1
|
Yang M, Gao Z, Cheng S, Wang Z, Ei-Seedi H, Du M. Novel Peptide Derived from Gadus morhua Stimulates Osteoblastic Differentiation and Mineralization through Wnt/β-Catenin and BMP Signaling Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:9691-9702. [PMID: 38639219 DOI: 10.1021/acs.jafc.3c06700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
Marine biodiversity offers a wide array of active ingredient resources. Gadus morhua peptides (GMPs) showed excellent osteoprotective effects in ovariectomized mice. However, the potential osteogenesis mechanisms of key osteogenic peptides in GMP were seldom reported. In this study, a novel osteogenic peptide (GETNPADSKPGSIR, P-GM-2) was screened from GMP. P-GM-2 has a high stability coefficient and a strong interaction with epidermal growth factor receptor. Cell culture experiments showed that P-GM-2 stimulated the expression of osteogenic differentiation markers to promote osteoblast proliferation, differentiation, and mineralization. Additionally, P-GM-2 phosphorylates GSK-3β, leading to the stabilization of β-catenin and its translocation to the nucleus, thus initiating the activation of the Wnt/β-catenin signaling pathway. Meanwhile, P-GM-2 could also regulate the osteogenic differentiation of preosteoblasts by triggering the BMP/Smad and mitogen-activated protein kinase signaling pathways. Further validation with specific inhibitors (ICG001 and Noggin) demonstrated that the osteogenic activity of P-GM-2 was revealed by the activation of the BMP and Wnt/β-catenin pathways. In summary, these results provide theoretical and practical insights into P-GM-2 as an effective antiosteoporosis active ingredient.
Collapse
Affiliation(s)
- Meilian Yang
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, SKL of Marine Food Processing & Safety Control, Dalian Polytechnic University, Dalian 116034, China
| | - Zengli Gao
- Inner Mongolia Enterprise Key Laboratory of Dairy Nutrition, Health & Safety, Inner Mongolia Mengniu Dairy (Group) Co. Ltd., Huhhot 011500, P. R. China
| | - Shuzhen Cheng
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, SKL of Marine Food Processing & Safety Control, Dalian Polytechnic University, Dalian 116034, China
| | - Zhenyu Wang
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, SKL of Marine Food Processing & Safety Control, Dalian Polytechnic University, Dalian 116034, China
| | - Hesham Ei-Seedi
- Pharmacognosy Group, Department of Medicinal Chemistry, Uppsala Biomedical Center, Uppsala University, Uppsala 75 123, Sweden
| | - Ming Du
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, SKL of Marine Food Processing & Safety Control, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
2
|
Jahangirnezhad M, Mahmoudinezhad SS, Moradi M, Moradi K, Rohani A, Tayebi L. Bone Scaffold Materials in Periodontal and Tooth-supporting Tissue Regeneration: A Review. Curr Stem Cell Res Ther 2024; 19:449-460. [PMID: 36578254 DOI: 10.2174/1574888x18666221227142055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND OBJECTIVES Periodontium is an important tooth-supporting tissue composed of both hard (alveolar bone and cementum) and soft (gingival and periodontal ligament) sections. Due to the multi-tissue architecture of periodontium, reconstruction of each part can be influenced by others. This review focuses on the bone section of the periodontium and presents the materials used in tissue engineering scaffolds for its reconstruction. MATERIALS AND METHODS The following databases (2015 to 2021) were electronically searched: ProQuest, EMBASE, SciFinder, MRS Online Proceedings Library, Medline, and Compendex. The search was limited to English-language publications and in vivo studies. RESULTS Eighty-three articles were found in primary searching. After applying the inclusion criteria, seventeen articles were incorporated into this study. CONCLUSION In complex periodontal defects, various types of scaffolds, including multilayered ones, have been used for the functional reconstruction of different parts of periodontium. While there are some multilayered scaffolds designed to regenerate alveolar bone/periodontal ligament/cementum tissues of periodontium in a hierarchically organized construct, no scaffold could so far consider all four tissues involved in a complete periodontal defect. The progress and material considerations in the regeneration of the bony part of periodontium are presented in this work to help investigators develop tissue engineering scaffolds suitable for complete periodontal regeneration.
Collapse
Affiliation(s)
- Mahmood Jahangirnezhad
- Department of Periodontics, School of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sadaf Sadat Mahmoudinezhad
- Department of Periodontics, School of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Melika Moradi
- Department of Periodontics, School of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Kooshan Moradi
- Department of Periodontics, School of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Rohani
- Department of Periodontics, School of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Lobat Tayebi
- School of Dentistry, Marquette University, Milwaukee, WI, 53233, USA
| |
Collapse
|
3
|
Bello SA, Cruz-Lebrón J, Rodríguez-Rivera OA, Nicolau E. Bioactive Scaffolds as a Promising Alternative for Enhancing Critical-Size Bone Defect Regeneration in the Craniomaxillofacial Region. ACS APPLIED BIO MATERIALS 2023; 6:4465-4503. [PMID: 37877225 DOI: 10.1021/acsabm.3c00432] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
Reconstruction of critical-size bone defects (CSDs) in the craniomaxillofacial (CMF) region remains challenging. Scaffold-based bone-engineered constructs have been proposed as an alternative to the classical treatments made with autografts and allografts. Scaffolds, a key component of engineered constructs, have been traditionally viewed as biologically passive temporary replacements of deficient bone lacking intrinsic cues to promote osteogenesis. Nowadays, scaffolds are functionalized, giving rise to bioactive scaffolds promoting bone regeneration more effectively than conventional counterparts. This review focuses on the three approaches most used to bioactivate scaffolds: (1) conferring microarchitectural designs or surface nanotopography; (2) loading bioactive molecules; and (3) seeding stem cells on scaffolds, providing relevant examples of in vivo (preclinical and clinical) studies where these methods are employed to enhance CSDs healing in the CMF region. From these, adding bioactive molecules (specifically bone morphogenetic proteins or BMPs) to scaffolds has been the most explored to bioactivate scaffolds. Nevertheless, the downsides of grafting BMP-loaded scaffolds in patients have limited its successful translation into clinics. Despite these drawbacks, scaffolds containing safer, cheaper, and more effective bioactive molecules, combined with stem cells and topographical cues, remain a promising alternative for clinical use to treat CSDs in the CMF complex replacing autografts and allografts.
Collapse
Affiliation(s)
- Samir A Bello
- Department of Chemistry, University of Puerto Rico, Rio Piedras Campus, PO Box 23346, San Juan, Puerto Rico 00931, United States
- Molecular Sciences Research Center, University of Puerto Rico, 1390 Ponce De León Ave, Suite 1-7, San Juan, Puerto Rico 00926, United States
| | - Junellie Cruz-Lebrón
- Department of Chemistry, University of Puerto Rico, Rio Piedras Campus, PO Box 23346, San Juan, Puerto Rico 00931, United States
- Molecular Sciences Research Center, University of Puerto Rico, 1390 Ponce De León Ave, Suite 1-7, San Juan, Puerto Rico 00926, United States
| | - Osvaldo A Rodríguez-Rivera
- Department of Chemistry, University of Puerto Rico, Rio Piedras Campus, PO Box 23346, San Juan, Puerto Rico 00931, United States
- Molecular Sciences Research Center, University of Puerto Rico, 1390 Ponce De León Ave, Suite 1-7, San Juan, Puerto Rico 00926, United States
| | - Eduardo Nicolau
- Department of Chemistry, University of Puerto Rico, Rio Piedras Campus, PO Box 23346, San Juan, Puerto Rico 00931, United States
- Molecular Sciences Research Center, University of Puerto Rico, 1390 Ponce De León Ave, Suite 1-7, San Juan, Puerto Rico 00926, United States
| |
Collapse
|
4
|
Parisi L, Ghezzi B, Toffoli A, Macaluso GM, Lumetti S. Aptamer-enriched scaffolds for tissue regeneration: a systematic review of the literature. Front Bioeng Biotechnol 2023; 11:1199651. [PMID: 37265990 PMCID: PMC10229892 DOI: 10.3389/fbioe.2023.1199651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/04/2023] [Indexed: 06/03/2023] Open
Abstract
Introduction: Aptamers are a brand-new class of receptors that can be exploited to improve the bioactivity of tissue engineering grafts. The aim of this work was to revise the current literature on in vitro and in vivo studies in order to i) identify current strategies adopted to improve scaffold bioactivity by aptamers; ii) assess effects of aptamer functionalization on cell behavior and iii) on tissue regeneration. Methods: Using a systematic search approach original research articles published up to 30 April 2022, were considered and screened. Results: In total, 131 records were identified and 18 were included in the final analysis. Included studies showed that aptamers can improve the bioactivity of biomaterials by specific adsorption of adhesive molecules or growth factors from the surrounding environment, or by capturing specific cell types. All the studies showed that aptamers ameliorate scaffold colonization by cells without modifying the physicochemical characteristics of the bare scaffold. Additionally, aptamers seem to promote the early stages of tissue healing and to promote anatomical and functional regeneration. Discussion: Although a metanalysis could not be performed due to the limited number of studies, we believe these findings provide solid evidence supporting the use of aptamers as a suitable modification to improve the bioactivity of tissue engineering constructs.
Collapse
Affiliation(s)
- Ludovica Parisi
- Laboratory for Oral Molecular Biology, Department of Orthodontics and Dentofacial Orthopedics, University of Bern, Bern, Switzerland
| | - Benedetta Ghezzi
- Centro Universitario di Odontoiatria, Dipartimento di Medicina e Chirurgia, University of Parma, Parma, Italy
- Istituto dei Materiali per l’Elettronica ed il Magnetismo, Consiglio Nazionale delle Ricerche, Parma, Italy
| | - Andrea Toffoli
- Centro Universitario di Odontoiatria, Dipartimento di Medicina e Chirurgia, University of Parma, Parma, Italy
| | - Guido M. Macaluso
- Centro Universitario di Odontoiatria, Dipartimento di Medicina e Chirurgia, University of Parma, Parma, Italy
- Istituto dei Materiali per l’Elettronica ed il Magnetismo, Consiglio Nazionale delle Ricerche, Parma, Italy
| | - Simone Lumetti
- Centro Universitario di Odontoiatria, Dipartimento di Medicina e Chirurgia, University of Parma, Parma, Italy
- Istituto dei Materiali per l’Elettronica ed il Magnetismo, Consiglio Nazionale delle Ricerche, Parma, Italy
| |
Collapse
|
5
|
Xu Y, Yang Y, Hua Z, Li S, Yang Z, Liu Q, Fu G, Ji P, Wu Q. BMP2 immune complexes promote new bone formation by facilitating the direct contact between osteoclasts and osteoblasts. Biomaterials 2021; 275:120890. [PMID: 34130144 DOI: 10.1016/j.biomaterials.2021.120890] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/22/2021] [Accepted: 05/06/2021] [Indexed: 11/17/2022]
Abstract
BMP2 antibody is proposed as a promising replacement for rhBMP2 in bone tissue engineering. Although studies have demonstrated its osteoinductive efficacy, the underlying osteogenic mechanism and adverse reactions of specific BMP2 antibody are not clarified yet, making it difficult to optimize the antibody for future application. By establishing BMP2 immune complexes (BMP2-ICs) ex vivo, we were able to introduce BMP2-ICs directly in vivo and found that BMP2-ICs promoted bone formation while suppressing osteoclastogenesis. However, ex vivo osteoclastogenic assays showed that BMP2-ICs promoted osteoclastogenesis by binding FcγR and activating PLCγ2 phosphorylation. Given that BMP2-ICs react with osteoblast and osteoclast lineage cells by the conjugated BMP2 domain and the Fc domain respectively, we introduced BMP2-ICs into coculture system of the two lineage cells and found that BMP2-ICs promoted osteogenesis while suppressing osteoclastogenesis by facilitating osteoblast-osteoclast contact and activating the EphrinB2-EphB4 signaling. This bidirectional function of BMP2-ICs was reproduced in the cranial bone resorption model, where osteoblast and osteoclast lineage cells co-localized. This study excluded the hidden problem of osteoclast overactivation that usually comes with rhBMP2 and clarified the first evidence of the mechanism of antibody-mediated bone regeneration, suggesting BMP2-ICs may present a promising therapy for bone diseases related with disrupted osteoclast-osteoblast interaction.
Collapse
Affiliation(s)
- Yamei Xu
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Yao Yang
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Ziyi Hua
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Shuang Li
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Zhenyu Yang
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Qianzi Liu
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Gang Fu
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China; Department of Oral Implantology, Stomatological Hospital of Chongqing Medical University, Chongqing, 401147, China
| | - Ping Ji
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Qingqing Wu
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China; Department of Oral Implantology, Stomatological Hospital of Chongqing Medical University, Chongqing, 401147, China.
| |
Collapse
|
6
|
Ouyang L, Cao J, Dai Q, Qiu D. New insight of immuno-engineering in osteoimmunomodulation for bone regeneration. Regen Ther 2021; 18:24-29. [PMID: 33778136 PMCID: PMC7985270 DOI: 10.1016/j.reth.2021.03.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 02/28/2021] [Accepted: 03/02/2021] [Indexed: 12/14/2022] Open
Abstract
With the continuous development of bone tissue engineering, the importance of immune response in bone tissue regeneration is gradually recognized. The new bone tissue engineering products should possess immunoregulatory functions, harmonizing the interactions between the bone's immune defense and regeneration systems, and promoting tissue regeneration. This article will interpret the relationship between the bone immune system, bone tissue regeneration, as well as the immunoregulatory function of bone biomaterials and seed stem cells in bone tissue engineering. This review locates arears for foucusing efforts at providing novel designs ideas about the development of immune-mediation targeted bone tissue engineering products and the evaluation strategy for the osteoimmunomodulation property of bone biomaterials.
Collapse
Affiliation(s)
- Long Ouyang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiankun Cao
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiang Dai
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Daojing Qiu
- Department of Orthopedics, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Functionalized Scaffold and Barrier Membrane with Anti-BMP-2 Monoclonal Antibodies for Alveolar Ridge Preservation in a Canine Model. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6153724. [PMID: 33029518 PMCID: PMC7530509 DOI: 10.1155/2020/6153724] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 08/04/2020] [Indexed: 01/14/2023]
Abstract
Introduction The aim of this study was to investigate the ability of anti-bone morphogenetic protein 2 monoclonal antibody (anti-BMP-2 mAb) to functionalize scaffolds to mediate bone regeneration in a canine model. Materials and Methods The mandibular right premolar 4 (PM4) was extracted in eight beagle dogs and grafted with anti-BMP-2 mAb+anorganic bovine bone mineral with 10% collagen (ABBM-C) and porcine bilayer native collagen membrane (CM). The ABBM-C and CM were functionalized with either anti-BMP-2 mAb (test group) or an isotype matched control mAb (control group). Animals were euthanized at 12 weeks for radiographic, histologic, and histomorphometric analyses. Outcomes were compared between groups. Results 3D imaging using cone beam computed tomography (CBCT) revealed that sites treated with ABBM-C and CM functionalized with anti-BMP-2 mAb exhibited significantly more remaining bone width near the alveolar crest, as well as buccal bone height, compared with control groups. Histologic and histomorphometric analyses demonstrated that in anti-BMP-2 mAb-treated sites, total tissue volume was significantly higher in the coronal part of the alveolar bone crest compared with control sites. In anti-BMP-2 mAb-treated sites, bone formation was observed under the barrier membrane. Conclusion Functionalization of the ABBM-C scaffold and CM appeared to have led to bone formation within healing alveolar bone sockets.
Collapse
|
8
|
The sialylation profile of IgG determines the efficiency of antibody directed osteogenic differentiation of iMSCs by modulating local immune responses and osteoclastogenesis. Acta Biomater 2020; 114:221-232. [PMID: 32771590 DOI: 10.1016/j.actbio.2020.07.055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 07/03/2020] [Accepted: 07/31/2020] [Indexed: 12/25/2022]
Abstract
Antibody-mediated osseous regeneration (AMOR) has been proved as a promising strategy for osteogenic differentiation of induced pluripotent stem cells derived MSCs (iMSCs). The key characteristic of antibody that determines the AMOR potential is largely unknown. The glycosylation profile of immunoglobulin G (IgG) represents a key checkpoint that determines its effector functions. Herein, we modified the sialylation profile of BMP2 antibodies to investigate the effects of glycosylation on antibody-mediated osteogenic differentiation of iMSCs. We found that over-sialylated BMP2 antibodies stimulated the highest amount of new bone while those non- or low-sialylated led to bone porosity and collapse. The immune response aroused by BMP2 immune complexes (BMP2-ICs) was intensified by desialylation, which contributed to an environment that favored osteoclastogenesis while inhibited osteoblastogenesis. In vitro study further demonstrated that the osteogenic potential of BMP2-ICs was not significantly affected by the degree of sialylation. On the other hand, BMP2-ICs could stimulate osteoclastogenesis by binding FcγRs on preosteoclasts directly, which was significantly intensified by desialylation and attenuated by over-sialylation. Bone defects implanted with alginate microbeads loaded with iMSCs and over-sialylated antibodies showed more bone formation than those sites with non- or low sialylated antibodies. Taken together, our study demonstrated that sialylation profile is one of the traits that decide the AMOR potential of BMP2 antibodies. Enhancement of sialylation may be a promising strategy to optimize antibody for iMSCs application in bone tissue engineering. STATEMENT OF SIGNIFICANCE: Antibody-mediated osseous regeneration (AMOR) is a promising strategy for bone tissue engineering that takes advantage of the specific reactivity of antibodies to sequester endogenous BMP2 and present it to osteoprogenitor cells. We previously demonstrated that BMP2 immune complex can drive iPSCs derived MSCs to osteogenic lineage. In this study, we analyze the effects of glycosylation profile on antibody directed osteogenic differentiation of iMSCs because glycosylation profile represents a key checkpoint that determines the effector functions of antibodies, and it is susceptible to variations in different clones. The results showed that sialylation profile is one of the traits that decides the AMOR potential of BMP2 antibody, and the enhancement of sialylation maybe a promising strategy to optimize antibodies for AMOR.
Collapse
|
9
|
Parisi L, Toffoli A, Ghezzi B, Mozzoni B, Lumetti S, Macaluso GM. A glance on the role of fibronectin in controlling cell response at biomaterial interface. JAPANESE DENTAL SCIENCE REVIEW 2019; 56:50-55. [PMID: 31890058 PMCID: PMC6928270 DOI: 10.1016/j.jdsr.2019.11.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 10/26/2019] [Accepted: 11/21/2019] [Indexed: 12/22/2022] Open
Abstract
The bioactivity of biomaterials is closely related to cell response in contact with them. However, shortly after their insertion, materials are soon covered with proteins that constitute the biological fluids, and which render the direct surface recognition by cells almost impossible. The control of protein adsorption at the interface is therefore desirable. Extracellular matrix proteins are of particular interest in this sense, due to their well-known ability to modulate cell behavior. Particularly, fibronectin plays a leading role, being present in both healthy and injured tissues undergoing healing and regeneration. The aim of the present work is to give an overview on fibronectin and on its involvement in the control of cell behavior providing evidence of its pivotal role in the control of cell adhesion, spreading, migration, proliferation and differentiation. A deep insight into methods to enrich biomaterials surface with fibronectin will be then discussed, as well as new cues on the possibility to design tailored platforms able to specifically retain fibronectin from the surrounding extracellular milieu.
Collapse
Affiliation(s)
- Ludovica Parisi
- Centro Universitario di Odontoiatria, Dipartimento di Medicina e Chirurgia, Università di Parma, Via Gramsci 14, 43126 Parma, Italy
- Labor für Orale Molekularbiologie, Klinik für Kieferorthopädie, Zahnmedizinische Klinik, Universität Bern, Freiburgstrasse 7, 3008 Bern, Switzerland
- Corresponding author. Present address: Labor für Orale Molekularbiologie, Klinik für Kieferorthopädie, Zahnmedizinische Kliniken, Universität Bern, Freiburgstrasse 7, CH-3010 Bern, Switzerland.
| | - Andrea Toffoli
- Centro Universitario di Odontoiatria, Dipartimento di Medicina e Chirurgia, Università di Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Benedetta Ghezzi
- Centro Universitario di Odontoiatria, Dipartimento di Medicina e Chirurgia, Università di Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Beatrice Mozzoni
- Centro Universitario di Odontoiatria, Dipartimento di Medicina e Chirurgia, Università di Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Simone Lumetti
- Centro Universitario di Odontoiatria, Dipartimento di Medicina e Chirurgia, Università di Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Guido M. Macaluso
- Centro Universitario di Odontoiatria, Dipartimento di Medicina e Chirurgia, Università di Parma, Via Gramsci 14, 43126 Parma, Italy
- Istituto dei Materiali per l’Elettronica e l’Elettromagnetismo, Consiglio Nazionale delle Ricerche, Parco Area delle Scienze 17/A, 43124 Parma, Italy
| |
Collapse
|
10
|
Bracey DN, Jinnah AH, Willey JS, Seyler TM, Hutchinson ID, Whitlock PW, Smith TL, Danelson KA, Emory CL, Kerr BA. Investigating the Osteoinductive Potential of a Decellularized Xenograft Bone Substitute. Cells Tissues Organs 2019; 207:97-113. [PMID: 31655811 PMCID: PMC6935535 DOI: 10.1159/000503280] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/09/2019] [Indexed: 12/14/2022] Open
Abstract
Bone grafting is the second most common tissue transplantation procedure worldwide. One of the alternative methods for bone repair under investigation is a tissue-engineered bone substitute. An ideal property of tissue-engineered bone substitutes is osteoinductivity, defined as the ability to stimulate primitive cells to differentiate into a bone-forming lineage. In the current study, we use a decellularization and oxidation protocol to produce a porcine bone scaffold and examine whether it possesses osteoinductive potential and can be used to create a tissue-engineered bone microenvironment. The decellularization protocol was patented by our lab and consists of chemical decellularization and oxidation steps using combinations of deionized water, trypsin, antimicrobials, peracetic acid, and triton-X100. To test if the bone scaffold was a viable host, preosteoblasts were seeded and analyzed for markers of osteogenic differentiation. The osteoinductive potential was observed in vitro with similar osteogenic markers being expressed in preosteoblasts seeded on the scaffolds and demineralized bone matrix. To assess these properties in vivo, scaffolds with and without preosteoblasts preseeded were subcutaneously implanted in mice for 4 weeks. MicroCT scanning revealed 1.6-fold increased bone volume to total volume ratio and 1.4-fold increase in trabecular thickness in scaffolds after implantation. The histological analysis demonstrates new bone formation and blood vessel formation with pentachrome staining demonstrating osteogenesis and angiogenesis, respectively, within the scaffold. Furthermore, CD31+ staining confirmed the endothelial lining of the blood vessels. These results demonstrate that porcine bone maintains its osteoinductive properties after the application of a patented decellularization and oxidation protocol developed in our laboratory. Future work must be performed to definitively prove osteogenesis of human mesenchymal stem cells, biocompatibility in large animal models, and osteoinduction/osseointegration in a relevant clinical model in vivo. The ability to create a functional bone microenvironment using decellularized xenografts will impact regenerative medicine, orthopedic reconstruction, and could be used in the research of multiple diseases.
Collapse
Affiliation(s)
- Daniel N. Bracey
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
| | - Alexander H. Jinnah
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
| | - Jeffrey S. Willey
- Wake Forest Baptist Medical Center, Radiation Oncology, Winston Salem, NC, USA
| | | | | | | | - Thomas L. Smith
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
| | - Kerry A. Danelson
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
| | - Cynthia L. Emory
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
| | - Bethany A. Kerr
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
- Virginia Tech-Wake Forest University School for Bioengineering and Sciences, Winston Salem, NC, USA
- Wake Forest School of Medicine, Cancer Biology, Winston Salem, NC, USA
| |
Collapse
|
11
|
Wu Q, Yang B, Cao C, Hu K, Wang P, Man Y. Therapeutic antibody directed osteogenic differentiation of induced pluripotent stem cell derived MSCs. Acta Biomater 2018; 74:222-235. [PMID: 29778895 DOI: 10.1016/j.actbio.2018.05.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 05/14/2018] [Accepted: 05/16/2018] [Indexed: 12/31/2022]
Abstract
Induced pluripotent stem cells (iPSCs) are regarded as a new cell source for regenerative medicine. Recent advances in tissue engineering have brought to light the therapeutic application of induced pluripotent stem cells (iPSCs) in bone defect repair. However, a safe and efficient way to differentiate iPSCs into osteogenic lineage remains to be a major challenge. Here we describe an approach using anti-BMP2 antibodies (Abs) to mediate osteogenic differentiation of iPSC-derived mesenchymal stromal cells (iMSCs). We first proved that 3G7 (an anti-BMP2 Ab) not only bound to BMP2, but also allowed the bound BMP2 to engage the BMP2 receptors on iMSCs. Subcutaneous implantation sites loaded with iMSCs + 3G7 group showed significant bone formation and vascularization in mice while those sites with exogenous BMP2 exhibited dystrophic calcification and significantly lower vascularization. Our in vitro study demonstrated that the anti-BMP2 Ab/BMP2 immune complex were capable of dictating the acquisition of osteogenic phenotype of iMSCs and subsequent mineralization. The study provided the first evidence of antibody-mediated differentiation of iMSCs and osseous regeneration in vivo. This novel strategy takes full advantage of the endogenous bioactive molecules for osseous regeneration and its potential therapeutic application is promising. STATEMENT OF SIGNIFICANCE Induced pluripotent stem cells (iPSCs) and its derived cells hold significant promise for the treatment of bone defects. In present study, we carried out the concept of antibody-mediated bone regeneration into the iPSC research for the first time. We demonstrated that anti-BMP2 Ab/BMP2 immune complex was capable of promoting osteogenic differentiation of iPSC-derived MSCs (iMSCs), likely through the classical BMP2/Smad1/Runx2 pathway. Subcutaneous co-delivery of iMSCs and anti-BMP2 Abs resulted in significant bone formation and vascularization. These findings suggested antibody mediated osteogenic differentiation may be a favorable approach for iPSC-based bone tissue engineering.
Collapse
|
12
|
Antibody-Mediated Osseous Regeneration for Bone Tissue Engineering in Canine Segmental Defects. BIOMED RESEARCH INTERNATIONAL 2018; 2018:9508721. [PMID: 29682573 PMCID: PMC5851338 DOI: 10.1155/2018/9508721] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 12/16/2017] [Accepted: 01/16/2018] [Indexed: 01/29/2023]
Abstract
Among many applications of therapeutic monoclonal antibodies (mAbs), a unique approach for regenerative medicine has entailed antibody-mediated osseous regeneration (AMOR). In an effort to identify a clinically relevant model of craniofacial defect, the present study investigated the efficacy of mAb specific for bone morphogenetic protein- (BMP-) 2 to repair canine segmental mandibular continuity defect model. Accordingly, a 15 mm unilateral segmental defect was created in mandible and fixated with a titanium plate. Anorganic bovine bone mineral with 10% collagen (ABBM-C) was functionalized with 25 μg/mL of either chimeric anti-BMP-2 mAb or isotype-matched mAb (negative control). Recombinant human (rh) BMP-2 served as positive control. Morphometric analyses were performed on computed tomography (CT) and histologic images. Bone densities within healed defect sites at 12 weeks after surgery were 1360.81 ± 10.52 Hounsfield Unit (HU), 1044.27 ± 141.16 HU, and 839.45 ± 179.41 HU, in sites with implanted anti-BMP-2 mAb, rhBMP-2, and isotype mAb groups, respectively. Osteoid bone formation in anti-BMP-2 mAb (42.99% ± 8.67) and rhBMP-2 (48.97% ± 2.96) groups was not significantly different but was higher (p < 0.05) than in sites with isotype control mAb (26.8% ± 5.35). In view of the long-term objective of translational application of AMOR in humans, the results of the present study demonstrated the feasibility of AMOR in a large clinically relevant animal model.
Collapse
|
13
|
Lin Y, An B, Bagheri M, Wang Q, Harden JL, Kaplan DL. Electrochemically Directed Assembly of Designer Coiled-Coil Telechelic Proteins. ACS Biomater Sci Eng 2017; 3:3195-3206. [PMID: 33445361 DOI: 10.1021/acsbiomaterials.7b00599] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We report the design and characterization of a de novo electrogelation protein comprising a central spider silk glue motif flanked by terminal pH-triggered coiled-coil domains. The coiled-coiled domains were designed to form intramolecular helix bundles below a sharply defined pH-trigger point (∼pH 5.3), whereas the spider silk glue protein, because of its substantial Glu content, serves both as an anionic electrophoretic transport element at neutral and elevated pH and as a disordered linker chain between the associated helix bundles at reduced pH. We show that in an electrochemical cell, a solution of these telechelic proteins migrates toward the anode where the terminal coiled-coil domains are triggered to form coiled-coil assemblies that act as transient cross-links for the e-gel state. Upon cessation of the current, the coiled-coil domains become denatured and the e-gel transforms back into a fluid solution of polypeptides in a fully reversible manner. This simplified triblock protein design mimics many of the characteristics of more complex electrogelation proteins, such as silk fibroin. As such, it provides some insight into possible general mechanisms of protein electrogelation. Moreover, this general class of electrogelation proteins has the potential for biomedical applications of electrochemically triggered gelation, such as externally switchable delivery of therapeutic cell and drugs from a responsive matrix.
Collapse
Affiliation(s)
- Yinan Lin
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Bo An
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Mehran Bagheri
- Department of Physics, University of Ottawa, Ontario K1N 6N5, Canada
| | - Qianrui Wang
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - James L Harden
- Department of Physics, University of Ottawa, Ontario K1N 6N5, Canada
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
14
|
Ansari S, Diniz IM, Chen C, Sarrion P, Tamayol A, Wu BM, Moshaverinia A. Human Periodontal Ligament- and Gingiva-derived Mesenchymal Stem Cells Promote Nerve Regeneration When Encapsulated in Alginate/Hyaluronic Acid 3D Scaffold. Adv Healthc Mater 2017; 6:10.1002/adhm.201700670. [PMID: 29076281 PMCID: PMC5813692 DOI: 10.1002/adhm.201700670] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 08/29/2017] [Indexed: 12/25/2022]
Abstract
Repair or regeneration of damaged nerves is still a challenging clinical task in reconstructive surgeries and regenerative medicine. Here, it is demonstrated that periodontal ligament stem cells (PDLSCs) and gingival mesenchymal stem cells (GMSCs) isolated from adult human periodontal and gingival tissues assume neuronal phenotype in vitro and in vivo via a subcutaneous transplantation model in nude mice. PDLSCs and GMSCs are encapsulated in a 3D scaffold based on alginate and hyaluronic acid hydrogels capable of sustained release of human nerve growth factor (NGF). The elasticity of the hydrogels affects the proliferation and differentiation of encapsulated MSCs within scaffolds. Moreover, it is observed that PDLSCs and GMSCs are stained positive for βIII-tubulin, while exhibiting high levels of gene expression related to neurogenic differentiation (βIII-tubulin and glial fibrillary acidic protein) via quantitative polymerase chain reaction (qPCR). Western blot analysis shows the importance of elasticity of the matrix and the presence of NGF in the neurogenic differentiation of encapsulated MSCs. In vivo, immunofluorescence staining for neurogenic specific protein markers confirms islands of dense positively stained structures inside transplanted hydrogels. As far as it is known, this study is the first demonstration of the application of PDLSCs and GMSCs as promising cell therapy candidates for nerve regeneration.
Collapse
Affiliation(s)
- Sahar Ansari
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA, 90095, USA
| | - Ivana M Diniz
- Faculdade de Odontologia da UFMG, Departamento de Odontologia Restauradora, Av. Antonio Carlos, 6627, Belo Horizonte, MG, 31270-910, Brazil
| | - Chider Chen
- School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, PA, 19104, USA
| | - Patricia Sarrion
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA, 90095, USA
| | - Ali Tamayol
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, NE 68508, Lincoln
| | - Benjamin M Wu
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA, 90095, USA
| | - Alireza Moshaverinia
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA, 90095, USA
| |
Collapse
|
15
|
Three-dimensional macroporous materials for tissue engineering of craniofacial bone. Br J Oral Maxillofac Surg 2017; 55:875-891. [PMID: 29056355 DOI: 10.1016/j.bjoms.2017.09.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 09/18/2017] [Indexed: 12/15/2022]
Abstract
Repair of critical-size defects caused by trauma, removal of a tumour, or congenital abnormalities is a challenge in the craniomaxillofacial region because of the limitations associated with treatment. We have reviewed research papers and updated information relevant to the various types of macroporous scaffolds. We have included papers on several biomaterials and their use in various craniofacial defects such as mandibular, calvarial, and others, as well as the latest technological developments such as 3-dimensional printed scaffolds. We selected all papers about scaffolds, stem cells, and growth factors for review. Initial selection was by review of titles and abstracts, and the full texts of potentially suitable articles were then assessed. Methods of tissue engineering for repair of critical-size defects in the craniofacial bones seem to be viable options for surgical treatment in the future. Macroporous scaffolds with interconnected pores are of great value in regeneration of bone in the craniofacial region. In recent years, various natural or synthetic materials, or both, have been developed, on which macroporous scaffolds can be based. In this review we present a review on the various types of three-dimensional macroporous scaffolds that have been developed in recent years, and evaluate their potential for regeneration of craniofacial bone.
Collapse
|
16
|
Guo L, Min S, Su Y, Tang J, Du J, Goh BT, Saigo L, Wang S, Ansari S, Moshaverinia A, Zadeh HH, Liu Y. Collagen sponge functionalized with chimeric anti-BMP-2 monoclonal antibody mediates repair of nonunion tibia defects in a nonhuman primate model: An exploratory study. J Biomater Appl 2017; 32:425-432. [DOI: 10.1177/0885328217733262] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Lijia Guo
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
- Department of Orthodontics, Capital Medical University School of Stomatology, Beijing, China
| | - Seiko Min
- Laboratory for Immunoregulation and Tissue Engineering (LITE), Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Yingying Su
- Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University School of Stomatology, Beijing, China
| | - Jianxia Tang
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital, Central South University, Changsha, Hunan, China
| | - Juan Du
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Bee Tin Goh
- Department of Oral & Maxillofacial Surgery, National Dental Centre, Singapore
| | - Leonardo Saigo
- Department of Oral & Maxillofacial Surgery, National Dental Centre, Singapore
| | - Songlin Wang
- Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Sahar Ansari
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Alireza Moshaverinia
- Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Homayoun H Zadeh
- Laboratory for Immunoregulation and Tissue Engineering (LITE), Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| |
Collapse
|
17
|
Lee JC, Volpicelli EJ. Bioinspired Collagen Scaffolds in Cranial Bone Regeneration: From Bedside to Bench. Adv Healthc Mater 2017; 6:10.1002/adhm.201700232. [PMID: 28585295 PMCID: PMC5831258 DOI: 10.1002/adhm.201700232] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/11/2017] [Indexed: 12/24/2022]
Abstract
Calvarial defects are common reconstructive dilemmas secondary to a variety of etiologies including traumatic brain injury, cerebrovascular disease, oncologic resection, and congenital anomalies. Reconstruction of the calvarium is generally undertaken for the purposes of cerebral protection, contour restoration for psychosocial well-being, and normalization of neurological dysfunction frequently found in patients with massive cranial defects. Current methods for reconstruction using autologous grafts, allogeneic grafts, or alloplastic materials have significant drawbacks that are unique to each material. The combination of wide medical relevance and the need for a better clinical solution render defects of the cranial skeleton an ideal target for development of regenerative strategies focused on calvarial bone. With the improved understanding of the instructive properties of tissue-specific extracellular matrices and the advent of precise nanoscale modulation in materials science, strategies in regenerative medicine have shifted in paradigm. Previously considered to be simple carriers of stem cells and growth factors, increasing evidence exists for differential materials directing lineage specific differentiation of progenitor cells and tissue regeneration. In this work, we review the clinical challenges for calvarial reconstruction, the anatomy and physiology of bone, and extracellular matrix-inspired, collagen-based materials that have been tested for in vivo cranial defect healing.
Collapse
Affiliation(s)
- Justine C Lee
- Greater Los Angeles Veterans Affairs Research Service, Los Angeles, California
- University of California Los Angeles Division of Plastic and Reconstructive Surgery, Los Angeles, California
| | - Elizabeth J Volpicelli
- Greater Los Angeles Veterans Affairs Research Service, Los Angeles, California
- University of California Los Angeles Division of Plastic and Reconstructive Surgery, Los Angeles, California
| |
Collapse
|
18
|
Yao Q, Sandhurst ES, Liu Y, Sun H. BBP-Functionalized Biomimetic Nanofibrous Scaffold Can Capture BMP2 and Promote Osteogenic Differentiation. J Mater Chem B 2017; 5:5196-5205. [PMID: 29250330 DOI: 10.1039/c7tb00744b] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Bone morphogenetic proteins (BMPs, e.g., BMP2 and 7) are potent mediators for bone repair, however, their clinical use has been limited by their safety and cost-effectiveness. Therefore, innovative strategies that can improve the efficacy of BMPs, and thereby, use a lower dose of exogenous BMPs are highly desired. Inspired by the natural interaction between extracellular matrix (ECM) and growth factors, we hypothesize that bone matrix-mimicking nanofibrous scaffold functionalized with BMP binding moieties can selectively capture and stabilize BMPs, and thereby, promote BMP-induced osteogenic differentiation. To test our hypothesis, a gelatin nanofibrous scaffold was fabricated using thermally induced phase separation together with a porogen leaching technique (TIPS&P) and functionalized by a BMP-binding peptide (BBP) through cross-linking. Our data indicated that BBP decoration largely improved the BMP2 binding and retention capacity of the nanofibrous scaffolds without compromising their macro/microstructure and mechanical properties. Importantly, the BBP-functionalized gelatin scaffolds were able to significantly promote BMP2-induced osteogenic differentiation. Moreover, BBP alone was able to significantly stimulate endogenous BMP2 expression and improve osteogenic differentiation. Compared to other affinity-based drug delivery strategies, e.g., heparin and antibody-mediated growth factor delivering techniques, we expect BBP-functionalized scaffolds will be a safer, more feasible and selective strategy for endogenous BMP stimulating and binding. Therefore, our data suggests a promising application of using the BBP-decorated gelatin nanofibrous scaffold to stimulate/capture BMPs and promote endogenous bone formation in situ in contrast to relying on the administration of high doses of exogenous BMPs and transplantation of cells.
Collapse
Affiliation(s)
- Qingqing Yao
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD 57107, USA.,BioSNTR, Sioux Falls, SD 57107, USA.,School of Ophthalmology and Optometry, Wenzhou Medical University, 270 Xueyuan Xi Road, Wenzhou, Zhejiang 325027, China.,Institute of Advanced Materials for Nano-Bio Applications, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Eric S Sandhurst
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD 57107, USA.,BioSNTR, Sioux Falls, SD 57107, USA
| | - Yangxi Liu
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD 57107, USA.,BioSNTR, Sioux Falls, SD 57107, USA
| | - Hongli Sun
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD 57107, USA.,BioSNTR, Sioux Falls, SD 57107, USA
| |
Collapse
|
19
|
Collagen Sponge Functionalized with Chimeric Anti-BMP-2 Monoclonal Antibody Mediates Repair of Critical-Size Mandibular Continuity Defects in a Nonhuman Primate Model. BIOMED RESEARCH INTERNATIONAL 2017; 2017:8094152. [PMID: 28401163 PMCID: PMC5376406 DOI: 10.1155/2017/8094152] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/05/2017] [Accepted: 01/19/2017] [Indexed: 11/25/2022]
Abstract
Antibody-mediated osseous regeneration (AMOR) has been introduced by our research group as a tissue engineering approach to capture of endogenous growth factors through the application of specific monoclonal antibodies (mAbs) immobilized on a scaffold. Specifically, anti-Bone Morphogenetic Protein- (BMP-) 2 mAbs have been demonstrated to be efficacious in mediating bone repair in a number of bone defects. The present study sought to investigate the application of AMOR for repair of mandibular continuity defect in nonhuman primates. Critical-sized mandibular continuity defects were created in Macaca fascicularis locally implanted with absorbable collagen sponges (ACS) functionalized with chimeric anti-BMP-2 mAb or isotype control mAb. 2D and 3D analysis of cone beam computed tomography (CBCT) imaging demonstrated increased bone density and volume observed within mandibular continuity defects implanted with collagen scaffolds functionalized with anti-BMP-2 mAb, compared with isotype-matched control mAb. Both CBCT imaging and histologic examination demonstrated de novo bone formation that was in direct apposition to the margins of the resected bone. It is hypothesized that bone injury may be necessary for AMOR. This is evidenced by de novo bone formation adjacent to resected bone margins, which may be the source of endogenous BMPs captured by anti-BMP-2 mAb, in turn mediating bone repair.
Collapse
|
20
|
Wang T, Wu D, Li Y, Li W, Zhang S, Hu K, Zhou H. Substance P incorporation in calcium phosphate cement for dental alveolar bone defect restoration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 69:546-53. [DOI: 10.1016/j.msec.2016.07.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 06/30/2016] [Accepted: 07/05/2016] [Indexed: 02/06/2023]
|
21
|
Ansari S, Chen C, Xu X, Annabi N, Zadeh HH, Wu BM, Khademhosseini A, Shi S, Moshaverinia A. Muscle Tissue Engineering Using Gingival Mesenchymal Stem Cells Encapsulated in Alginate Hydrogels Containing Multiple Growth Factors. Ann Biomed Eng 2016; 44:1908-20. [PMID: 27009085 DOI: 10.1007/s10439-016-1594-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 03/14/2016] [Indexed: 12/15/2022]
Abstract
Repair and regeneration of muscle tissue following traumatic injuries or muscle diseases often presents a challenging clinical situation. If a significant amount of tissue is lost the native regenerative potential of skeletal muscle will not be able to grow to fill the defect site completely. Dental-derived mesenchymal stem cells (MSCs) in combination with appropriate scaffold material, present an advantageous alternative therapeutic option for muscle tissue engineering in comparison to current treatment modalities available. To date, there has been no report on application of gingival mesenchymal stem cells (GMSCs) in three-dimensional scaffolds for muscle tissue engineering. The objectives of the current study were to develop an injectable 3D RGD-coupled alginate scaffold with multiple growth factor delivery capacity for encapsulating GMSCs, and to evaluate the capacity of encapsulated GMSCs to differentiate into myogenic tissue in vitro and in vivo where encapsulated GMSCs were transplanted subcutaneously into immunocompromised mice. The results demonstrate that after 4 weeks of differentiation in vitro, GMSCs as well as the positive control human bone marrow mesenchymal stem cells (hBMMSCs) exhibited muscle cell-like morphology with high levels of mRNA expression for gene markers related to muscle regeneration (MyoD, Myf5, and MyoG) via qPCR measurement. Our quantitative PCR analyzes revealed that the stiffness of the RGD-coupled alginate regulates the myogenic differentiation of encapsulated GMSCs. Histological and immunohistochemical/fluorescence staining for protein markers specific for myogenic tissue confirmed muscle regeneration in subcutaneous transplantation in our in vivo animal model. GMSCs showed significantly greater capacity for myogenic regeneration in comparison to hBMMSCs (p < 0.05). Altogether, our findings confirmed that GMSCs encapsulated in RGD-modified alginate hydrogel with multiple growth factor delivery capacity is a promising candidate for muscle tissue engineering.
Collapse
Affiliation(s)
- Sahar Ansari
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Chider Chen
- School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xingtian Xu
- Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Nasim Annabi
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA.,Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Homayoun H Zadeh
- Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Benjamin M Wu
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prothodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Songtao Shi
- School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alireza Moshaverinia
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prothodontics, School of Dentistry, University of California, Los Angeles, CA, USA.
| |
Collapse
|
22
|
Diniz IMA, Chen C, Ansari S, Zadeh HH, Moshaverinia M, Chee D, Marques MM, Shi S, Moshaverinia A. Gingival Mesenchymal Stem Cell (GMSC) Delivery System Based on RGD-Coupled Alginate Hydrogel with Antimicrobial Properties: A Novel Treatment Modality for Peri-Implantitis. J Prosthodont 2016; 25:105-15. [PMID: 26216081 PMCID: PMC4729657 DOI: 10.1111/jopr.12316] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2015] [Indexed: 11/26/2022] Open
Abstract
PURPOSE Peri-implantitis is one of the most common inflammatory complications in dental implantology. Similar to periodontitis, in peri-implantitis, destructive inflammatory changes take place in the tissues surrounding a dental implant. Bacterial flora at the failing implant sites resemble the pathogens in periodontal disease and consist of Gram-negative anaerobic bacteria including Aggregatibacter actinomycetemcomitans (Aa). Here we demonstrate the effectiveness of a silver lactate (SL)-containing RGD-coupled alginate hydrogel scaffold as a promising stem cell delivery vehicle with antimicrobial properties. MATERIALS AND METHODS Gingival mesenchymal stem cells (GMSCs) or human bone marrow mesenchymal stem cells (hBMMSCs) were encapsulated in SL-loaded alginate hydrogel microspheres. Stem cell viability, proliferation, and osteo-differentiation capacity were analyzed. RESULTS Our results showed that SL exhibited antimicrobial properties against Aa in a dose-dependent manner, with 0.50 mg/ml showing the greatest antimicrobial properties while still maintaining cell viability. At this concentration, SL-containing alginate hydrogel was able to inhibit Aa growth on the surface of Ti discs and significantly reduce the bacterial load in Aa suspensions. Silver ions were effectively released from the SL-loaded alginate microspheres for up to 2 weeks. Osteogenic differentiation of GMSCs and hBMMSCs encapsulated in the SL-loaded alginate microspheres were confirmed by the intense mineral matrix deposition and high expression of osteogenesis-related genes. CONCLUSION Taken together, our findings confirm that GMSCs encapsulated in RGD-modified alginate hydrogel containing SL show promise for bone tissue engineering with antimicrobial properties against Aa bacteria in vitro.
Collapse
Affiliation(s)
- Ivana M. A. Diniz
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA
- Restorative Dentistry Department, School of Dentistry, Universidade de São Paulo, São Paulo, Brazil
| | - Chider Chen
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania
| | - Sahar Ansari
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA
- Laboratory for Immunoregulation and Tissue Engineering (LITE), Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA
| | - Homayoun H. Zadeh
- Laboratory for Immunoregulation and Tissue Engineering (LITE), Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA
| | - Maryam Moshaverinia
- Department of Oral and Maxillofacial Medicine, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Daniel Chee
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA
| | - Márcia M. Marques
- Restorative Dentistry Department, School of Dentistry, Universidade de São Paulo, São Paulo, Brazil
| | - Songtao Shi
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania
| | - Alireza Moshaverinia
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA
| |
Collapse
|
23
|
Ansari S, Phark JH, Duarte S, Paulino da Silva M, Sharifzadeh N, Moshaverinia A, Zadeh HH. Biomechanical analysis of engineered bone with anti-BMP2 antibody immobilized on different scaffolds. J Biomed Mater Res B Appl Biomater 2015; 104:1465-73. [PMID: 26252572 DOI: 10.1002/jbm.b.33492] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 06/16/2015] [Accepted: 07/18/2015] [Indexed: 11/09/2022]
Abstract
Recently we have demonstrated the ability of monoclonal antibodies (mAb) specific for bone morphogenetic protein (BMP)-2 immobilized on different scaffolds to mediate bone formation, a process referred to as Antibody Mediated Osseous Regeneration (AMOR). One of the key properties of regenerated bone is its biomechanical strength, in particular in load-bearing areas. This study sought to test the hypothesis that the biomechanical strength of regenerated bone depends of the mode of regeneration, as well as the scaffold used. Four different scaffolds, namely titanium granules (Ti), alginate hydrogel, anorganic bovine bone mineral (ABBM), and absorbable collagen sponge (ACS) were functionalized with anti-BMP-2 or isotype control mAb and implanted into rat critical-size calvarial defects. The morphology, density and strength of the regenerated bone were evaluated after 8 weeks. Results demonstrated that scaffolds functionalized with anti-BMP-2 mAb exhibited varying degrees of bone volume and density. Ti and ABBM achieved the highest bone volume, density, and strength of bone. When anti-BMP-2 mAb was immobilized on Ti or ABBM, the strength of the regenerated bone were 80% and 77% of native bone respectively, compared with 60% of native bone in sites implanted with rh-BMP-2. Control interventions with isotype mAb did not promote considerable bone regeneration and exhibited significantly lower mechanical properties. SEM analysis showed specimens immobilized with anti-BMP-2 mAb formed new bone with organized structure bridging the crack areas. Altogether, the present data demonstrated that the morphological and mechanical properties of bone bioengineered through AMOR could approximate that of native bone, when appropriate scaffolds are used. © 2015 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 104B: 1465-1473, 2016.
Collapse
Affiliation(s)
- Sahar Ansari
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA
| | - Jin-Ho Phark
- Division of Restorative Sciences, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA
| | - Sillas Duarte
- Division of Restorative Sciences, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA
| | - Maike Paulino da Silva
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, Sao Paulo, Brazil
| | - Navid Sharifzadeh
- Division of Periodontology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA
| | - Alireza Moshaverinia
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA
| | - Homayoun H Zadeh
- Division of Periodontology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA.
| |
Collapse
|
24
|
Ansari S, Freire M, Choi MG, Tavari A, Almohaimeed M, Moshaverinia A, Zadeh HH. Effects of the orientation of anti-BMP2 monoclonal antibody immobilized on scaffold in antibody-mediated osseous regeneration. J Biomater Appl 2015; 30:558-67. [PMID: 26184354 DOI: 10.1177/0885328215594704] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Recently, we have shown that anti-BMP2 monoclonal antibodies (mAbs) can trap endogenous osteogenic BMP ligands, which can in turn mediate osteodifferentiation of progenitor cells. The effectiveness of this strategy requires the availability of the anti-BMP-2 monoclonal antibodies antigen-binding sites for anti-BMP-2 monoclonal antibodies to bind to the scaffold through a domain that will leave its antigen-binding region exposed and available for binding to an osteogenic ligand. We examined whether antibodies bound to a scaffold by passive adsorption versus through Protein G as a linker will exhibit differences in mediating bone formation. In vitro anti-BMP-2 monoclonal antibodies was immobilized on absorbable collagen sponge (ACS) with Protein G as a linker to bind the antibody through its Fc region and implanted into rat calvarial defects. The biomechanical strength of bone regenerated by absorbable collagen sponge/Protein G/anti-BMP-2 monoclonal antibodies immune complex was compared to ACS/anti-BMP-2 monoclonal antibodies or ACS/Protein G/isotype mAb control group. Results demonstrated higher binding of anti-BMP-2 monoclonal antibodies/BMPs to C2C12 cells, when the mAb was initially attached to recombinant Protein G or Protein G-coupled microbeads. After eight weeks, micro-CT and histomorphometric analyses revealed increased bone formation within defects implanted with absorbable collagen sponge/Protein G/anti-BMP-2 monoclonal antibodies compared with defects implanted with absorbable collagen sponge/anti-BMP-2 monoclonal antibodies (p < 0.05). Confocal laser scanning microscopy (CLSM) confirmed increased BMP-2, -4, and -7 detection in sites implanted with absorbable collagen sponge/Protein G/anti-BMP-2 monoclonal antibodies in vivo. Biomechanical analysis revealed the regenerated bone in sites with Protein G/anti-BMP-2 monoclonal antibodies had higher mechanical strength in comparison to anti-BMP-2 monoclonal antibodies. The negative control group, Protein G/isotype mAb, did not promote bone regeneration and exhibited significantly lower mechanical properties (p < 0.05). Altogether, our results demonstrated that application of Protein G as a linker to adsorb anti-BMP-2 monoclonal antibodies onto the scaffold was accompanied by increased in vitro binding of the anti-BMP-2 mAb/BMP immune complex to BMP-receptor positive cell, as well as increased volume and strength of de novo bone formation in vivo.
Collapse
Affiliation(s)
- Sahar Ansari
- Laboratory for Immunoregulation and Tissue Engineering (LITE), Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA, USA Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA, USA
| | - Marcelo Freire
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Moon G Choi
- Laboratory for Immunoregulation and Tissue Engineering (LITE), Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA, USA
| | - Azadeh Tavari
- Laboratory for Immunoregulation and Tissue Engineering (LITE), Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA, USA
| | - Mohammad Almohaimeed
- Dental Research Center (DRC), Tissue Engineering and Biomaterials Research Unit (TEBRU), College of Dentistry, Qassim University, Qassim, Saudi Arabia
| | - Alireza Moshaverinia
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA, USA
| | - Homayoun H Zadeh
- Laboratory for Immunoregulation and Tissue Engineering (LITE), Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
25
|
Moshaverinia A, Chen C, Xu X, Ansari S, Zadeh HH, Schricker SR, Paine ML, Moradian-Oldak J, Khademhosseini A, Snead ML, Shi S. Regulation of the Stem Cell-Host Immune System Interplay Using Hydrogel Coencapsulation System with an Anti-Inflammatory Drug. ADVANCED FUNCTIONAL MATERIALS 2015; 25:2296-2307. [PMID: 26120294 PMCID: PMC4478611 DOI: 10.1002/adfm.201500055] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The host immune system is known to influence mesenchymal stem cell (MSC)-mediated bone tissue regeneration. However, the therapeutic capacity of hydrogel biomaterial to modulate the interplay between MSCs and T-lymphocytes is unknown. Here it is shown that encapsulating hydrogel affects this interplay when used to encapsulate MSCs for implantation by hindering the penetration of pro-inflammatory cells and/or cytokines, leading to improved viability of the encapsulated MSCs. This combats the effects of the host pro-inflammatory T-lymphocyte-induced nuclear factor kappaB pathway, which can reduce MSC viability through the CASPASE-3 and CAS-PASE-8 associated proapoptotic cascade, resulting in the apoptosis of MSCs. To corroborate rescue of engrafted MSCs from the insult of the host immune system, the incorporation of the anti-inflammatory drug indomethacin into the encapsulating alginate hydrogel further regulates the local microenvironment and prevents pro-inflammatory cytokine-induced apoptosis. These findings suggest that the encapsulating hydrogel can regulate the MSC-host immune cell interplay and direct the fate of the implanted MSCs, leading to enhanced tissue regeneration.
Collapse
Affiliation(s)
- Alireza Moshaverinia
- Center for Craniofacial Molecular Biology (CCMB), Ostrow School of Dentistry, University of Southern California, 2250 Alcazar St, Los Angeles, CA 90033, USA
| | - Chider Chen
- School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, PA 19104, USA
| | - Xingtian Xu
- Center for Craniofacial Molecular Biology (CCMB), Ostrow School of Dentistry, University of Southern California, 2250 Alcazar St, Los Angeles, CA 90033, USA
| | - Sahar Ansari
- Center for Craniofacial Molecular Biology (CCMB), Ostrow School of Dentistry, University of Southern California, 2250 Alcazar St, Los Angeles, CA 90033, USA
| | - Homayoun H. Zadeh
- Center for Craniofacial Molecular Biology (CCMB), Ostrow School of Dentistry, University of Southern California, 2250 Alcazar St, Los Angeles, CA 90033, USA
| | - Scott R. Schricker
- College of Dentistry, Ohio State University, 305 W 12th Ave, Columbus, OH 43210, USA
| | - Michael L. Paine
- Center for Craniofacial Molecular Biology (CCMB), Ostrow School of Dentistry, University of Southern California, 2250 Alcazar St, Los Angeles, CA 90033, USA
| | - Janet Moradian-Oldak
- Center for Craniofacial Molecular Biology (CCMB), Ostrow School of Dentistry, University of Southern California, 2250 Alcazar St, Los Angeles, CA 90033, USA
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center Harvard Medical School 65 Landsdowne St, Rm 252, Cambridge, MA 02139, USA
| | - Malcolm L. Snead
- Center for Craniofacial Molecular Biology (CCMB), Ostrow School of Dentistry, University of Southern California, 2250 Alcazar St, Los Angeles, CA 90033, USA
| | - Songtao Shi
- School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, PA 19104, USA
| |
Collapse
|
26
|
Biomimetic approaches in bone tissue engineering: Integrating biological and physicomechanical strategies. Adv Drug Deliv Rev 2015; 84:1-29. [PMID: 25236302 DOI: 10.1016/j.addr.2014.09.005] [Citation(s) in RCA: 294] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Revised: 09/01/2014] [Accepted: 09/05/2014] [Indexed: 02/06/2023]
Abstract
The development of responsive biomaterials capable of demonstrating modulated function in response to dynamic physiological and mechanical changes in vivo remains an important challenge in bone tissue engineering. To achieve long-term repair and good clinical outcomes, biologically responsive approaches that focus on repair and reconstitution of tissue structure and function through drug release, receptor recognition, environmental responsiveness and tuned biodegradability are required. Traditional orthopedic materials lack biomimicry, and mismatches in tissue morphology, or chemical and mechanical properties ultimately accelerate device failure. Multiple stimuli have been proposed as principal contributors or mediators of cell activity and bone tissue formation, including physical (substrate topography, stiffness, shear stress and electrical forces) and biochemical factors (growth factors, genes or proteins). However, optimal solutions to bone regeneration remain elusive. This review will focus on biological and physicomechanical considerations currently being explored in bone tissue engineering.
Collapse
|
27
|
Zhang J, Zhou A, Deng A, Yang Y, Gao L, Zhong Z, Yang S. Pore architecture and cell viability on freeze dried 3D recombinant human collagen-peptide (RHC)–chitosan scaffolds. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2015; 49:174-182. [DOI: 10.1016/j.msec.2014.12.076] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 11/19/2014] [Accepted: 12/22/2014] [Indexed: 12/25/2022]
|
28
|
Diniz IMA, Chen C, Xu X, Ansari S, Zadeh HH, Marques MM, Shi S, Moshaverinia A. Pluronic F-127 hydrogel as a promising scaffold for encapsulation of dental-derived mesenchymal stem cells. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2015; 26:153. [PMID: 25773231 PMCID: PMC4477746 DOI: 10.1007/s10856-015-5493-4] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 02/09/2015] [Indexed: 05/16/2023]
Abstract
Dental-derived mesenchymal stem cells (MSCs) provide an advantageous therapeutic option for tissue engineering due to their high accessibility and bioavailability. However, delivering MSCs to defect sites while maintaining a high MSC survival rate is still a critical challenge in MSC-mediated tissue regeneration. Here, we tested the osteogenic and adipogenic differentiation capacity of dental pulp stem cells (DPSCs) in a thermoreversible Pluronic F127 hydrogel scaffold encapsulation system in vitro. DPSCs were encapsulated in Pluronic (®) F-127 hydrogel and stem cell viability, proliferation and differentiation into adipogenic and osteogenic tissues were evaluated. The degradation profile and swelling kinetics of the hydrogel were also analyzed. Our results confirmed that Pluronic F-127 is a promising and non-toxic scaffold for encapsulation of DPSCs as well as control human bone marrow MSCs (hBMMSCs), yielding high stem cell viability and proliferation. Moreover, after 2 weeks of differentiation in vitro, DPSCs as well as hBMMSCs exhibited high levels of mRNA expression for osteogenic and adipogenic gene markers via PCR analysis. Our histochemical staining further confirmed the ability of Pluronic F-127 to direct the differentiation of these stem cells into osteogenic and adipogenic tissues. Furthermore, our results revealed that Pluronic F-127 has a dense tubular and reticular network morphology, which contributes to its high permeability and solubility, consistent with its high degradability in the tested conditions. Altogether, our findings demonstrate that Pluronic F-127 is a promising scaffold for encapsulation of DPSCs and can be considered for cell delivery purposes in tissue engineering.
Collapse
Affiliation(s)
- Ivana M. A. Diniz
- />Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, 2250 Alcazar Street - CSA 103, Los Angeles, CA 90033 USA
- />Restorative Dentistry Department, School of Dentistry, Universidade de São Paulo, São Paulo, SP Brazil
| | - Chider Chen
- />Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, 2250 Alcazar Street - CSA 103, Los Angeles, CA 90033 USA
| | - Xingtian Xu
- />Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, 2250 Alcazar Street - CSA 103, Los Angeles, CA 90033 USA
| | - Sahar Ansari
- />Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, 2250 Alcazar Street - CSA 103, Los Angeles, CA 90033 USA
- />Laboratory for Immunoregulation and Tissue Engineering (LITE), Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA USA
| | - Homayoun H. Zadeh
- />Laboratory for Immunoregulation and Tissue Engineering (LITE), Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA USA
| | - Márcia M. Marques
- />Restorative Dentistry Department, School of Dentistry, Universidade de São Paulo, São Paulo, SP Brazil
| | - Songtao Shi
- />Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, 2250 Alcazar Street - CSA 103, Los Angeles, CA 90033 USA
| | - Alireza Moshaverinia
- />Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, 2250 Alcazar Street - CSA 103, Los Angeles, CA 90033 USA
| |
Collapse
|
29
|
Application of AMOR in craniofacial rabbit bone bioengineering. BIOMED RESEARCH INTERNATIONAL 2015; 2015:628769. [PMID: 25705677 PMCID: PMC4325208 DOI: 10.1155/2015/628769] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 08/09/2014] [Indexed: 12/17/2022]
Abstract
Endogenous molecular and cellular mediators modulate tissue repair and regeneration. We have recently described antibody mediated osseous regeneration (AMOR) as a novel strategy for bioengineering bone in rat calvarial defect. This entails application of anti-BMP-2 antibodies capable of in vivo capturing of endogenous osteogenic BMPs (BMP-2, BMP-4, and BMP-7). The present study sought to investigate the feasibility of AMOR in other animal models. To that end, we examined the efficacy of a panel of anti-BMP-2 monoclonal antibodies (mAbs) and a polyclonal Ab immobilized on absorbable collagen sponge (ACS) to mediate bone regeneration within rabbit calvarial critical size defects. After 6 weeks, de novo bone formation was demonstrated by micro-CT imaging, histology, and histomorphometric analysis. Only certain anti-BMP-2 mAb clones mediated significant in vivo bone regeneration, suggesting that the epitopes with which anti-BMP-2 mAbs react are critical to AMOR. Increased localization of BMP-2 protein and expression of osteocalcin were observed within defects, suggesting accumulation of endogenous BMP-2 and/or increased de novo expression of BMP-2 protein within sites undergoing bone repair by AMOR. Considering the ultimate objective of translation of this therapeutic strategy in humans, preclinical studies will be necessary to demonstrate the feasibility of AMOR in progressively larger animal models.
Collapse
|
30
|
Immobilization of murine anti-BMP-2 monoclonal antibody on various biomaterials for bone tissue engineering. BIOMED RESEARCH INTERNATIONAL 2014; 2014:940860. [PMID: 25147826 PMCID: PMC4132312 DOI: 10.1155/2014/940860] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 07/02/2014] [Indexed: 11/18/2022]
Abstract
Biomaterials are widely used as scaffolds for tissue engineering. We have developed a strategy for bone tissue engineering that entails application of immobilized anti-BMP-2 monoclonal antibodies (mAbs) to capture endogenous BMPs in vivo and promote antibody-mediated osseous regeneration (AMOR). The purpose of the current study was to compare the efficacy of immobilization of a specific murine anti-BMP-2 mAb on three different types of biomaterials and to evaluate their suitability as scaffolds for AMOR. Anti-BMP-2 mAb or isotype control mAb was immobilized on titanium (Ti) microbeads, alginate hydrogel, and ACS. The treated biomaterials were surgically implanted in rat critical-sized calvarial defects. After 8 weeks, de novo bone formation was assessed using micro-CT and histomorphometric analyses. Results showed de novo bone regeneration with all three scaffolds with immobilized anti-BMP-2 mAb, but not isotype control mAb. Ti microbeads showed the highest volume of bone regeneration, followed by ACS. Alginate showed the lowest volume of bone. Localization of BMP-2, -4, and -7 antigens was detected on all 3 scaffolds with immobilized anti-BMP-2 mAb implanted in calvarial defects. Altogether, these data suggested a potential mechanism for bone regeneration through entrapment of endogenous BMP-2, -4, and -7 proteins leading to bone formation using different types of scaffolds via AMOR.
Collapse
|