1
|
Torres-Guzman RA, Avila FR, Maita KC, Garcia JP, De Sario GD, Borna S, Eldaly AS, Quinones-Hinojosa A, Zubair AC, Ho OA, Forte AJ. Bone Morphogenic Protein and Mesenchymal Stem Cells to Regenerate Bone in Calvarial Defects: A Systematic Review. J Clin Med 2023; 12:4064. [PMID: 37373757 DOI: 10.3390/jcm12124064] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND The use of bone morphogenic protein and mesenchymal stem cells has shown promise in promoting bone regeneration in calvarial defects. However, a systematic review of the available literature is needed to evaluate the efficacy of this approach. METHODS We comprehensively searched electronic databases using MeSH terms related to skull defects, bone marrow mesenchymal stem cells, and bone morphogenic proteins. Eligible studies included animal studies that used BMP therapy and mesenchymal stem cells to promote bone regeneration in calvarial defects. Reviews, conference articles, book chapters, and non-English language studies were excluded. Two independent investigators conducted the search and data extraction. RESULTS Twenty-three studies published between 2010 and 2022 met our inclusion criteria after a full-text review of the forty-five records found in the search. Eight of the 23 studies used mice as models, while 15 used rats. The most common mesenchymal stem cell was bone marrow-derived, followed by adipose-derived. BMP-2 was the most popular. Stem cells were embedded in Scaffold (13), Transduction (7), and Transfection (3), and they were delivered BMP to cells. Each treatment used 2 × 104-1 × 107 mesenchymal stem cells, averaging 2.26 × 106. Most BMP-transduced MSC studies used lentivirus. CONCLUSIONS This systematic review examined BMP and MSC synergy in biomaterial scaffolds or alone. BMP therapy and mesenchymal stem cells in calvarial defects, alone, or with a scaffold regenerated bone. This method treats skull defects in clinical trials. The best scaffold material, therapeutic dosage, administration method, and long-term side effects need further study.
Collapse
Affiliation(s)
| | - Francisco R Avila
- Division of Plastic Surgery, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL 32224, USA
| | - Karla C Maita
- Division of Plastic Surgery, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL 32224, USA
| | - John P Garcia
- Division of Plastic Surgery, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL 32224, USA
| | - Gioacchino D De Sario
- Division of Plastic Surgery, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL 32224, USA
| | - Sahar Borna
- Division of Plastic Surgery, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL 32224, USA
| | - Abdullah S Eldaly
- Division of Plastic Surgery, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL 32224, USA
| | | | - Abba C Zubair
- Department of Laboratory Medicine and Pathology, Transfusion Medicines and Stem Cell Therapy, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Olivia A Ho
- Division of Plastic Surgery, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL 32224, USA
| | - Antonio J Forte
- Division of Plastic Surgery, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL 32224, USA
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
2
|
Liu G, Chen J, Wang X, Liu Y, Ma Y, Tu X. Functionalized 3D-Printed ST2/Gelatin Methacryloyl/Polcaprolactone Scaffolds for Enhancing Bone Regeneration with Vascularization. Int J Mol Sci 2022; 23:ijms23158347. [PMID: 35955478 PMCID: PMC9368581 DOI: 10.3390/ijms23158347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 02/01/2023] Open
Abstract
Growth factors were often used to improve the bioactivity of biomaterials in order to fabricate biofunctionalized bone grafts for bone defect repair. However, supraphysiological concentrations of growth factors for improving bioactivity could lead to serious side effects, such as ectopic bone formation, radiculitis, swelling of soft tissue in the neck, etc. Therefore, safely and effectively applying growth factors in bone repair biomaterials comes to be an urgent problem that needs to be addressed. In this study, an appropriate concentration (50 ng/mL) of Wnt3a was used to pretreat the 3D-bioprinting gelatin methacryloyl(GelMA)/polycaprolactone(PCL) scaffold loaded with bone marrow stromal cell line ST2 for 24 h. This pretreatment promoted the cell proliferation, osteogenic differentiation, and mineralization of ST2 in the scaffold in vitro, and enhanced angiogenesis and osteogenesis after being implanted in critical-sized mouse calvarial defects. On the contrary, the inhibition of Wnt/β-catenin signaling in ST2 cells reduced the bone repair effect of this scaffold. These results suggested that ST2/GelMA/PCL scaffolds pretreated with an appropriate concentration of Wnt3a in culture medium could effectively enhance the osteogenic and angiogenic activity of bone repair biomaterials both in vitro and in vivo. Moreover, it would avoid the side effects caused by the supraphysiological concentrations of growth factors. This functionalized scaffold with osteogenic and angiogenic activity might be used as an outstanding bone substitute for bone regeneration and repair.
Collapse
|
3
|
Amirthalingam S, Lee SS, Rajendran AK, Kim I, Hwang NS, Rangasamy J. Addition of lactoferrin and substance P in a chitin/PLGA-CaSO 4 hydrogel for regeneration of calvarial bone defects. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 126:112172. [PMID: 34082973 DOI: 10.1016/j.msec.2021.112172] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 04/24/2021] [Accepted: 05/03/2021] [Indexed: 11/25/2022]
Abstract
Calcium-based injectable hydrogels with various bioactive active molecules possess a great potential for bone regeneration. Herein, we have synthesized a chitin-PLGA-calcium sulfate hydrogel (CSG) containing bioactive molecules - lactoferrin (LF) and substance P (SP). SEM and XRD analysis revealed that CS crystal growth was altered with the addition of LF. Rheological measurements indicated that the injectability of the hydrogels was maintained after the addition of LF, however, there was a reduction in storage modulus after LF addition. The addition of LF increased stem cell proliferation whereas, SP enhanced the cell migration. Osteogenic gene expression revealed that LF concentration at 25 μg/mg of CSG was optimal for a favourable outcome. To this optimized LF containing CSG, SP was incorporated and 0.05 μg/mg was found to be most effective (CSG-L3S2) in vitro studies. Further, the μ-CT and histological studies confirmed that CSG-L3S2 showed enhanced bone regeneration compared to the controls in critical-sized calvarial defect of mice. Thus the results indicate that a combination of the chemotactic agent (SP), pleiotropic growth protein (LF), and CS in the chitin-PLGA hydrogel could be a promising approach for non-load bearing bone defects.
Collapse
Affiliation(s)
- Sivashanmugam Amirthalingam
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi-682041, India; School of Chemical and Biological Engineering, the Institute of Chemical Processes, Seoul National University, Seoul, 151-742, Republic of Korea
| | - Seunghun S Lee
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 151-742, Republic of Korea
| | - Arun Kumar Rajendran
- School of Chemical and Biological Engineering, the Institute of Chemical Processes, Seoul National University, Seoul, 151-742, Republic of Korea
| | - Inseon Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 151-742, Republic of Korea
| | - Nathaniel S Hwang
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 151-742, Republic of Korea; School of Chemical and Biological Engineering, the Institute of Chemical Processes, Seoul National University, Seoul, 151-742, Republic of Korea; Bio-MAX Institute, Institute of Bio-Engineering, Seoul National University, Seoul, 151-742, Republic of Korea.
| | - Jayakumar Rangasamy
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi-682041, India.
| |
Collapse
|
4
|
Dubey S, Mishra R, Roy P, Singh RP. 3-D macro/microporous-nanofibrous bacterial cellulose scaffolds seeded with BMP-2 preconditioned mesenchymal stem cells exhibit remarkable potential for bone tissue engineering. Int J Biol Macromol 2020; 167:934-946. [PMID: 33189758 DOI: 10.1016/j.ijbiomac.2020.11.049] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/30/2020] [Accepted: 11/07/2020] [Indexed: 12/13/2022]
Abstract
Bone repair using BMP-2 is a promising therapeutic approach in clinical practices, however, high dosages required to be effective pose issues of cost and safety. The present study explores the potential of low dose BMP-2 treatment via tissue engineering approach, which amalgamates 3-D macro/microporous-nanofibrous bacterial cellulose (mNBC) scaffolds and low dose BMP-2 primed murine mesenchymal stem cells (C3H10T1/2 cells). Initial studies on cell-scaffold interaction using unprimed C3H10T1/2 cells confirmed that scaffolds provided a propitious environment for cell adhesion, growth, and infiltration, owing to its ECM-mimicking nano-micro-macro architecture. Osteogenic studies were conducted by preconditioning the cells with 50 ng/mL BMP-2 for 15 min, followed by culturing on mNBC scaffolds for up to three weeks. The results showed an early onset and significantly enhanced bone matrix secretion and maturation in the scaffolds seeded with BMP-2 primed cells compared to the unprimed ones. Moreover, mNBC scaffolds alone were able to facilitate the mineralization of cells to some extent. These findings suggest that, with the aid of 'osteoinduction' from low dose BMP-2 priming of stem cells and 'osteoconduction' from nano-macro/micro topography of mNBC scaffolds, a cost-effective bone tissue engineering strategy can be designed for quick and excellent in vivo osseointegration.
Collapse
Affiliation(s)
- Swati Dubey
- Microbial Biotechnology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, India.
| | - Rutusmita Mishra
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Partha Roy
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - R P Singh
- Microbial Biotechnology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, India.
| |
Collapse
|
5
|
Vordemvenne T, Wähnert D, Koettnitz J, Merten M, Fokin N, Becker A, Büker B, Vogel A, Kronenberg D, Stange R, Wittenberg G, Greiner JFW, Hütten A, Kaltschmidt C, Kaltschmidt B. Bone Regeneration: A Novel Osteoinductive Function of Spongostan by the Interplay between Its Nano- and Microtopography. Cells 2020; 9:cells9030654. [PMID: 32156086 PMCID: PMC7140719 DOI: 10.3390/cells9030654] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 02/28/2020] [Accepted: 03/05/2020] [Indexed: 12/16/2022] Open
Abstract
Scaffold materials for bone regeneration are crucial for supporting endogenous healing after accidents, infections, or tumor resection. Although beneficial impacts of microtopological or nanotopological cues in scaffold topography are commonly acknowledged, less consideration is given to the interplay between the microscale and nanoscale. Here, micropores with a 60.66 ± 24.48 µm diameter ordered by closely packed collagen fibers are identified in pre-wetted Spongostan, a clinically-approved collagen sponge. On a nanoscale level, a corrugated surface of the collagen sponge is observable, leading to the presence of 32.97 ± 1.41 nm pores. This distinct micro- and nanotopography is shown to be solely sufficient for guiding osteogenic differentiation of human stem cells in vitro. Transplantation of Spongostan into a critical-size calvarial rat bone defect further leads to fast regeneration of the lesion. However, masking the micro- and nanotopographical cues using SiO2 nanoparticles prevents bone regeneration in vivo. Therefore, we demonstrate that the identified micropores allow migration of stem cells, which are further driven towards osteogenic differentiation by scaffold nanotopography. The present findings emphasize the necessity of considering both micro- and nanotopographical cues to guide intramembranous ossification, and might provide an optimal cell- and growth-factor-free scaffold for bone regeneration in clinical settings.
Collapse
Affiliation(s)
- Thomas Vordemvenne
- Protestant Hospital of Bethel Foundation, Department of Trauma and Orthopedic Surgery, Burgsteig 13, 33617 Bielefeld, Germany; (T.V.); (D.W.); (J.K.)
| | - Dirk Wähnert
- Protestant Hospital of Bethel Foundation, Department of Trauma and Orthopedic Surgery, Burgsteig 13, 33617 Bielefeld, Germany; (T.V.); (D.W.); (J.K.)
| | - Julian Koettnitz
- Protestant Hospital of Bethel Foundation, Department of Trauma and Orthopedic Surgery, Burgsteig 13, 33617 Bielefeld, Germany; (T.V.); (D.W.); (J.K.)
| | - Madlen Merten
- Molecular Neurobiology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany; (M.M.); (A.V.)
| | - Nadine Fokin
- Thin Films & Physics of Nanostructures, Universitätsstrasse 25, 33615 Bielefeld, Germany; (N.F.); (A.B.); (B.B.); (A.H.)
| | - Andreas Becker
- Thin Films & Physics of Nanostructures, Universitätsstrasse 25, 33615 Bielefeld, Germany; (N.F.); (A.B.); (B.B.); (A.H.)
| | - Björn Büker
- Thin Films & Physics of Nanostructures, Universitätsstrasse 25, 33615 Bielefeld, Germany; (N.F.); (A.B.); (B.B.); (A.H.)
| | - Asaria Vogel
- Molecular Neurobiology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany; (M.M.); (A.V.)
| | - Daniel Kronenberg
- Department of Regenerative Musculoskeletal Medicine, Institute for Musculoskeletal Medicine, University Hospital Muenster, Westfaelische Wilhelms University Muenster, Albert-Schweitzer-Campus 1, Building D3, 48149 Muenster, Germany; (D.K.); (R.S.)
| | - Richard Stange
- Department of Regenerative Musculoskeletal Medicine, Institute for Musculoskeletal Medicine, University Hospital Muenster, Westfaelische Wilhelms University Muenster, Albert-Schweitzer-Campus 1, Building D3, 48149 Muenster, Germany; (D.K.); (R.S.)
| | - Günther Wittenberg
- Protestant Hospital of Bethel Foundation, Department of Diagnostic and Interventional Radiology, Burgsteig 13, 33617 Bielefeld, Germany;
| | - Johannes FW Greiner
- Department of Cell Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany (C.K.)
| | - Andreas Hütten
- Thin Films & Physics of Nanostructures, Universitätsstrasse 25, 33615 Bielefeld, Germany; (N.F.); (A.B.); (B.B.); (A.H.)
- Bielefeld Institute for Nanoscience (BINAS), Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany
| | - Christian Kaltschmidt
- Department of Cell Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany (C.K.)
- Bielefeld Institute for Nanoscience (BINAS), Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany
| | - Barbara Kaltschmidt
- Molecular Neurobiology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany; (M.M.); (A.V.)
- Department of Cell Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany (C.K.)
- Bielefeld Institute for Nanoscience (BINAS), Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany
- Correspondence: ; Tel.: +49-521-106-5624
| |
Collapse
|
6
|
Koleva PM, Keefer JH, Ayala AM, Lorenzo I, Han CE, Pham K, Ralston SE, Kim KD, Lee CC. Hyper-Crosslinked Carbohydrate Polymer for Repair of Critical-Sized Bone Defects. Biores Open Access 2019; 8:111-120. [PMID: 31346493 PMCID: PMC6657362 DOI: 10.1089/biores.2019.0021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
This study evaluated the safety and efficacy of a novel hyper-crosslinked carbohydrate polymer (HCCP) for the repair of critical-sized bone defects in comparison to two alternative treatments: autologous bone and poly(lactide-co-glycolide) with hyaluronic acid (PLGA/HA). Bilateral critical-sized defects were created in the lateral femoral condyles of skeletally mature New Zealand White rabbits, and they were subsequently implanted with HCCP, PLGA/HA, or autologous bone in a randomized manner. Clinical and behavioral observations were made daily, and radiological and histopathological evaluations were performed at 4, 10, and 16 weeks postimplantation. Defects implanted with HCCP showed progressive bone regeneration and bridging of the defect without adverse histological events. No signs of infection or inflammation associated with the implant material were observed in all animals that received HCCP implantation. A radiographic assessment performed at 16 weeks post-implantation showed significantly higher bone density and volume in defects implanted with HCCP compared to PLGA/HA. No statistically significant difference was observed in bone density and volume between HCCP and autologous bone. These findings demonstrate that HCCP is biocompatible, osteoconductive, and capable of promoting bone regeneration in vivo; therefore, it is suitable for both tissue engineering and the repair of critical-sized bone defects.
Collapse
Affiliation(s)
| | | | | | | | | | - Kristen Pham
- Molecular Matrix, Inc., West Sacramento, California
| | | | - Kee D Kim
- Department of Neurological Surgery, UC Davis School of Medicine, Sacramento, California
| | - Charles C Lee
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California, Davis, California
| |
Collapse
|
7
|
Cui Y, Fu S, Sun D, Xing J, Hou T, Wu X. EPC-derived exosomes promote osteoclastogenesis through LncRNA-MALAT1. J Cell Mol Med 2019; 23:3843-3854. [PMID: 31025509 PMCID: PMC6533478 DOI: 10.1111/jcmm.14228] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 01/21/2019] [Accepted: 01/27/2019] [Indexed: 12/17/2022] Open
Abstract
Bone repair involves bone resorption through osteoclastogenesis and the stimulation of neovascularization and osteogenesis by endothelial progenitor cells (EPCs). However, the role of EPCs in osteoclastogenesis is unclear. In this study, we assess the effects of EPC-derived exosomes on the migration and osteoclastic differentiation of primary mouse bone marrow-derived macrophages (BMMs) in vitro using immunofluorescence, western blotting, RT-PCR and Transwell assays. We also evaluated the effects of EPC-derived exosomes on the homing and osteoclastic differentiation of transplanted BMMs in a mouse bone fracture model in vivo. We found that EPCs cultured with BMMs secreted exosomes into the medium and, compared with EPCs, exosomes had a higher expression level of LncRNA-MALAT1. We confirmed that LncRNA-MALAT1 directly binds to miR-124 to negatively control miR-124 activity. Moreover, overexpression of miR-124 could reverse the migration and osteoclastic differentiation of BMMs induced by EPC-derived exosomes. A dual-luciferase reporter assay indicated that the integrin ITGB1 is the target of miR-124. Mice treated with EPC-derived exosome-BMM co-transplantations exhibited increased neovascularization at the fracture site and enhanced fracture healing compared with those treated with BMMs alone. Overall, our results suggest that EPC-derived exosomes can promote bone repair by enhancing recruitment and differentiation of osteoclast precursors through LncRNA-MALAT1.
Collapse
Affiliation(s)
- Yigong Cui
- Department of OrthopaedicsSouthwest HospitalThe Third Military Medical UniversityChongqingP.R. China
| | - Shenglong Fu
- Department of OrthopaedicsJinan Fifth People's HospitalShandongP.R. China
| | - Dong Sun
- Department of OrthopaedicsSouthwest HospitalThe Third Military Medical UniversityChongqingP.R. China
| | - Junchao Xing
- Department of OrthopaedicsSouthwest HospitalThe Third Military Medical UniversityChongqingP.R. China
| | - Tianyong Hou
- Department of OrthopaedicsSouthwest HospitalThe Third Military Medical UniversityChongqingP.R. China
| | - Xuehui Wu
- Department of OrthopaedicsSouthwest HospitalThe Third Military Medical UniversityChongqingP.R. China
| |
Collapse
|
8
|
Echave MC, Pimenta-Lopes C, Pedraz JL, Mehrali M, Dolatshahi-Pirouz A, Ventura F, Orive G. Enzymatic crosslinked gelatin 3D scaffolds for bone tissue engineering. Int J Pharm 2019; 562:151-161. [PMID: 30853482 DOI: 10.1016/j.ijpharm.2019.02.043] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/17/2019] [Accepted: 02/27/2019] [Indexed: 02/07/2023]
Abstract
Bone tissue engineering is an emerging medical field that has been developed in recent years to address pathologies with limited ability of bones to regenerate. Here we report the fabrication and characterization of microbial transglutaminase crosslinked gelatin-based scaffolds designed for serving as both cell substrate and growth factor release system. In particular, morphological, biomechanical and biological features have been analyzed. The enzyme ratio applied during the fabrication of the scaffolds affects the swelling capacity and the mechanical properties of the final structure. The developed systems are not cytotoxic according to the biocompatibility tests. The biological performance of selected formulations was studied using L-929 fibroblasts, D1 MSC and MG63 osteoblasts. Moreover, scaffolds allowed efficient osteogenic differentiation and signaling of MSCs. MSC cultured on the scaffolds not only presented lower proliferative and stemness profile, but also increased expression of osteoblast-related genes (Col1a1, Runx2, Osx). Furthermore, the in vitro release kinetics of vascular endothelial growth factor (VEGF) and bone morphogenetic protein -2 (BMP-2) from the scaffolds were also investigated. The release of the growth factors produced from the scaffolds followed a first order kinetics. These results highlight that the scaffolds designed and developed in this work may be suitable candidates for bone tissue regeneration purposes.
Collapse
Affiliation(s)
- Mari Carmen Echave
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, Vitoria-Gasteiz 01006, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - Carolina Pimenta-Lopes
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, C/ Feixa Llarga s/n 08907, Hospitalet de Llobregat, Spain
| | - José Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, Vitoria-Gasteiz 01006, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - Mehdi Mehrali
- Technical University of Denmark, Department of Health Technology, Center for Intestinal Absorption and Transport of Biopharmaceutical, 2800 Kgs, Denmark
| | - Alireza Dolatshahi-Pirouz
- Technical University of Denmark, Department of Health Technology, Center for Intestinal Absorption and Transport of Biopharmaceutical, 2800 Kgs, Denmark; Department of Dentistry - Regenerative Biomaterials, Radboud University Medical Center, Philips van Leydenlaan 25, Nijmegen 6525 EX, The Netherlands
| | - Fransesc Ventura
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, C/ Feixa Llarga s/n 08907, Hospitalet de Llobregat, Spain
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, Vitoria-Gasteiz 01006, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain; Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore.
| |
Collapse
|
9
|
Aquino-Martínez R, Monroe DG, Ventura F. Calcium mimics the chemotactic effect of conditioned media and is an effective inducer of bone regeneration. PLoS One 2019; 14:e0210301. [PMID: 30608979 PMCID: PMC6319750 DOI: 10.1371/journal.pone.0210301] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/18/2018] [Indexed: 12/22/2022] Open
Abstract
Background After bone resorption, ions and degraded organic components are co-released into the extracellular space. Ions and growth factors, although different in their biological nature, induce a common and coordinated chemotactic effect. Conditioned media has been used successfully in bone regeneration by promoting endogenous cell recruitment. Likewise, calcium alone act as a paracrine chemotactic signal, inducing the host’s undifferentiated progenitor cell infiltration into the implanted biomaterials. The aim of the present study was to compare the chemotactic effect of calcium and conditioned media in primary calvarial cells. Methods The chemotactic cell response was evaluated in vitro using an agarose spot and a wound healing assay. In addition, we used a calvarial bone explant model ex-vivo. The healing potential was also tested through an in vivo model, a critical-size calvarial bone defect in mice. For the in vivo experiment, cell-free calcium-containing or conditioned media-containing scaffolds were implanted, and MSC’s seeded scaffolds were used as positive control. After seven weeks post-implantation, samples were retrieved, and bone regeneration was evaluated by μCT and histological analysis. Osteogenic gene expression was evaluated by qPCR. Results We found that chemotactic cell migration in response to either calcium or conditioned media was equivalent in vitro and ex vivo. Accordingly, μCT analysis showed that bone regeneration induced by the MSC’s seeded scaffolds was similar to that obtained with cell-free calcium or conditioned media-containing scaffolds. Pre-treatment with SB202190, a highly selective p38 inhibitor, abrogated the chemotactic effect induced by conditioned media. In contrast, p38 activity was not essential for the calcium-induced chemotaxis. Moreover, BAPTA-AM treatment, a cytosolic calcium chelator, decreased the chemotactic effect and the expression of key osteogenic genes induced by calcium or conditioned media. Conclusion We show that calcium ions alone not only mimic the conditioned media chemotactic effect, but also induce an osteogenic effect similar to that produced by transplanted MSC’s in vivo. Furthermore, the chemotactic effect induced by conditioned media is calcium and p38 dependent. The rise in cytosolic calcium might integrate the different signaling pathways triggered by conditioned media and extracellular Ca2+. This calcium-driven in situ bone regeneration is a promising and convenient alternative to promote endogenous cell recruitment into the injured bone site. This pre-clinical cell-free and growth factor-free approach might avoid the disadvantages of the ex vivo cell manipulation.
Collapse
Affiliation(s)
- Rubén Aquino-Martínez
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
- Department of Medicine, Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, United States of America
| | - David G. Monroe
- Department of Medicine, Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, United States of America
| | - Francesc Ventura
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
- * E-mail:
| |
Collapse
|
10
|
Sivashanmugam A, Charoenlarp P, Deepthi S, Rajendran A, Nair SV, Iseki S, Jayakumar R. Injectable Shear-Thinning CaSO 4/FGF-18-Incorporated Chitin-PLGA Hydrogel Enhances Bone Regeneration in Mice Cranial Bone Defect Model. ACS APPLIED MATERIALS & INTERFACES 2017; 9:42639-42652. [PMID: 29143524 DOI: 10.1021/acsami.7b15845] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
For craniofacial bone regeneration, shear-thinning injectable hydrogels are favored over conventional scaffolds because of their improved defect margin adaptability, easier handling, and ability to be injected manually into deeper tissues. The most accepted method, after autografting, is the use of recombinant human bone morphogenetic protein-2 (BMP-2); however, complications such as interindividual variations, edema, and poor cost-efficiency in supraphysiological doses have been reported. The endogenous synthesis of BMP-2 is desirable, and a molecule which induces this is fibroblast growth factor-18 (FGF-18) because it can upregulate the BMP-2 expression by supressing noggin. We developed a chitin-poly(lactide-co-glycolide) (PLGA) composite hydrogel by regeneration chemistry and then incorporated CaSO4 and FGF-18 for this purpose. Rheologically, a 7-fold increase in the elastic modulus was observed in the CaSO4-incorporated chitin-PLGA hydrogels as compared to the chitin-PLGA hydrogel. Shear-thinning Herschel-Bulkley fluid nature was observed for both hydrogels. Chitin-PLGA/CaSO4 gel showed sustained release of FGF-18. In vitro osteogenic differentiation showed an enhanced alkaline phosphatase (ALP) expression in the FGF-18-containing chitin-PLGA/CaSO4 gel when compared to cells alone. Further, it was confirmed by studying the expression of osteogenic genes [RUNX2, ALP, BMP-2, osteocalcin (OCN), and osteopontin (OPN)], immunofluorescence staining of BMP-2, OCN, and OPN, and alizarin red S staining. Incorporation of FGF-18 in the hydrogel increased the endothelial cell migration. Further, the regeneration potential of the prepared hydrogels was tested in vivo, and longitudinal live animal μ-CT was performed. FGF-18-loaded chitin-PLGA/CaSO4 showed early and almost complete bone healing in comparison with chitin-PLGA/CaSO4, chitin-PLGA/FGF-18, chitin-PLGA, and sham control systems, as confirmed by hematoxylin and eosin and osteoid tetrachrome stainings. This shows that the CaSO4 and FGF-18-incorporated hydrogel is a potential candidate for craniofacial bone defect regeneration.
Collapse
Affiliation(s)
- A Sivashanmugam
- Center for Nanosciences and Molecular Medicine, Amrita University , Kochi 682041, India
| | - Pornkawee Charoenlarp
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University , Tokyo 113 8510, Japan
| | - S Deepthi
- Center for Nanosciences and Molecular Medicine, Amrita University , Kochi 682041, India
| | - Arunkumar Rajendran
- Center for Nanosciences and Molecular Medicine, Amrita University , Kochi 682041, India
| | - Shantikumar V Nair
- Center for Nanosciences and Molecular Medicine, Amrita University , Kochi 682041, India
| | - Sachiko Iseki
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University , Tokyo 113 8510, Japan
| | - R Jayakumar
- Center for Nanosciences and Molecular Medicine, Amrita University , Kochi 682041, India
| |
Collapse
|
11
|
Aquino-Martínez R, Angelo AP, Pujol FV. Calcium-containing scaffolds induce bone regeneration by regulating mesenchymal stem cell differentiation and migration. Stem Cell Res Ther 2017; 8:265. [PMID: 29145866 PMCID: PMC5689169 DOI: 10.1186/s13287-017-0713-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/18/2017] [Accepted: 10/26/2017] [Indexed: 11/17/2022] Open
Abstract
Background Osteoinduction and subsequent bone formation rely on efficient mesenchymal stem cell (MSC) recruitment. It is also known that migration is induced by gradients of growth factors and cytokines. Degradation of Ca2+-containing biomaterials mimics the bone remodeling compartment producing a localized calcium-rich osteoinductive microenvironment. The aim of our study was to determine the effect of calcium sulfate (CaSO4) on MSC migration. In addition, to evaluate the influence of CaSO4 on MSC differentiation and the potential molecular mechanisms involved. Methods A circular calvarial bone defect (5 mm diameter) was created in the parietal bone of 35 Balb-C mice. We prepared and implanted a cell-free agarose/gelatin scaffold alone or in combination with different CaSO4 concentrations into the bone defects. After 7 weeks, we determined the new bone regenerated by micro-CT and histological analysis. In vitro, we evaluated the CaSO4 effects on MSC migration by both wound healing and agarose spot assays. Osteoblastic gene expression after BMP-2 and CaSO4 treatment was also evaluated by qPCR. Results CaSO4 increased MSC migration and bone formation in a concentration-dependent manner. Micro-CT analysis showed that the addition of CaSO4 significantly enhanced bone regeneration compared to the scaffold alone. The histological evaluation confirmed an increased number of endogenous cells recruited into the cell-free CaSO4-containing scaffolds. Furthermore, MSC migration in vitro and active AKT levels were attenuated when CaSO4 and BMP-2 were in combination. Addition of LY294002 and Wortmannin abrogated the CaSO4 effects on MSC migration. Conclusions Specific CaSO4 concentrations induce bone regeneration of calvarial defects in part by acting on the host’s undifferentiated MSCs and promoting their migration. Progenitor cell recruitment is followed by a gradual increment in osteoblast gene expression. Moreover, CaSO4 regulates BMP-2-induced MSC migration by differentially activating the PI3K/AKT pathway. Altogether, these results suggest that CaSO4 scaffolds could have potential applications for bone regeneration. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0713-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rubén Aquino-Martínez
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain.,Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Alcira P Angelo
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Francesc Ventura Pujol
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
12
|
Aquino-Martínez R, Artigas N, Gámez B, Rosa JL, Ventura F. Extracellular calcium promotes bone formation from bone marrow mesenchymal stem cells by amplifying the effects of BMP-2 on SMAD signalling. PLoS One 2017; 12:e0178158. [PMID: 28542453 PMCID: PMC5444778 DOI: 10.1371/journal.pone.0178158] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 05/09/2017] [Indexed: 11/23/2022] Open
Abstract
Understanding the molecular events that regulate osteoblast differentiation is essential for the development of effective approaches to bone regeneration. In this study, we analysed the osteoinductive properties of extracellular calcium in bone marrow-derived mesenchymal stem cell (BM-MSC) differentiation. We cultured BM-MSCs in 3D gelatin scaffolds with Ca2+ and BMP-2 as osteoinductive agents. Early and late osteogenic gene expression and bone regeneration in a calvarial critical-size defect model demonstrate that extracellular Ca2+ enhances the effects of BMP-2 on Osteocalcin, Runx2 and Osterix expression and promotes bone regeneration in vivo. Moreover, we analysed the molecular mechanisms involved and observed an antagonistic effect between Ca2+ and BMP-2 on SMAD1/5, ERK and S6K signalling after 24 hours. More importantly, a cooperative effect between Ca2+ and BMP-2 on the phosphorylation of SMAD1/5, S6, GSK3 and total levels of β-CATENIN was observed at a later differentiation time (10 days). Furthermore, Ca2+ alone favoured the phosphorylation of SMAD1, which correlates with the induction of Bmp2 and Bmp4 gene expression. These data suggest that Ca2+ and BMP-2 cooperate and promote an autocrine/paracrine osteogenic feed-forward loop. On the whole, these results demonstrate the usefulness of calcium-based bone grafts or the addition of exogenous Ca2+ in bone tissue engineering.
Collapse
Affiliation(s)
- Rubén Aquino-Martínez
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L’Hospitalet de Llobregat, Spain
| | - Natalia Artigas
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L’Hospitalet de Llobregat, Spain
| | - Beatriz Gámez
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L’Hospitalet de Llobregat, Spain
| | - José Luis Rosa
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L’Hospitalet de Llobregat, Spain
| | - Francesc Ventura
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L’Hospitalet de Llobregat, Spain
- * E-mail:
| |
Collapse
|
13
|
Osteogenic differentiation of preosteoblasts on a hemostatic gelatin sponge. Sci Rep 2016; 6:32884. [PMID: 27616161 PMCID: PMC5018723 DOI: 10.1038/srep32884] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 08/16/2016] [Indexed: 02/07/2023] Open
Abstract
Bone tissue engineering provides many advantages for repairing skeletal defects. Although many different kinds of biomaterials have been used for bone tissue engineering, safety issues must be considered when using them in a clinical setting. In this study, we examined the effects of using a common clinical item, a hemostatic gelatin sponge, as a scaffold for bone tissue engineering. The use of such a clinically acceptable item may hasten the translational lag from laboratory to clinical studies. We performed both degradation and biocompatibility studies on the hemostatic gelatin sponge, and cultured preosteoblasts within the sponge scaffold to demonstrate its osteogenic differentiation potential. In degradation assays, the gelatin sponge demonstrated good stability after being immersed in PBS for 8 weeks (losing only about 10% of its net weight and about 54% decrease of mechanical strength), but pepsin and collagenases readily biodegraded it. The gelatin sponge demonstrated good biocompatibility to preosteoblasts as demonstrated by MTT assay, confocal microscopy, and scanning electron microscopy. Furthermore, osteogenic differentiation and the migration of preosteoblasts, elevated alkaline phosphatase activity, and in vitro mineralization were observed within the scaffold structure. Each of these results indicates that the hemostatic gelatin sponge is a suitable scaffold for bone tissue engineering.
Collapse
|
14
|
Wu Q, Yang B, Hu K, Cao C, Man Y, Wang P. Deriving Osteogenic Cells from Induced Pluripotent Stem Cells for Bone Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2016; 23:1-8. [PMID: 27392674 DOI: 10.1089/ten.teb.2015.0559] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Induced pluripotent stem cells (iPSCs), reprogrammed from adult somatic cells using defined transcription factors, are regarded as a promising cell source for tissue engineering. For the purpose of bone tissue regeneration, efficient in vitro differentiation of iPSCs into downstream cells, such as mesenchymal stem cells (MSCs), osteoblasts, or osteocyte-like cells, before use is necessary to limit undesired tumorogenesis associated with the pluripotency of iPSCs. Until recently numerous techniques on the production of iPSC-derived osteogenic progenitors have been introduced. We reviewed these protocols and provided a perspective on the comparisons of osteogenic potentials of (1) iPSC-derived osteogenic cells produced by different protocols, (2) iPSCs from different somatic origins, and (3) iPSC-derived MSC-like cells and bone marrow stem cells. Finally, we discussed the potential application of the diseased iPSCs for systematic bone disorders.
Collapse
Affiliation(s)
- Qingqing Wu
- 1 State Key Laboratory of Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University , Chengdu, China
| | - Bo Yang
- 1 State Key Laboratory of Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University , Chengdu, China
| | - Kevin Hu
- 2 University of Maryland Dental School , Baltimore, Maryland
| | - Cong Cao
- 3 Department of Stomatology, China-Japan Friendship Hospital , Beijing, China
| | - Yi Man
- 1 State Key Laboratory of Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University , Chengdu, China
| | - Ping Wang
- 1 State Key Laboratory of Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University , Chengdu, China .,2 University of Maryland Dental School , Baltimore, Maryland
| |
Collapse
|