. We have previously demonstrated that unique skeletal phenotypes observed in human embryonic stem cells carrying the monogenic FBN1 mutation (MFS cells) are faithfully phenocopied by cells differentiated from induced pluripotent-stem cells (MFSiPS) derived independently from MFS patient fibroblasts. In this study, we aimed to determine further the biochemical features of transducing signaling(s) in MFS stem cells and MFSiPS cells highlighting a crosstalk between TGFβ and BMP signaling. Our results revealed that enhanced activation of TGFβ signaling observed in MFS cells decreased their endogenous BMP signaling. Moreover, exogenous BMP antagonized the enhanced TGFβ signaling in both MFS stem cells and MFSiPS cells therefore, rescuing their ability to undergo osteogenic differentiation. This study advances our understanding of molecular mechanisms underlying the pathogenesis of bone loss/abnormal skeletogenesis in human diseases caused by mutations in FBN1.
Collapse
Affiliation(s)
- Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, California 94305, USA.
| | | | | | | |
Collapse
|
50
|
Deferoxamine restores callus size, mineralization, and mechanical strength in fracture healing after radiotherapy. Plast Reconstr Surg 2013; 131:711e-719e. [PMID: 23629110 DOI: 10.1097/prs.0b013e3182865c57] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Therapeutic augmentation of fracture-site angiogenesis with deferoxamine has proven to increase vascularity, callus size, and mineralization in long-bone fracture models. The authors posit that the addition of deferoxamine would enhance pathologic fracture healing in the setting of radiotherapy in a model where nonunions are the most common outcome. METHODS Thirty-five Sprague-Dawley rats were divided into three groups. Fracture, irradiated fracture, and irradiated fracture plus deferoxamine. The irradiated fracture and irradiated fracture plus deferoxamine groups received a human equivalent dose of radiotherapy [7 Gy/day for 5 days, (35 Gy)] 2 weeks before mandibular osteotomy and external fixation. The irradiated fracture plus deferoxamine group received injections of deferoxamine into the fracture callus after surgery. After a 40-day healing period, mandibles were dissected, clinically assessed for bony union, imaged with micro-computed tomography, and tension tested to failure. RESULTS Compared with irradiated fractures, metrics of callus size, mineralization, and strength in deferoxamine-treated mandibles were significantly increased. These metrics were restored to a level demonstrating no statistical difference from control fractures. In addition, the authors observed an increased rate of achieving bony unions in the irradiated fracture plus deferoxamine-treated group when compared with irradiated fracture (67 percent and 20 percent, respectively). CONCLUSIONS The authors' data demonstrate nearly total restoration of callus size, mineralization, and biomechanical strength, and a threefold increase in the rate of union with the use of deferoxamine. The authors' results suggest that the administration of deferoxamine may have the potential for clinical translation as a new treatment paradigm for radiation-induced pathologic fractures.
Collapse
|