1
|
Le Pennec J, Makshakova O, Nevola P, Fouladkar F, Gout E, Machillot P, Friedel-Arboleas M, Picart C, Perez S, Vortkamp A, Vivès RR, Migliorini E. Glycosaminoglycans exhibit distinct interactions and signaling with BMP2 according to their nature and localization. Carbohydr Polym 2024; 341:122294. [PMID: 38876708 DOI: 10.1016/j.carbpol.2024.122294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/07/2024] [Accepted: 05/17/2024] [Indexed: 06/16/2024]
Abstract
The role of glycosaminoglycans (GAGs) in modulating bone morphogenetic protein (BMP) signaling represents a recent and underexplored area. Conflicting reports suggest a dual effect: some indicate a positive influence, while others demonstrate a negative impact. This duality suggests that the localization of GAGs (either at the cell surface or within the extracellular matrix) or the specific type of GAG may dictate their signaling role. The precise sulfation patterns of heparan sulfate (HS) responsible for BMP2 binding remain elusive. BMP2 exhibits a preference for binding to HS over other GAGs. Using well-characterized biomaterials mimicking the extracellular matrix, our research reveals that HS promotes BMP2 signaling in the extracellular space, contrary to chondroitin sulfate (CS), which enhances BMP2 bioactivity at the cell surface. Further observations indicate that a central IdoA (2S)-GlcNS (6S) tri-sulfated motif within HS hexasaccharides enhances binding. Nevertheless, BMP2 exhibits a degree of adaptability to various HS sulfation types and sequences. Molecular dynamic simulations attribute this adaptability to the BMP2 N-terminal end flexibility. Our findings illustrate the complex interplay between GAGs and BMP signaling, highlighting the importance of localization and specific sulfation patterns. This understanding has implications for the development of biomaterials with tailored properties for therapeutic applications targeting BMP signaling pathways.
Collapse
Affiliation(s)
- Jean Le Pennec
- Univ. Grenoble Alpes, INSERM, CEA, CNRS, U1292 Biosanté, EMR 5000, Grenoble, France
| | - Olga Makshakova
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, Synthetic Biology of Signalling Processes Lab, University of Freiburg, 79104 Freiburg, Germany
| | - Paola Nevola
- Univ. Grenoble Alpes, INSERM, CEA, CNRS, U1292 Biosanté, EMR 5000, Grenoble, France; Dipartimento di Ingegneria Chimica dei Materiali e della Produzione Industriale, University of Naples Federico II, Napoli, Italy
| | - Farah Fouladkar
- Univ. Grenoble Alpes, INSERM, CEA, CNRS, U1292 Biosanté, EMR 5000, Grenoble, France
| | - Evelyne Gout
- Univ. Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | - Paul Machillot
- Univ. Grenoble Alpes, INSERM, CEA, CNRS, U1292 Biosanté, EMR 5000, Grenoble, France
| | | | - Catherine Picart
- Univ. Grenoble Alpes, INSERM, CEA, CNRS, U1292 Biosanté, EMR 5000, Grenoble, France
| | - Serge Perez
- Univ. Grenoble Alpes, CNRS, Centre de Recherche sur les Macromolécules Végétales, Grenoble, France
| | - Andrea Vortkamp
- Developmental Biology, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| | | | - Elisa Migliorini
- Univ. Grenoble Alpes, INSERM, CEA, CNRS, U1292 Biosanté, EMR 5000, Grenoble, France.
| |
Collapse
|
2
|
Behera DP, Subadini S, Freudenberg U, Sahoo H. Sulfation of hyaluronic acid reconfigures the mechanistic pathway of bone morphogenetic protein-2 aggregation. Int J Biol Macromol 2024; 263:130128. [PMID: 38350587 DOI: 10.1016/j.ijbiomac.2024.130128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/03/2024] [Accepted: 02/09/2024] [Indexed: 02/15/2024]
Abstract
Bone morphogenetic protein-2 (BMP-2) is a critical growth factor of bone extracellular matrix (ECM), pivotal for osteogenesis. Glycosaminoglycans (GAGs), another vital ECM biomolecules, interact with growth factors, affecting signal transduction. Our study primarily focused on hyaluronic acid (HA), a prevalent GAG, and its sulfated derivative (SHA). We explored their impact on BMP-2's conformation, aggregation, and mechanistic pathways of aggregation using diverse optical and rheological methods. In the presence of HA and SHA, the secondary structure of BMP-2 underwent a structured transformation, characterized by a substantial increase in beta sheet content, and a detrimental alteration, manifesting as a shift towards unstructured content, respectively. Although both HA and SHA induced BMP-2 aggregation, their mechanisms differed. SHA led to rapid amorphous aggregates, while HA promoted amyloid fibrils with a lag phase and sigmoidal kinetics. Aggregate size and shape varied; HA produced larger structures, SHA smaller. Each aggregation type followed distinct pathways influenced by viscosity and excluded volume. Higher viscosity, low diffusivity of protein and higher excluded volume In the presence of HA promotes fibrillation having size in micrometer range. Low viscosity, high diffusivity of protein and lesser excluded volume leads to amorphous aggregate of size in nanometer range.
Collapse
Affiliation(s)
- Devi Prasanna Behera
- Biophysical and Protein Chemistry Lab, Department of Chemistry, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Suchismita Subadini
- Biophysical and Protein Chemistry Lab, Department of Chemistry, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Uwe Freudenberg
- Institute of Polymer Research, Technical University Dresden, 01307 Dresden, Germany
| | - Harekrushna Sahoo
- Biophysical and Protein Chemistry Lab, Department of Chemistry, National Institute of Technology, Rourkela 769008, Odisha, India; Center for Nanomaterials, National Institute of Technology, Rourkela 769008, Odisha, India.
| |
Collapse
|
3
|
Le Pennec J, Picart C, Vivès RR, Migliorini E. Sweet but Challenging: Tackling the Complexity of GAGs with Engineered Tailor-Made Biomaterials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312154. [PMID: 38011916 DOI: 10.1002/adma.202312154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Indexed: 11/29/2023]
Abstract
Glycosaminoglycans (GAGs) play a crucial role in tissue homeostasis by regulating the activity and diffusion of bioactive molecules. Incorporating GAGs into biomaterials has emerged as a widely adopted strategy in medical applications, owing to their biocompatibility and ability to control the release of bioactive molecules. Nevertheless, immobilized GAGs on biomaterials can elicit distinct cellular responses compared to their soluble forms, underscoring the need to understand the interactions between GAG and bioactive molecules within engineered functional biomaterials. By controlling critical parameters such as GAG type, density, and sulfation, it becomes possible to precisely delineate GAG functions within a biomaterial context and to better mimic specific tissue properties, enabling tailored design of GAG-based biomaterials for specific medical applications. However, this requires access to pure and well-characterized GAG compounds, which remains challenging. This review focuses on different strategies for producing well-defined GAGs and explores high-throughput approaches employed to investigate GAG-growth factor interactions and to quantify cellular responses on GAG-based biomaterials. These automated methods hold considerable promise for improving the understanding of the diverse functions of GAGs. In perspective, the scientific community is encouraged to adopt a rational approach in designing GAG-based biomaterials, taking into account the in vivo properties of the targeted tissue for medical applications.
Collapse
Affiliation(s)
- Jean Le Pennec
- U1292 Biosanté, INSERM, CEA, Univ. Grenoble Alpes, CNRS EMR 5000 Biomimetism and Regenerative Medicine, Grenoble, F-38054, France
| | - Catherine Picart
- U1292 Biosanté, INSERM, CEA, Univ. Grenoble Alpes, CNRS EMR 5000 Biomimetism and Regenerative Medicine, Grenoble, F-38054, France
| | | | - Elisa Migliorini
- U1292 Biosanté, INSERM, CEA, Univ. Grenoble Alpes, CNRS EMR 5000 Biomimetism and Regenerative Medicine, Grenoble, F-38054, France
| |
Collapse
|
4
|
Ivanjko N, Stokovic N, Milesevic M, Rumenovic V, Windhager R, Sampath KT, Kovacic N, Grcevic D, Vukicevic S. rhBMP6 in autologous blood coagulum is a preferred osteoinductive device to rhBMP2 on bovine collagen sponge in the rat ectopic bone formation assay. Biomed Pharmacother 2023; 169:115844. [PMID: 37948990 DOI: 10.1016/j.biopha.2023.115844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/02/2023] [Accepted: 11/05/2023] [Indexed: 11/12/2023] Open
Abstract
Osteoinductive BMPs require a suitable delivery system for treating various pathological conditions of the spine and segmental bone defects. INFUSE, the only commercially available BMP-based osteoinductive device, consisting of rhBMP2 on bovine absorbable collagen sponge (ACS) showed major disadvantages due to serious side effects. A novel osteoinductive device, OSTEOGROW, comprised of rhBMP6 dispersed within autologous blood coagulum (ABC) is a promising therapy for bone regeneration, subjected to several clinical trials for diaphysial bone repair and spinal fusion. In the present study, we have examined the release dynamics showing that the ABC carrier provided a slower, more steady BMP release in comparison to the ACS. Rat subcutaneous assay was employed to evaluate cellular events and the time course of ectopic osteogenesis. The host cellular response to osteoinductive implants was evaluated by flow cytometry, while dynamics of bone formation and maintenance in time were evaluated by histology, immunohistochemistry and micro CT analyses. Flow cytometry revealed that the recruitment of lymphoid cell populations was significantly higher in rhBMP6/ABC implants, while rhBMP2/ACS implants recruited more myeloid populations. Furthermore, rhBMP6/ABC implants more efficiently attracted early and committed progenitor cells. Dynamics of bone formation induced by rhBMP2/ACS was characterized by a delayed endochondral ossification process in comparison to rhBMP6/ABC implants. Besides, rhBMP6/ABC implants induced more ectopic bone volume in all observed time points in comparison to rhBMP2/ACS implants. These results indicate that OSTEOGROW was superior to INFUSE due to ABC's advantages as a carrier and rhBMP6 superior efficacy in inducing bone.
Collapse
Affiliation(s)
- Natalia Ivanjko
- Laboratory for Mineralized Tissues, School of Medicine, University of Zagreb, Zagreb, Croatia; Scientific Center of Excellence for Reproductive and Regenerative Medicine, Zagreb, Croatia
| | - Nikola Stokovic
- Laboratory for Mineralized Tissues, School of Medicine, University of Zagreb, Zagreb, Croatia; Scientific Center of Excellence for Reproductive and Regenerative Medicine, Zagreb, Croatia
| | - Marina Milesevic
- Laboratory for Mineralized Tissues, School of Medicine, University of Zagreb, Zagreb, Croatia; Scientific Center of Excellence for Reproductive and Regenerative Medicine, Zagreb, Croatia
| | - Viktorija Rumenovic
- Laboratory for Mineralized Tissues, School of Medicine, University of Zagreb, Zagreb, Croatia; Scientific Center of Excellence for Reproductive and Regenerative Medicine, Zagreb, Croatia
| | - Reinhard Windhager
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | | | - Natasa Kovacic
- Croatian Institute for Brain Research, Laboratory for Molecular Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Danka Grcevic
- Croatian Institute for Brain Research, Laboratory for Molecular Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Slobodan Vukicevic
- Laboratory for Mineralized Tissues, School of Medicine, University of Zagreb, Zagreb, Croatia; Scientific Center of Excellence for Reproductive and Regenerative Medicine, Zagreb, Croatia.
| |
Collapse
|
5
|
Seok JM, Kim MJ, Park JH, Kim D, Lee D, Yeo SJ, Lee JH, Lee K, Byun JH, Oh SH, Park SA. A bioactive microparticle-loaded osteogenically enhanced bioprinted scaffold that permits sustained release of BMP-2. Mater Today Bio 2023; 21:100685. [PMID: 37545560 PMCID: PMC10401289 DOI: 10.1016/j.mtbio.2023.100685] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 05/03/2023] [Accepted: 05/29/2023] [Indexed: 08/08/2023] Open
Abstract
Extrusion-based bioprinting technology is widely used for tissue regeneration and reconstruction. However, the method that uses only hydrogel as the bioink base material exhibits limited biofunctional properties and needs improvement to achieve the desired tissue regeneration. In this study, we present a three-dimensionally printed bioactive microparticle-loaded scaffold for use in bone regeneration applications. The unique structure of the microparticles provided sustained release of growth factor for > 4 weeks without the use of toxic or harmful substances. Before and after printing, the optimal particle ratio in the bioink for cell viability demonstrated a survival rate of ≥ 85% over 7 days. Notably, osteogenic differentiation and mineralization-mediated by human periosteum-derived cells in scaffolds with bioactive microparticles-increased over a 2-week interval. Here, we present an alternative bioprinting strategy that uses the sustained release of bioactive microparticles to improve biofunctional properties in a manner that is acceptable for clinical bone regeneration applications.
Collapse
Affiliation(s)
- Ji Min Seok
- Nano-Convergence Manufacturing Systems Research Division, Korea Institute of Machinery and Materials (KIMM), Daejeon, 34103, Republic of Korea
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Min Ji Kim
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jin Ho Park
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Dahong Kim
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dongjin Lee
- Nano-Convergence Manufacturing Systems Research Division, Korea Institute of Machinery and Materials (KIMM), Daejeon, 34103, Republic of Korea
| | - Seon Ju Yeo
- Nano-Convergence Manufacturing Systems Research Division, Korea Institute of Machinery and Materials (KIMM), Daejeon, 34103, Republic of Korea
| | - Jun Hee Lee
- Nano-Convergence Manufacturing Systems Research Division, Korea Institute of Machinery and Materials (KIMM), Daejeon, 34103, Republic of Korea
| | - Kangwon Lee
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
- Research Institute for Convergence Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - June-Ho Byun
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Se Heang Oh
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Su A Park
- Nano-Convergence Manufacturing Systems Research Division, Korea Institute of Machinery and Materials (KIMM), Daejeon, 34103, Republic of Korea
| |
Collapse
|
6
|
Zhu X, Wang C, Bai H, Zhang J, Wang Z, Li Z, Zhao X, Wang J, Liu H. Functionalization of biomimetic mineralized collagen for bone tissue engineering. Mater Today Bio 2023; 20:100660. [PMID: 37214545 PMCID: PMC10199226 DOI: 10.1016/j.mtbio.2023.100660] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/18/2023] [Accepted: 05/04/2023] [Indexed: 05/24/2023] Open
Abstract
Mineralized collagen (MC) is the basic unit of bone structure and function and is the main component of the extracellular matrix (ECM) in bone tissue. In the biomimetic method, MC with different nanostructures of neo-bone have been constructed. Among these, extra-fibrous MC has been approved by regulatory agencies and applied in clinical practice to play an active role in bone defect repair. However, in the complex microenvironment of bone defects, such as in blood supply disorders and infections, MC is unable to effectively perform its pro-osteogenic activities and needs to be functionalized to include osteogenesis and the enhancement of angiogenesis, anti-infection, and immunomodulation. This article aimed to discuss the preparation and biological performance of MC with different nanostructures in detail, and summarize its functionalization strategy. Then we describe the recent advances in the osteo-inductive properties and multifunctional coordination of MC. Finally, the latest research progress of functionalized biomimetic MC, along with the development challenges and future trends, are discussed. This paper provides a theoretical basis and advanced design philosophy for bone tissue engineering in different bone microenvironments.
Collapse
Affiliation(s)
- Xiujie Zhu
- Department of Orthopedics, The Second Hospital of Jilin University, 4110 Yatai Street, Changchun, 130041, PR China
| | - Chenyu Wang
- Department of Plastic and Reconstruct Surgery, The First Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, PR China
| | - Haotian Bai
- Department of Orthopedics, The Second Hospital of Jilin University, 4110 Yatai Street, Changchun, 130041, PR China
| | - Jiaxin Zhang
- Department of Orthopedics, The Second Hospital of Jilin University, 4110 Yatai Street, Changchun, 130041, PR China
| | - Zhonghan Wang
- Department of Orthopedics, The Second Hospital of Jilin University, 4110 Yatai Street, Changchun, 130041, PR China
| | - Zuhao Li
- Department of Orthopedics, The Second Hospital of Jilin University, 4110 Yatai Street, Changchun, 130041, PR China
| | - Xin Zhao
- Department of Orthopedics, The Second Hospital of Jilin University, 4110 Yatai Street, Changchun, 130041, PR China
| | - Jincheng Wang
- Department of Orthopedics, The Second Hospital of Jilin University, 4110 Yatai Street, Changchun, 130041, PR China
| | - He Liu
- Department of Orthopedics, The Second Hospital of Jilin University, 4110 Yatai Street, Changchun, 130041, PR China
| |
Collapse
|
7
|
Saito H, Shoji S, Kuroda A, Inoue G, Tazawa R, Sekiguchi H, Fukushima K, Miyagi M, Takaso M, Uchida K. In situ-formed hyaluronan gel/BMP-2/hydroxyapatite composite promotes bone union in refractory fracture model mice. Biomed Mater Eng 2023; 34:537-544. [PMID: 37334576 DOI: 10.3233/bme-230021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
BACKGROUND A combination of synthetic porous materials and BMP-2 has been used to promote fracture healing. For bone healing to be successful, it is important to use growth factor delivery systems that enable continuous release of BMP-2 at the fracture site. We previously reported that in situ-formed gels (IFGs) consisting of hyaluronan (HyA)-tyramine (TA), horseradish peroxidase and hydrogen peroxide enhance the bone formation ability of hydroxyapatite (Hap)/BMP-2 composites in a posterior lumbar fusion model. OBJECTIVE We examined the effectiveness of IFGs-HyA/Hap/BMP-2 composites for facilitating osteogenesis in refractory fracture model mice. METHODS After establishing the refractory fracture model, animals were either treated at the site of fracture with Hap harboring BMP-2 (Hap/BMP-2) or IFGs-HyA with Hap harboring BMP-2 (IFGs-HyA/Hap/BMP-2) (n = 10 each). Animals that underwent the fracture surgery but did not receive any treatment were considered the control group (n = 10). We determined the extent of bone formation at the fracture site according to findings on micro-computed tomography and histological studies four weeks following treatment. RESULTS Animals treated with IFGs-HyA/Hap/BMP-2 demonstrated significantly greater bone volume, bone mineral content and bone union than those treated with vehicle or IFG-HyA/Hap alone. CONCLUSIONS IFGs-HyA/Hap/BMP-2 could be an effective treatment option for refractory fractures.
Collapse
Affiliation(s)
- Hiroki Saito
- Department of Orthopedic Surgery, Kitasato University School of Medicine, Sagamihara City, Japan
| | - Shintaro Shoji
- Department of Orthopedic Surgery, Kitasato University School of Medicine, Sagamihara City, Japan
| | - Akiyoshi Kuroda
- Department of Orthopedic Surgery, Kitasato University School of Medicine, Sagamihara City, Japan
| | - Gen Inoue
- Department of Orthopedic Surgery, Kitasato University School of Medicine, Sagamihara City, Japan
| | - Ryo Tazawa
- Department of Orthopedic Surgery, Kitasato University School of Medicine, Sagamihara City, Japan
| | - Hiroyuki Sekiguchi
- Shonan University of Medical Sciences Research Institute, Chigasaki City, Japan
| | - Kensuke Fukushima
- Department of Orthopedic Surgery, Kitasato University School of Medicine, Sagamihara City, Japan
| | - Masayuki Miyagi
- Department of Orthopedic Surgery, Kitasato University School of Medicine, Sagamihara City, Japan
| | - Masashi Takaso
- Department of Orthopedic Surgery, Kitasato University School of Medicine, Sagamihara City, Japan
| | - Kentaro Uchida
- Department of Orthopedic Surgery, Kitasato University School of Medicine, Sagamihara City, Japan
- Shonan University of Medical Sciences Research Institute, Chigasaki City, Japan
| |
Collapse
|
8
|
Gélébart P, Cuenot S, Sinquin C, Halgand B, Sourice S, Le Visage C, Guicheux J, Colliec-Jouault S, Zykwinska A. Microgels based on Infernan, a glycosaminoglycan-mimetic bacterial exopolysaccharide, as BMP-2 delivery systems. Carbohydr Polym 2022; 284:119191. [DOI: 10.1016/j.carbpol.2022.119191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023]
|
9
|
Kim S, Fan J, Lee CS, Chen C, Lee M. Sulfonate Hydrogel-siRNA Conjugate Facilitates Osteogenic Differentiation of Mesenchymal Stem Cells by Controlled Gene Silencing and Activation of BMP Signaling. ACS APPLIED BIO MATERIALS 2021; 4:5189-5200. [PMID: 34661086 DOI: 10.1021/acsabm.1c00369] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hydrogels have been widely used in bone tissue engineering due to their tunable characteristics that allow facile modifications with various biochemical properties to support cell growth and guide proper cell functions. Herein, we report a design of hydrogel-siRNA conjugate that facilitates osteogenesis via gene silencing and activation of bone morphogenetic protein (BMP) signaling. A sulfonate hydrogel is prepared by modifying chitosan with sulfoacetic acid to mimic a natural sulfated polysaccharide and to provide a hydrogel surface that enables BMP binding. Then, siRNA targeting noggin, an endogenous extracellular antagonist of BMP signaling, is covalently conjugated to the sulfonate hydrogel by visible blue light crosslinking. The sulfonate hydrogel-siRNA conjugate is efficient to bind BMPs and also successfully prolongs the release of siRNA for sustained noggin suppression, thereby resulting in significantly increased osteogenic differentiation. Lastly, demineralized bone matrix (DBM) is incorporated into the sulfonate hydrogel-siRNA conjugate, wherein the DBM incorporation induces noggin expression via a negative feedback mechanism that regulates BMP signaling in DBM. However, simultaneous delivery of siRNA downregulates noggin thus facilitating endogenous BMP activity and enhancing the osteogenic efficacy of DBM. These findings support a promising hydrogel RNA silencing platform for bone tissue engineering applications.
Collapse
Affiliation(s)
- Soyon Kim
- Division of Advanced Prosthodontics, University of California, Los Angeles, USA
| | - Jiabing Fan
- Division of Advanced Prosthodontics, University of California, Los Angeles, USA
| | - Chung-Sung Lee
- Division of Advanced Prosthodontics, University of California, Los Angeles, USA
| | - Chen Chen
- Division of Advanced Prosthodontics, University of California, Los Angeles, USA
| | - Min Lee
- Division of Advanced Prosthodontics, University of California, Los Angeles, USA.,Department of Bioengineering, University of California, Los Angeles, USA
| |
Collapse
|
10
|
Terauchi M, Tamura A, Arisaka Y, Masuda H, Yoda T, Yui N. Cyclodextrin-Based Supramolecular Complexes of Osteoinductive Agents for Dental Tissue Regeneration. Pharmaceutics 2021; 13:136. [PMID: 33494320 PMCID: PMC7911178 DOI: 10.3390/pharmaceutics13020136] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/13/2021] [Accepted: 01/18/2021] [Indexed: 12/16/2022] Open
Abstract
Oral tissue regeneration has received growing attention for improving the quality of life of patients. Regeneration of oral tissues such as alveolar bone and widely defected bone has been extensively investigated, including regenerative treatment of oral tissues using therapeutic cells and growth factors. Additionally, small-molecule drugs that promote bone formation have been identified and tested as new regenerative treatment. However, treatments need to progress to realize successful regeneration of oral functions. In this review, we describe recent progress in development of regenerative treatment of oral tissues. In particular, we focus on cyclodextrin (CD)-based pharmaceutics and polyelectrolyte complexation of growth factors to enhance their solubility, stability, and bioactivity. CDs can encapsulate hydrophobic small-molecule drugs into their cavities, resulting in inclusion complexes. The inclusion complexation of osteoinductive small-molecule drugs improves solubility of the drugs in aqueous solutions and increases in vitro osteogenic differentiation efficiency. Additionally, various anionic polymers such as heparin and its mimetic polymers have been developed to improve stability and bioactivity of growth factors. These polymers protect growth factors from deactivation and degradation by complex formation through electrostatic interaction, leading to potentiation of bone formation ability. These approaches using an inclusion complex and polyelectrolyte complexes have great potential in the regeneration of oral tissues.
Collapse
Affiliation(s)
- Masahiko Terauchi
- Department of Maxillofacial Surgery, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo, Tokyo 113-8549, Japan; (M.T.); (H.M.); (T.Y.)
| | - Atsushi Tamura
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan; (Y.A.); (N.Y.)
| | - Yoshinori Arisaka
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan; (Y.A.); (N.Y.)
| | - Hiroki Masuda
- Department of Maxillofacial Surgery, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo, Tokyo 113-8549, Japan; (M.T.); (H.M.); (T.Y.)
| | - Tetsuya Yoda
- Department of Maxillofacial Surgery, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo, Tokyo 113-8549, Japan; (M.T.); (H.M.); (T.Y.)
| | - Nobuhiko Yui
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan; (Y.A.); (N.Y.)
| |
Collapse
|
11
|
Migliorini E, Guevara-Garcia A, Albiges-Rizo C, Picart C. Learning from BMPs and their biophysical extracellular matrix microenvironment for biomaterial design. Bone 2020; 141:115540. [PMID: 32730925 PMCID: PMC7614069 DOI: 10.1016/j.bone.2020.115540] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 01/19/2023]
Abstract
It is nowadays well-accepted that the extracellular matrix (ECM) is not a simple reservoir for growth factors but is an organization center of their biological activity. In this review, we focus on the ability of the ECM to regulate the biological activity of BMPs. In particular, we survey the role of the ECM components, notably the glycosaminoglycans and fibrillary ECM proteins, which can be promoters or repressors of the biological activities mediated by the BMPs. We examine how a process called mechano-transduction induced by the ECM can affect BMP signaling, including BMP internalization by the cells. We also focus on the spatio-temporal regulation of the BMPs, including their release from the ECM, which enables to modulate their spatial localization as well as their local concentration. We highlight how biomaterials can recapitulate some aspects of the BMPs/ECM interactions and help to answer fundamental questions to reveal previously unknown molecular mechanisms. Finally, the design of new biomaterials inspired by the ECM to better present BMPs is discussed, and their use for a more efficient bone regeneration in vivo is also highlighted.
Collapse
Affiliation(s)
- Elisa Migliorini
- CNRS, Grenoble Institute of Technology, LMGP, UMR 5628, 3 Parvis Louis Néel, 38016 Grenoble, France; CEA, Institute of Interdisciplinary Research of Grenoble (IRIG), Biomimetism and Regenerative Medicine Lab, ERL 5000, Université Grenoble-Alpes (UGA)/CEA/CNRS, Grenoble France.
| | - Amaris Guevara-Garcia
- CNRS, Grenoble Institute of Technology, LMGP, UMR 5628, 3 Parvis Louis Néel, 38016 Grenoble, France; CEA, Institute of Interdisciplinary Research of Grenoble (IRIG), Biomimetism and Regenerative Medicine Lab, ERL 5000, Université Grenoble-Alpes (UGA)/CEA/CNRS, Grenoble France; Université Grenoble Alpes, Institut for Advances Biosciences, Institute Albert Bonniot, INSERM U1209, CNRS 5309, La Tronche, France
| | - Corinne Albiges-Rizo
- Université Grenoble Alpes, Institut for Advances Biosciences, Institute Albert Bonniot, INSERM U1209, CNRS 5309, La Tronche, France
| | - Catherine Picart
- CNRS, Grenoble Institute of Technology, LMGP, UMR 5628, 3 Parvis Louis Néel, 38016 Grenoble, France; CEA, Institute of Interdisciplinary Research of Grenoble (IRIG), Biomimetism and Regenerative Medicine Lab, ERL 5000, Université Grenoble-Alpes (UGA)/CEA/CNRS, Grenoble France.
| |
Collapse
|
12
|
Osteogenic-differentiated mesenchymal stem cell-secreted extracellular matrix as a bone morphogenetic protein-2 delivery system for ectopic bone formation. Acta Biomater 2020; 116:186-200. [PMID: 32911108 DOI: 10.1016/j.actbio.2020.09.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 08/28/2020] [Accepted: 09/02/2020] [Indexed: 02/04/2023]
Abstract
While human bone morphogenetic protein-2 (BMP-2) is a promising growth factor for bone regeneration, a major challenge in biomedical applications is finding an optimal carrier for its delivery at the site of injury. Because of their natural affinities for growth factors (including BMP-2) as well as their role in instructing cell function, cultured cell-derived extracellular matrices (ECM) are of special interest. We hereby hypothesized that a "bony matrix" containing mineralized, osteogenic ECM is a potential efficacious carrier of BMP-2 for promoting bone formation and, therefore, compared the efficacy of the decellularized ECM derived from osteogenic-differentiated human mesenchymal stem cells (hMSCs) to the one obtained from ECM from undifferentiated hMSCs. Our results provided evidence that both ECMs can bind BMP-2 and promote bone formation when implanted ectopically in mice. The osteoinductive potential of BMP-2, however, was greater when loaded within an osteogenic MSC-derived ECM; this outcome was correlated with higher sequestration capacity of BMP-2 over time in vivo. Interestingly, although the BMP-2 mainly bound onto the mineral crystals contained within the osteogenic MSC derived-ECM, these mineral components were not involved in the observed higher osteoinductivity, suggesting that the organic components were the critical components for the matrix efficacy as BMP-2 carrier.
Collapse
|
13
|
Son HJ, Lee MN, Kim Y, Choi H, Jeong BC, Oh SH, Kim JW, Kwon SH, Kim SH, Song SC, Lee SE, Koh JT. Bone Generation Following Repeated Administration of Recombinant Bone Morphogenetic Protein 2. Tissue Eng Regen Med 2020; 18:155-164. [PMID: 33058016 DOI: 10.1007/s13770-020-00290-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 07/14/2020] [Accepted: 07/22/2020] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND The delivery of recombinant human bone morphogenetic protein 2 (rhBMP2) by using various carriers has been used to successfully induce bone formation in many animal models. However, the effect of multiple administration of rhBMP2 on bone formation and BMP2 antibody production has not been determined. Our aim was to examine the bone formation activity of rhBMP2 and serum levels of anti-BMP2 antibodies following the repeated administration of rhBMP2 in mice. METHODS Absorbable collagen sponges or polyphosphazene hydrogels containing rhBMP2 were subcutaneously implanted or injected into one side on the back of six-week-old C57BL/6 mice. Three or 4 weeks later, the same amount of rhBMP2 was administered again with the same carrier into the subcutaneous regions on the other side of the back or into calvarial defects. The effects of a single administration of rhBMP2 on the osteoinductive ability in the ectopic model were compared with those of repeated administrations. In vivo ectopic or orthotopic bone formation was evaluated using microradiography and histological analyses. Serum concentrations of anti-rhBMP2 antibodies were measured by ELISAs. RESULTS Re-administration of the same amount of rhBMP2 into the subcutaneous area showed a comparable production of ectopic bone as after the first administration. The bone forming ability of repeated rhBMP2 administrations was equal to that of single rhBMP2 administration. The administration of rhBMP2 into calvarial defects, following the first subcutaneous administration of rhBMP2 on the back, completely recovered the defect area with newly regenerated bone within 3 weeks. Repeated administration of rhBMP2 at 4-week intervals did not significantly alter the serum levels of anti-BMP2 antibodies and did not induce any inflammatory response. The serum obtained from rhBMP2-exposed mice had no effect on the ability of rhBMP2 to induce osteogenic gene expressions in MC3T3-E1. CONCLUSION We suggest that the osteoinductive ability of rhBMP2 is not compromised by repeated administrations. Thus, rhBMP2 can be repeatedly used for bone regeneration at various sites within a short duration.
Collapse
Affiliation(s)
- Hye-Ju Son
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, 33 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Mi Nam Lee
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, 33 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea.,Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, 33 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Yuri Kim
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, 33 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Hyuck Choi
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, 33 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Byung-Chul Jeong
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, 33 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Sin-Hye Oh
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, 33 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea.,Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, 33 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Jung-Woo Kim
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, 33 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea.,Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, 33 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Seung-Hee Kwon
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, 33 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea.,Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, 33 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Sun-Hun Kim
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, 33 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea.,Department of Oral Anatomy, School of Dentistry, Chonnam National University, 33 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Soo-Chang Song
- Center for Biomaterials, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Shee Eun Lee
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, 33 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea.,Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, 33 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Jeong-Tae Koh
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, 33 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea. .,Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, 33 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
14
|
Shoji S, Uchida K, Satio W, Sekiguchi H, Inoue G, Miyagi M, Takata K, Yokozeki Y, Takaso M. Acceleration of bone union by in situ-formed hydrogel containing bone morphogenetic protein-2 in a mouse refractory fracture model. J Orthop Surg Res 2020; 15:426. [PMID: 32948214 PMCID: PMC7501615 DOI: 10.1186/s13018-020-01953-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 09/09/2020] [Indexed: 11/30/2022] Open
Abstract
Background An enzymatic crosslinking strategy using hydrogen peroxide and horseradish peroxidase is receiving increasing attention for application with in situ-formed hydrogels (IFHs). Several studies have reported the application of IFHs in cell delivery and tissue engineering. IFHs may also be ideal carrier materials for bone repair, although their potential as a carrier for bone morphogenetic protein (BMP)-2 has yet to be examined. Here, we examined the effect of an IFH made of hyaluronic acid (IFH-HA) containing BMP-2 in promoting osteogenesis in a mouse refractory fracture model. Methods Immediately following a fracture procedure, animals either received no treatment (control) or an injection of IFH-HA/PBS or IFH-HA containing 2 μg BMP-2 (IFH-HA/BMP-2) into the fracture site (n = 16, each treatment). Results Fracture sites injected with IFH-HA/BMP-2 showed significantly greater bone volume, bone mineral content, and bone union compared with sites receiving no treatment or treated with IFH-HA/PBS alone (each n = 10). Gene expression levels of osteogenic markers, Alpl, Bglap, and Osx, were significantly raised in the IFH-HA/BMP-2 group compared to the IFH-HA/PBS and control groups (each n = 6). Conclusion IFH-HA/BMP-2 may contribute to the treatment of refractory fractures.
Collapse
Affiliation(s)
- Shintaro Shoji
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara City, Kanagawa, 252-0374, Japan
| | - Kentaro Uchida
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara City, Kanagawa, 252-0374, Japan. .,Shonan University of Medical Sciences Research Institute, Nishikubo 500, Chigasaki City, Kanagawa, 253-0083, Japan.
| | - Wataru Satio
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara City, Kanagawa, 252-0374, Japan
| | - Hiroyuki Sekiguchi
- Shonan University of Medical Sciences Research Institute, Nishikubo 500, Chigasaki City, Kanagawa, 253-0083, Japan
| | - Gen Inoue
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara City, Kanagawa, 252-0374, Japan
| | - Masayuki Miyagi
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara City, Kanagawa, 252-0374, Japan
| | - Ken Takata
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara City, Kanagawa, 252-0374, Japan
| | - Yuji Yokozeki
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara City, Kanagawa, 252-0374, Japan
| | - Masashi Takaso
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara City, Kanagawa, 252-0374, Japan
| |
Collapse
|
15
|
Halloran D, Durbano HW, Nohe A. Bone Morphogenetic Protein-2 in Development and Bone Homeostasis. J Dev Biol 2020; 8:E19. [PMID: 32933207 PMCID: PMC7557435 DOI: 10.3390/jdb8030019] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/01/2020] [Accepted: 09/11/2020] [Indexed: 12/11/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are multi-functional growth factors belonging to the Transforming Growth Factor-Beta (TGF-β) superfamily. These proteins are essential to many developmental processes, including cardiogenesis, neurogenesis, and osteogenesis. Specifically, within the BMP family, Bone Morphogenetic Protein-2 (BMP-2) was the first BMP to be characterized and has been well-studied. BMP-2 has important roles during embryonic development, as well as bone remodeling and homeostasis in adulthood. Some of its specific functions include digit formation and activating osteogenic genes, such as Runt-Related Transcription Factor 2 (RUNX2). Because of its diverse functions and osteogenic potential, the Food and Drug Administration (FDA) approved usage of recombinant human BMP-2 (rhBMP-2) during spinal fusion surgery, tibial shaft repair, and maxillary sinus reconstructive surgery. However, shortly after initial injections of rhBMP-2, several adverse complications were reported, and alternative therapeutics have been developed to limit these side-effects. As the clinical application of BMP-2 is largely implicated in bone, we focus primarily on its role in bone. However, we also describe briefly the role of BMP-2 in development. We then focus on the structure of BMP-2, its activation and regulation signaling pathways, BMP-2 clinical applications, and limitations of using BMP-2 as a therapeutic. Further, this review explores other potential treatments that may be useful in treating bone disorders.
Collapse
Affiliation(s)
| | | | - Anja Nohe
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA; (D.H.); (H.W.D.)
| |
Collapse
|
16
|
Um IW, Ku JK, Kim YK, Lee BK, Leem DH. Histological Review of Demineralized Dentin Matrix as a Carrier of rhBMP-2. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:284-293. [PMID: 31928139 PMCID: PMC7310192 DOI: 10.1089/ten.teb.2019.0291] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
In 2007, recombinant human bone morphogenetic protein-2 (rhBMP-2) was approved for use in humans at a concentration of 1.5 mg/mL with absorbable collagen sponges as an alternative to autogenous bone grafts for alveolar ridge augmentation, defects associated with extraction sockets, and sinus augmentation. However, the use of supraphysiological doses and the insufficient retention of rhBMP-2, when delivered through collagen sponge, result in dose-dependent side effects related to off-label use. Demineralized dentin matrix (DDM), an osteoinducing bone substrate, has been used as an rhBMP-2 carrier since 1998. In addition, DDM has both microparticle and nanoparticle structures, which do not undergo remodeling, unlike bone. In vitro, DDM is a suitable carrier for BMP-2, with the continued release over 30 days at concentrations sufficient to stimulate osteogenic differentiation. In this review, we discuss the histological outcomes of DDM loaded with rhBMP-2 to highlight the biological functions of exogenous rhBMP-2 associated with the DDM carrier in clinical applications in implant dentistry. Impact Statement Demineralized dentin matrix (DDM) has been used as an recombinant human bone morphogenetic protein (rhBMP-2) carrier and osteo-inducing bone substrate to facilitate continued release and stimulate osteogenic differentiation. In this review, we discuss the histological outcomes of DDM loaded with rhBMP-2 in order to highlight the biological functions of exogenous rhBMP-2 associated with the DDM carrier in clinical applications in implant dentistry.
Collapse
Affiliation(s)
- In-Woong Um
- R&D Institute, Korea Tooth Bank, Seoul, Republic of Korea
| | - Jeong-Kui Ku
- Section of Dentistry, Department of Oral and Maxillofacial Surgery, Armed Forces Capital Hospital, Seongnam, Republic of Korea
- Department of Oral and Maxillofacial Surgery, Seoul Asan Medical Center, Seoul, Republic of Korea
- Address correspondence to: Jeong-Kui Ku, DDS, MMSc, FIBCOMS, Section of Dentistry, Department of Oral and Maxillofacial Surgery, Armed Forces Capital Hospital, 81, Saemaul-ro 117, Bundang-gu, Seongnam 13575, Republic of Korea
| | - Young-Kyun Kim
- Section of Dentistry, Department of Oral and Maxillofacial Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Bu-Kyu Lee
- Department of Oral and Maxillofacial Surgery, Seoul Asan Medical Center, Seoul, Republic of Korea
| | - Dae Ho Leem
- Department of Oral and Maxillofacial Surgery, School of Dentistry and Institute of Oral Bioscience, Research Institute of Clinical Medicine of Chonbuk National University, Jeonju, Republic of Korea
- Biomedical Research Institute of Chonbuk National University Hospital, Chonbuk National University, Jeonju, Republic of Korea
| |
Collapse
|
17
|
Hachim D, Whittaker TE, Kim H, Stevens MM. Glycosaminoglycan-based biomaterials for growth factor and cytokine delivery: Making the right choices. J Control Release 2019; 313:131-147. [PMID: 31629041 PMCID: PMC6900262 DOI: 10.1016/j.jconrel.2019.10.018] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 12/21/2022]
Abstract
Controlled, localized drug delivery is a long-standing goal of medical research, realization of which could reduce the harmful side-effects of drugs and allow more effective treatment of wounds, cancers, organ damage and other diseases. This is particularly the case for protein "drugs" and other therapeutic biological cargoes, which can be challenging to deliver effectively by conventional systemic administration. However, developing biocompatible materials that can sequester large quantities of protein and release them in a sustained and controlled manner has proven challenging. Glycosaminoglycans (GAGs) represent a promising class of bio-derived materials that possess these key properties and can additionally potentially enhance the biological effects of the delivered protein. They are a diverse group of linear polysaccharides with varied functionalities and suitabilities for different cargoes. However, most investigations so far have focused on a relatively small subset of GAGs - particularly heparin, a readily available, promiscuously-binding GAG. There is emerging evidence that for many applications other GAGs are in fact more suitable for regulated and sustained delivery. In this review, we aim to illuminate the beneficial properties of various GAGs with reference to specific protein cargoes, and to provide guidelines for informed choice of GAGs for therapeutic applications.
Collapse
Affiliation(s)
- Daniel Hachim
- Department of Materials, Imperial College London, London, SW7 2AZ, United Kingdom; Department of Bioengineering, Imperial College London, London, SW7 2AZ, United Kingdom; Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Thomas E Whittaker
- Department of Materials, Imperial College London, London, SW7 2AZ, United Kingdom; Department of Bioengineering, Imperial College London, London, SW7 2AZ, United Kingdom; Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Hyemin Kim
- Department of Materials, Imperial College London, London, SW7 2AZ, United Kingdom; Department of Bioengineering, Imperial College London, London, SW7 2AZ, United Kingdom; Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Molly M Stevens
- Department of Materials, Imperial College London, London, SW7 2AZ, United Kingdom; Department of Bioengineering, Imperial College London, London, SW7 2AZ, United Kingdom; Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, United Kingdom.
| |
Collapse
|
18
|
Lee JH, Jin YZ. Recombinant human bone morphogenetic protein-2 loaded porous β-tricalcium phosphate microsphere-hyaluronic acid composites promoted osseointegration around titanium implants. INT J POLYM MATER PO 2019. [DOI: 10.1080/00914037.2018.1446138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Jae Hyup Lee
- Department of Orthopedic Surgery, College of Medicine, SMG-SNU Boramae Medical Center, Seoul National University, Seoul, Korea
- Institute of Medical and Biological Engineering, Medical Research Center, Seoul National University, Seoul, Korea
| | - Yuan Zhe Jin
- Department of Orthopedic Surgery, College of Medicine, SMG-SNU Boramae Medical Center, Seoul National University, Seoul, Korea
| |
Collapse
|
19
|
Liu S, Zhou H, Liu H, Ji H, Fei W, Luo E. Fluorine-contained hydroxyapatite suppresses bone resorption through inhibiting osteoclasts differentiation and function in vitro and in vivo. Cell Prolif 2019; 52:e12613. [PMID: 30968984 PMCID: PMC6536412 DOI: 10.1111/cpr.12613] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 03/08/2019] [Accepted: 03/09/2019] [Indexed: 02/05/2023] Open
Abstract
Objectives Fluorine, an organic trace element, has been shown to unfavourably effect osteoclasts function at a low dose. Use of hydroxyapatite (HA) has been effective in exploring its roles in promoting bone repair. In this study, we used HA modified with fluorine to investigate whether it could influence osteoclastic activity in vitro and ovariectomy‐induced osteoclasts hyperfunction in vivo. Materials and methods Fluorohydroxyapatite (FHA) was obtained and characterized by scanning electron microscope (SEM). Osteoclasts proliferation and apoptosis treated with FHA were assessed by MTT and TUNEL assay. SEM, F‐actin, TRAP activity and bone resorption experiment were performed to determine the influence of FHA on osteoclasts differentiation and function. Moreover, HA and FHA were implanted into ovariectomized osteoporotic and sham surgery rats. Histology and Micro‐CT were examined for further verification. Results Fluorine released from FHA slowly and sustainably. FHA hampered osteoclasts proliferation, promoted osteoclasts apoptosis, suppressed osteoclasts differentiation and function. Experiments in vivo validated that FHA participation brought about an inhibitory effect on osteoclasts hyperfunction and less bone absorption. Conclusion The results indicated that FHA served as an efficient regulator to attenuate osteoclasts formation and function and was proposed as a candidature for bone tissue engineering applications.
Collapse
Affiliation(s)
- Shibo Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hao Zhou
- Department of Stomotology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu, China
| | - Hanghang Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Huanzhong Ji
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wei Fei
- Department of Stomotology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu, China
| | - En Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Anouz R, Repanas A, Schwarz E, Groth T. Novel Surface Coatings Using Oxidized Glycosaminoglycans as Delivery Systems of Bone Morphogenetic Protein 2 (BMP‐2) for Bone Regeneration. Macromol Biosci 2018; 18:e1800283. [DOI: 10.1002/mabi.201800283] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/03/2018] [Indexed: 12/28/2022]
Affiliation(s)
- Reema Anouz
- Department of Biomedical MaterialsMartin Luther University Halle‐Wittenberg Heinrich‐Damerow‐Strasse 4 06120 Halle (Saale) Germany
| | - Alexandros Repanas
- Department of Biomedical MaterialsMartin Luther University Halle‐Wittenberg Heinrich‐Damerow‐Strasse 4 06120 Halle (Saale) Germany
| | - Elisabeth Schwarz
- Institute of PharmacyMartin Luther University Halle‐Wittenberg Wolfgang‐Langenbeck‐Strasse 4 06120 Halle (Saale) Germany
| | - Thomas Groth
- Department of Biomedical MaterialsMartin Luther University Halle‐Wittenberg Heinrich‐Damerow‐Strasse 4 06120 Halle (Saale) Germany
- Interdisciplinary Center of Material Research and Interdisciplinary Center of Applied ResearchMartin Luther University Halle‐Wittenberg 06099 Halle (Saale) Germany
| |
Collapse
|
21
|
Kim HY, Park JH, Byun JH, Lee JH, Oh SH. BMP-2-Immobilized Porous Matrix with Leaf-Stacked Structure as a Bioactive GBR Membrane. ACS APPLIED MATERIALS & INTERFACES 2018; 10:30115-30124. [PMID: 30130399 DOI: 10.1021/acsami.8b09558] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
We developed an asymmetrically porous membrane with a leaf-stacked structure (LSS membrane; top with nanosized pores and bulk/bottom with leaf-stacked structure) via immersion-precipitation using polycarprolactone (PCL)/Pluronic F127 mixture solution (in tetraglycol). The bone morphogenetic protein-2 (BMP-2) is immobilized on the pore surfaces of the LSS membrane by immersing the membrane in the BMP-2 solution. The BMP-2 loaded in the LSS membrane is continuously released for 38 days (without additional modifications of the matrix) to improve osteogenic differentiation of cells and new bone formation (carvarial defect rat model). The leaf-stacked structure is recognized to be a physical stimulus for bone regeneration, and the stimulation effect is comparable to that of continuously released BMP-2. Moreover, we observe the combined effect of BMP-2 and the leaf-stacked structure for bone healing. Thus, we suggest that the BMP-2-immobilized LSS membrane may be a candidate as a bioactive guided bone regeneration (GBR) membrane for clinical applications, due to the use of clinically acceptable biomaterials and fabrication procedures as well as effective osteogenic differentiation and bone regeneration.
Collapse
Affiliation(s)
- Ho Yong Kim
- Department of Nanobiomedical Science , Dankook University , Cheonan 31116 , Republic of Korea
| | - Jin Hyun Park
- Department of Nanobiomedical Science , Dankook University , Cheonan 31116 , Republic of Korea
| | - June-Ho Byun
- Department of Oral and Maxillofacial Surgery , Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University , Jinju 52828 , Republic of Korea
| | - Jin Ho Lee
- Department of Advanced Materials and Chemical Engineering , Hannam University , Daejeon 34054 , Republic of Korea
| | - Se Heang Oh
- Department of Nanobiomedical Science , Dankook University , Cheonan 31116 , Republic of Korea
- Department of Pharmaceutical Engineering , Dankook University , Cheonan 31116 , Republic of Korea
| |
Collapse
|
22
|
De Pieri A, Ribeiro S, Tsiapalis D, Eglin D, Bohner M, Dubruel P, Procter P, Zeugolis DI, Bayon Y. Joint academic and industrial efforts towards innovative and efficient solutions for clinical needs. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2018; 29:129. [PMID: 30066293 DOI: 10.1007/s10856-018-6136-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 07/20/2018] [Indexed: 06/08/2023]
Abstract
The 4th Translational Research Symposium (TRS) was organised at the annual meeting of the European Society for Biomaterials (ESB) 2017, Athens, Greece, with a focus on 'Academia-Industry Clusters of Research for Innovation Catalysis'. Collaborations between research institutes and industry can be sustained in several ways such as: European Union (EU) funded consortiums; syndicates of academic institutes, clinicians and industries; funding from national governments; and private collaborations between universities and companies. Invited speakers from industry and research institutions presented examples of these collaborations in the translation of research ideas or concepts into marketable products. The aim of the present article is to summarize the key messages conveyed during these lectures. In particular, emphasis is put on the challenges to appropriately identify and select unmet clinical needs and their translation by ultimately implementing innovative and efficient solutions achieved through joint academic and industrial efforts.
Collapse
Affiliation(s)
- Andrea De Pieri
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), National University of Galway Ireland (NUI Galway), Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), National University of Galway Ireland (NUI Galway), Galway, Ireland
- Proxy Biomedical Ltd., Coilleach, Spiddal, Galway, Ireland
| | - Sofia Ribeiro
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), National University of Galway Ireland (NUI Galway), Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), National University of Galway Ireland (NUI Galway), Galway, Ireland
- Medtronic Sofradim Production, Trevoux, France
| | - Dimitrios Tsiapalis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), National University of Galway Ireland (NUI Galway), Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), National University of Galway Ireland (NUI Galway), Galway, Ireland
| | - David Eglin
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland
| | - Marc Bohner
- RMS Foundation, Bischmattstrasse 12, P.O. Box 203, 2544, Bettlach, Switzerland
| | - Peter Dubruel
- Polymer Chemistry & Biomaterials Research Group, Ghent University, Krijgslaan 281 S4 Bis, Ghent, 9000, Belgium
| | - Philip Procter
- CPP SARL Divonne les Bains, 01220, Divonne les Bains, France
- Applied Materials Science, Dept Eng. Sciences, Uppsala University, 752 37, Uppsala, Sweden
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), National University of Galway Ireland (NUI Galway), Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), National University of Galway Ireland (NUI Galway), Galway, Ireland
| | - Yves Bayon
- Medtronic Sofradim Production, Trevoux, France.
| |
Collapse
|
23
|
Zhang N, Ma L, Liu X, Jiang X, Yu Z, Zhao D, Zhang L, Zhang C, Huang F. In vitro and in vivo evaluation of xenogeneic bone putty with the carrier of hydrogel derived from demineralized bone matrix. Cell Tissue Bank 2018; 19:591-601. [PMID: 29974309 DOI: 10.1007/s10561-018-9708-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 06/29/2018] [Indexed: 02/06/2023]
Abstract
The demineralized bone matrix (DBM) putty is a traditional bone graft utilized to facilitate the repair and reconstruction of bone. Recent studies indicated the DBM putties with the various carriers were different in bone repairing ability. In order to prepare a kind of DBM putty with a good biocompatibility and bioactivity, the DBM gel was processed from the DBM and the feasibility as a carrier for the DBM putty was evaluated. After the bovine DBM gel was prepared, the BMPs content as well as the ability to promote osteogenic differentiation of MC3T3-E1 cells in vitro were investigated. Then the DBM putty was prepared and filled into the rat calvarial defect model to evaluate the bone repairing ability by micro-CT and histology. The result showed there was 2.953 ± 0.054 ng BMP contained in per gram of the DBM gel. And the ALP production of MC3T3-E1 cells in the DBM gels group increased with prolonged culturing, the mineralized nodules formed in MC3T3-E1 cells on 14th day after co-culture. The putty prepared by DBM gel was easy to handle without loss of DBM particles at room temperature. In the rat calvarial bone defect experiment, histological observation showed more mature bone formed in the DBM putty group than that in the type I collagen group at 12 weeks, which indicated the bone putty prepared by DBM gel exhibited a better bone repair capability.
Collapse
Affiliation(s)
- Naili Zhang
- Department of Human Anatomy, School of Basic Medical Science, Binzhou Medical University, 346 Guanhai Road, Laishan, Yantai, 264003, Shandong, China
| | - Lina Ma
- Department of Diagnostics, School of Medicine, Binzhou Medical University, 346 Guanhai Road, Laishan, Yantai, 264003, Shandong, China
| | - Xiaowei Liu
- Department of Human Anatomy, School of Basic Medical Science, Binzhou Medical University, 346 Guanhai Road, Laishan, Yantai, 264003, Shandong, China
| | - Xiaorui Jiang
- Department of Hand and Foot Surgery, Yuhuangding Hospital, 20 Yuhuangding East Road, Zhifu, Yantai, 264000, China
| | - Zhenhai Yu
- Department of Human Anatomy, School of Basic Medical Science, Binzhou Medical University, 346 Guanhai Road, Laishan, Yantai, 264003, Shandong, China
| | - Dongmei Zhao
- Department of Human Anatomy, School of Basic Medical Science, Binzhou Medical University, 346 Guanhai Road, Laishan, Yantai, 264003, Shandong, China
| | - Luping Zhang
- Department of Human Anatomy, School of Basic Medical Science, Binzhou Medical University, 346 Guanhai Road, Laishan, Yantai, 264003, Shandong, China
| | - Chunlei Zhang
- Department of Human Anatomy, School of Basic Medical Science, Binzhou Medical University, 346 Guanhai Road, Laishan, Yantai, 264003, Shandong, China
| | - Fei Huang
- Department of Human Anatomy, School of Basic Medical Science, Binzhou Medical University, 346 Guanhai Road, Laishan, Yantai, 264003, Shandong, China.
| |
Collapse
|
24
|
Kim HY, Lee JH, Lee HAR, Park JS, Woo DK, Lee HC, Rho GJ, Byun JH, Oh SH. Sustained Release of BMP-2 from Porous Particles with Leaf-Stacked Structure for Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2018; 10:21091-21102. [PMID: 29863327 DOI: 10.1021/acsami.8b02141] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Sustained release of bioactive molecules from delivery systems is a common strategy for ensuring their prolonged bioactivity and for minimizing safety issues. However, residual toxic reagents, the use of harsh organic solvents, and complex fabrication procedures in conventional delivery systems are considered enormous impediments toward clinical use. Herein, we describe bone morphogenetic protein-2 (BMP-2)-immobilized porous polycaprolactone particles with unique leaf-stacked structures (LSS particles) prepared using clinically feasible materials and procedures. The BMP-2 immobilized in these LSS particles is continuously released up to 36 days to provide an appropriate environment for osteogenic differentiation of human periosteum-derived cells and new bone formation. Thus, the leaf-stacked structures of these LSS particles provide a simple but clinically applicable platform for effectively delivering a variety of bioactive molecules, such as growth factors, hormones, cytokines, peptides, etc.
Collapse
Affiliation(s)
| | - Jin Ho Lee
- Department of Advanced Materials , Hannam University , Daejeon 34054 , Republic of Korea
| | | | | | | | | | | | - June-Ho Byun
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine, Gyeongsang National University Hospital, Institute of Health Sciences , Gyeongsang National University , Jinju 52727 , Republic of Korea
| | | |
Collapse
|
25
|
Smith RA, Chua R, Carnachan SM, Tan CL, Sims IM, Hinkley SF, Nurcombe V, Cool SM. Retention of the Structure and Function of Heparan Sulfate Biomaterials After Gamma Irradiation. Tissue Eng Part A 2018; 24:729-739. [DOI: 10.1089/ten.tea.2017.0263] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Raymond A.A. Smith
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - R.J.E. Chua
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - Susan M. Carnachan
- The Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Clarissa L.L. Tan
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - Ian M. Sims
- The Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Simon F.R. Hinkley
- The Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Victor Nurcombe
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University-Imperial College London, Singapore
| | - Simon M. Cool
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
26
|
Kim S, Cui ZK, Kim PJ, Jung LY, Lee M. Design of hydrogels to stabilize and enhance bone morphogenetic protein activity by heparin mimetics. Acta Biomater 2018; 72:45-54. [PMID: 29597024 DOI: 10.1016/j.actbio.2018.03.034] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 03/15/2018] [Accepted: 03/19/2018] [Indexed: 12/21/2022]
Abstract
Although bone morphogenetic protein-2 (BMP-2) is known to be the most potent stimulator available for bone formation, a major barrier to widespread clinical use is its inherent instability and absence of an adequate delivery system. Heparin is being widely used in controlled release systems due to its strong binding ability and protective effect for many growth factor proteins. In this work, we developed a hydrogel surface that can mimic heparin to stabilize BMP-2 and to enhance osteogenesis by introducing heparin-mimicking sulfonated molecules such as poly-vinylsulfonic acid (PVSA) or poly-4-styrenesulfonic acid (PSS), into photo-crosslinkable hydrogel. Bioactivity of BMP-2 was well preserved in the presence of polysulfonates during exposure to various therapeutically relevant stressors. The heparin-mimicking sulfonated hydrogels were effective to bind BMP-2 compared to unmodified MeGC hydrogel and significantly enhanced osteogenic differentiation of encapsulated bone marrow stromal cells (BMSCs) without the addition of exogenous BMP-2. The sulfonated hydrogels were effective in delivering exogenous BMP-2 with reduced initial burst and increased BMSCs osteogenesis induced by BMP-2. These findings suggest a novel hydrogel platform for sequestering and stabilizing BMP-2 to enhance osteoinductive activity in bone tissue engineering. STATEMENT OF SIGNIFICANCE Although bone morphogenetic protein-2 (BMP-2) is believed to be the most potent cytokine for bone regeneration, its clinical applications require supraphysiological BMP dosage due to its intrinsic instability and fast enzymatic degradation, leading to worrisome side effects. This study demonstrates a novel hydrogel platform that mimics a natural protector of BMPs, heparin, to sequester and stabilize BMP-2 for increased osteoinductive signaling. This study will achieve the stabilization of BMPs with prolonged bioactivity by a synthetic heparin mimic that has not been examined previously. Moreover, the heparin mimetic hydrogel surface can augment endogenous BMP activity by sequestering and localizing the cell-produced BMPs. The additional knowledge gained from this study may suggest basis for future development of material-based therapeutics for tissue engineering.
Collapse
|
27
|
Prolonged delivery of BMP-2 by a non-polymer hydrogel for bone defect regeneration. Drug Deliv Transl Res 2017; 8:178-190. [DOI: 10.1007/s13346-017-0451-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
28
|
Lukasova V, Buzgo M, Sovkova V, Dankova J, Rampichova M, Amler E. Osteogenic differentiation of 3D cultured mesenchymal stem cells induced by bioactive peptides. Cell Prolif 2017; 50. [PMID: 28714176 DOI: 10.1111/cpr.12357] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 05/10/2017] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVES Bioactive peptides derived from receptor binding motifs of native proteins are a potent source of bioactive molecules that can induce signalling pathways. These peptides could substitute for osteogenesis promoting supplements. The work presented here compares three kinds of bioactive peptides derived from collagen III, bone morphogenetic protein 7 (BMP-7) and BMP-2 with their potential osteogenic activity on the model of porcine mesenchymal stem cells (pMSCs). MATERIALS AND METHODS pMSCs were cultured on electrospun polycaprolactone nanofibrous scaffolds with different concentrations of the bioactive peptides without addition of any osteogenic supplement. Analysis of pMSCs cultures included measurement of the metabolic activity and proliferation, immunofluorescence staining and also qPCR. RESULTS Results showed no detrimental effect of the bioactive peptides to cultured pMSCs. Based on qPCR analysis, the bioactive peptides are specific for osteogenic differentiation with no detectable expression of collagen II. Our results further indicate that peptide derived from BMP-2 protein promoted the expression of mRNA for osteocalcin (OCN) and collagen I significantly compared to control groups and also supported deposition of OCN as observed by immunostaining method. CONCLUSION The data suggest that bioactive peptide with an amino acid sequence of KIPKASSVPTELSAISTLYL derived from BMP-2 protein was the most potent for triggering osteogenic differentiation of pMSCs.
Collapse
Affiliation(s)
- Vera Lukasova
- Faculty of Science, Charles University in Prague, Prague, Czech Republic.,Laboratory of Tissue Engineering, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Matej Buzgo
- Institute of Biophysics, 2nd Faculty of Medicine, Charles University in Prague, Prague, Czech Republic.,University Center for Energy Efficient Buildings, Czech Technical University in Prague, Bustehrad, Czech Republic
| | - Vera Sovkova
- Laboratory of Tissue Engineering, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic.,Institute of Biophysics, 2nd Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Jana Dankova
- Laboratory of Tissue Engineering, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic.,Institute of Biophysics, 2nd Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Michala Rampichova
- Laboratory of Tissue Engineering, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic.,University Center for Energy Efficient Buildings, Czech Technical University in Prague, Bustehrad, Czech Republic
| | - Evzen Amler
- Laboratory of Tissue Engineering, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic.,Institute of Biophysics, 2nd Faculty of Medicine, Charles University in Prague, Prague, Czech Republic.,University Center for Energy Efficient Buildings, Czech Technical University in Prague, Bustehrad, Czech Republic
| |
Collapse
|
29
|
Mansour A, Mezour MA, Badran Z, Tamimi F. * Extracellular Matrices for Bone Regeneration: A Literature Review. Tissue Eng Part A 2017; 23:1436-1451. [PMID: 28562183 DOI: 10.1089/ten.tea.2017.0026] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The gold standard material for bone regeneration is still autologous bone, a mesenchymal tissue that consists mainly of extracellular matrix (ECM) (90% v/v) and little cellular content (10% v/v). However, the fact that decellularized allogenic bone grafts often present a clinical performance comparable to autologous bone grafts demonstrates the crucial role of ECM in bone regeneration. For long, the mechanism by which bone allografts function was not clear, but recent research has unveiled many unique characteristics of ECM that seem to play a key role in tissue regeneration. This is further confirmed by the fact that synthetic biomaterials with composition and properties resembling bone ECM present excellent bone regeneration properties. In this context, ECM molecules such as glycosaminoglycans (GAGs) and self-assembly peptides (SAPs) can improve the performance of bone regeneration biomaterials. Moreover, decellularized ECM derived either from native tissues such as bone, cartilage, skin, and tooth germs or from cells such as osteoblasts, chondrocytes, and stem cells has shown promising results in bone regeneration applications. Understanding the role of ECM in bone regeneration is crucial for the development of the next generation of biomaterials for bone tissue engineering. In this sense, this review addresses the state-of-the-art on this subject matter.
Collapse
Affiliation(s)
- Alaa Mansour
- 1 Faculty of Dentistry, McGill University , Montreal, Canada
| | | | - Zahi Badran
- 1 Faculty of Dentistry, McGill University , Montreal, Canada .,2 Department of Periodontology (CHU/UIC 11, INSERM UMR 1229-RMeS), Faculty of Dental Surgery, University of Nantes , Nantes, France
| | - Faleh Tamimi
- 1 Faculty of Dentistry, McGill University , Montreal, Canada
| |
Collapse
|
30
|
Hettiaratchi MH, Chou C, Servies N, Smeekens JM, Cheng A, Esancy C, Wu R, McDevitt TC, Guldberg RE, Krishnan L. Competitive Protein Binding Influences Heparin-Based Modulation of Spatial Growth Factor Delivery for Bone Regeneration. Tissue Eng Part A 2017; 23:683-695. [PMID: 28338419 PMCID: PMC5549832 DOI: 10.1089/ten.tea.2016.0507] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 02/08/2017] [Indexed: 11/12/2022] Open
Abstract
Tissue engineering strategies involving the in vivo delivery of recombinant growth factors are often limited by the inability of biomaterials to spatially control diffusion of the delivered protein within the site of interest. The poor spatiotemporal control provided by porous collagen sponges, which are used for the clinical delivery of bone morphogenetic protein-2 (BMP-2) for bone regeneration, has necessitated the use of supraphysiological protein doses, leading to inflammation and heterotopic ossification. This study describes a novel tissue engineering strategy to spatially control rapid BMP-2 diffusion from collagen sponges in vivo by creating a high-affinity BMP-2 sink around the collagen sponge. We designed an electrospun poly-ɛ-caprolactone nanofiber mesh containing physically entrapped heparin microparticles, which have been previously demonstrated to bind and retain large amounts of BMP-2. Nanofiber meshes containing 0.05 and 0.10 mg of microparticles/cm2 demonstrated increased BMP-2 binding and decreased BMP-2 release in vitro compared with meshes without microparticles. However, when microparticle-containing meshes were used in vivo to limit the diffusion of BMP-2 delivered by using collagen sponges in a rat femoral defect, no differences in heterotopic ossification or biomechanical properties were observed. Further investigation revealed that, although BMP-2 binding to heparin microparticles was rapid, the presence of serum components attenuated microparticle-BMP-2 binding and increased BMP-2 release in vitro. These observations provide a plausible explanation for the results observed in vivo and suggest that competitive protein binding in vivo may hinder the ability of affinity-based biomaterials to modulate growth factor delivery.
Collapse
Affiliation(s)
- Marian H. Hettiaratchi
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - Catherine Chou
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - Nicholas Servies
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - Johanna M. Smeekens
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia
| | - Albert Cheng
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
- The George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia
| | - Camden Esancy
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
| | - Ronghu Wu
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
| | - Todd C. McDevitt
- The Gladstone Institute of Cardiovascular Disease, San Francisco, California
- The Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California
| | - Robert E. Guldberg
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
- The George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia
| | - Laxminarayanan Krishnan
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
31
|
Todeschi MR, El Backly RM, Varghese OP, Hilborn J, Cancedda R, Mastrogiacomo M. Host cell recruitment patterns by bone morphogenetic protein-2 releasing hyaluronic acid hydrogels in a mouse subcutaneous environment. Regen Med 2017; 12:525-539. [DOI: 10.2217/rme-2017-0023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: This study aimed to identify host cell recruitment patterns in a mouse model in response to rhBMP-2 releasing hyaluronic acid hydrogels and influence of added nano-hydroxyapatite particles on rhBMP-2 release and pattern of bone formation. Materials & methods: Implanted gels were retrieved after implantation and cells were enzymatically dissociated for flow cytometric analysis. Percentages of macrophages, progenitor endothelial cells and putative mesenchymal stem cells were measured. Implants were evaluated for BMP-2 release by ELISA and by histology to monitor tissue formation. Results & conclusion: Hyaluronic acid+BMP-2 gels influenced the inflammatory response in the bone healing microenvironment. Host-derived putative mesenchymal stem cells were major contributors. Addition of hydroxyapatite nanoparticles modified the release pattern of rhBMP-2, resulting in enhanced bone formation.
Collapse
Affiliation(s)
- Maria R Todeschi
- Department of Experimental Medicine (DIMES), University of Genoa & IRCCS Policlinico San Martino, Genoa, Italy
| | - Rania M El Backly
- Department of Experimental Medicine (DIMES), University of Genoa & IRCCS Policlinico San Martino, Genoa, Italy
- Endodontics, Conservative Dentistry Department, Faculty of Dentistry, Alexandria University, Alexandria, Egypt
| | - Oommen P Varghese
- Department of Chemistry, Angstrom Laboratory, Uppsala University, Uppsala, Sweden
| | - Jöns Hilborn
- Department of Chemistry, Angstrom Laboratory, Uppsala University, Uppsala, Sweden
| | - Ranieri Cancedda
- Department of Experimental Medicine (DIMES), University of Genoa & IRCCS Policlinico San Martino, Genoa, Italy
| | - Maddalena Mastrogiacomo
- Department of Experimental Medicine (DIMES), University of Genoa & IRCCS Policlinico San Martino, Genoa, Italy
| |
Collapse
|
32
|
Mafina MK, Sullivan AC, Hing KA. Use of a fluorescent probe to monitor the enhanced affinity of rh-BMP-2 to silicated-calcium phosphate synthetic bone graft substitutes under competitive conditions. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 80:207-212. [PMID: 28866158 DOI: 10.1016/j.msec.2017.05.142] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 05/21/2017] [Accepted: 05/28/2017] [Indexed: 11/24/2022]
Abstract
A comparative investigation was undertaken on 1-2mm sized granules of two forms of synthetic bone graft substitute (SBG) with identical pore structure but varied bulk chemistry, stoichiometric hydroxyapatite (HA) and silicate substituted (0.8wt% Si) hydroxyapatite (SA), to assess the influence of SBG chemistry on the relative affinity of an osteogenic growth factor (GF), recombinant human bone morphogenetic protein-2 (rhBMP-2). A previously described novel fluorescent probe, fluoresceinthioureidoaminocaproic acid (FTCA), was covalently attached to rhBMP-2 to give FTCA-rhBMP-2 and facilitate the quantitative monitoring of GF uptake and release from the two chemistries of SBG. The relative affinity of rhBMP-2 for the HA and SA granules was assessed at a physiologically relevant concentration of 300ngmL-1 from three (increasingly complex) environments; phosphate buffered saline (PBS), minimum Eagles' medium (MEM) and serum supplemented MEM (SCEM) in order to closely mimic clinical bone repair procedures. The results demonstrated that rhBMP-2 affinity to SBGs was highly sensitive to both SBG chemistry and the composition of the local environment. Under the most physiologically relevant competitive conditions of SCEM, rhBMP-2 showed greater affinity to SA (P<0.05) such that 50% of the rhBMP-2 in solution was adsorbed to the SA granules after only 15min, as compared to 30% adsorbed to the HA granules. Subsequent investigation of the desorption of adsorbed GF from the SBGs demonstrated that a significantly higher percentage of the adsorbed rhBMP-2 was desorbed from HA as compared to SA granules. Together, these observations suggested that at physiologically relevant concentrations and conditions, rhBMP-2 has a greater affinity to silicate-substituted hydroxyapatite as compared to stoichiometric hydroxyapatite, which may in part explain the enhanced osteoconductivity and reported osteoinductivity for silicate-substituted hydroxyapatite based SBGs.
Collapse
Affiliation(s)
- Marc-Krystelle Mafina
- School of Engineering and Materials Science, Queen Mary, University of London, Bancroft Road, Mile End, London E1 4NS, United Kingdom.
| | - Alice C Sullivan
- School of Biological and Chemical Sciences, Queen Mary, University of London, Bancroft Road, Mile End, London E1 4NS, United Kingdom.
| | - Karin A Hing
- School of Engineering and Materials Science, Queen Mary, University of London, Bancroft Road, Mile End, London E1 4NS, United Kingdom.
| |
Collapse
|
33
|
Huber E, Pobloth AM, Bormann N, Kolarczik N, Schmidt-Bleek K, Schell H, Schwabe P, Duda GN, Wildemann B. * Demineralized Bone Matrix as a Carrier for Bone Morphogenetic Protein-2: Burst Release Combined with Long-Term Binding and Osteoinductive Activity Evaluated In Vitro and In Vivo. Tissue Eng Part A 2017; 23:1321-1330. [PMID: 28351338 DOI: 10.1089/ten.tea.2017.0005] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
To allow bone defect regeneration, autologous bone grafting still represents the gold standard. However, autograft harvesting has limitations, including an additional surgery, donor site morbidity, and limited availability. Demineralized bone matrix (DBM) would represent an alternative, yet lacks sufficient osteoinductive properties. Combining DBM with a potent agent, such as bone morphogenetic protein-2 (BMP-2) might be a feasible alternative approach, optimizing an established grafting material with strong osteoinductive properties. A unique mixing device has been developed that enables perioperative handling to reach a homogeneous and standardized paste for bone defect filling. DBM proved in vitro to be a suitable carrier for BMP-2, with a documented release over 56 days at concentrations sufficient to stimulate osteogenic differentiation. At the end of the elution experiment, 56 days, bioactive BMP was still captured within the DBM. Using a sheep drill hole defect model, DBM perioperatively mixed with BMP-2 showed strong osteoinductive properties comparable to those of autologous bone and outnumbering the one of DBM alone or empty defects. Bone defect healing was enabled at diaphyseal and metaphyseal defects and thus BMP-2-doped DBM represented an easy perioperative enriching method and an efficient carrier for BMP-2. With the comparability to the clinical gold standard autologous bone, DBM mixed with BMP-2 might serve as possible alternative grafting material enabling a controlled osteogenic stimulation.
Collapse
Affiliation(s)
- Elisabeth Huber
- 1 Julius Wolff Institut, Charité-Universitätsmedizin Berlin , Berlin, Germany .,2 Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin , Berlin, Germany
| | - Anne-Marie Pobloth
- 1 Julius Wolff Institut, Charité-Universitätsmedizin Berlin , Berlin, Germany .,2 Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin , Berlin, Germany
| | - Nicole Bormann
- 1 Julius Wolff Institut, Charité-Universitätsmedizin Berlin , Berlin, Germany .,2 Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin , Berlin, Germany
| | - Nicolai Kolarczik
- 1 Julius Wolff Institut, Charité-Universitätsmedizin Berlin , Berlin, Germany .,2 Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin , Berlin, Germany
| | - Katharina Schmidt-Bleek
- 1 Julius Wolff Institut, Charité-Universitätsmedizin Berlin , Berlin, Germany .,2 Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin , Berlin, Germany
| | - Hanna Schell
- 1 Julius Wolff Institut, Charité-Universitätsmedizin Berlin , Berlin, Germany .,2 Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin , Berlin, Germany
| | - Philipp Schwabe
- 3 Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin , Berlin, Germany
| | - Georg N Duda
- 1 Julius Wolff Institut, Charité-Universitätsmedizin Berlin , Berlin, Germany .,2 Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin , Berlin, Germany
| | - Britt Wildemann
- 1 Julius Wolff Institut, Charité-Universitätsmedizin Berlin , Berlin, Germany .,2 Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin , Berlin, Germany
| |
Collapse
|
34
|
Picke AK, Salbach-Hirsch J, Hintze V, Rother S, Rauner M, Kascholke C, Möller S, Bernhardt R, Rammelt S, Pisabarro MT, Ruiz-Gómez G, Schnabelrauch M, Schulz-Siegmund M, Hacker MC, Scharnweber D, Hofbauer C, Hofbauer LC. Sulfated hyaluronan improves bone regeneration of diabetic rats by binding sclerostin and enhancing osteoblast function. Biomaterials 2016; 96:11-23. [DOI: 10.1016/j.biomaterials.2016.04.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 04/18/2016] [Accepted: 04/19/2016] [Indexed: 01/03/2023]
|
35
|
Kissling S, Seidenstuecker M, Pilz IH, Suedkamp NP, Mayr HO, Bernstein A. Sustained release of rhBMP-2 from microporous tricalciumphosphate using hydrogels as a carrier. BMC Biotechnol 2016; 16:44. [PMID: 27206764 PMCID: PMC4874020 DOI: 10.1186/s12896-016-0275-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 05/12/2016] [Indexed: 11/25/2022] Open
Abstract
Background Tissue engineering and bone substitutes are subjects of intensive ongoing research. If the healing of bone fractures is delayed, osteoinductive materials that induce mesenchymal stem cells (MSCs) to form bone are necessary. The use of Bone Morphogenetic Protein - 2 is a common means to enhance effectiveness and accelerate the healing process. A delivery system that maintains and releases BMP biological activity in controlled fashion at the surgical site while preventing systemic diffusion (and thereby the risk of undesirable effects by controlling the amount of protein implanted) is essential. In this study, we aimed to test a cylindrical TCP-scaffold (porosity ~ 40 %, mean pore size 5 μm, high interconnectivity) in comparison to BMP-2. Recombinant human BMP-2 was dissolved in different hydrogels as a carrier, namely gelatin and alginate cross-linked with CaCl2-solution, or a solution of GDL and CaCO3. FITC-labeled Protein A was used as a model substance for rhBMP-2 in the pre-trials. For loading, the samples were put in a flow chamber and sealed with silicone rings. Using a directional vacuum, the samples were loaded with the alginate-BMP-2-mixture and the loading success monitored by observing changes in a fluorescent dye (FITC labeled Protein A) under a fluorescence microscope. A fluorescence reader and ELISA were employed to measure the release. Efficacy was determined in cell culture experiments (MG63 cells) via Live-Dead-Assay, FACS, WST-1-Assay, pNPP alkaline phosphatase assay and confocal microscopy. For statistical analysis, we calculated the mean and standard deviation and carried out an analysis of variance. Results Directional vacuum makes it possible to load nearly 100 % of the interconnected micropores with alginate mixed with rhBMP-2. Using alginate hardened with CaCl2 as a carrier, BMP-2's release can be decelerated significantly longer than with other hydrogels - eg, for over 28 days. The effects on osteoblast-like cells were an increase of the growth rate and expression of alkaline phosphatase while triggering no toxic effect. Conclusion The rhBMP-2-loaded microporous TCP scaffolds possess proliferative and osteoinductive potential. Alginate helps to lower the local growth factor dose below the cytotoxic limit, and allows the release period to be lengthened by at least 28 days. Electronic supplementary material The online version of this article (doi:10.1186/s12896-016-0275-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Steffen Kissling
- Center for Surgery, Department of Orthopedics and Trauma Surgery, Medical Center - University of Freiburg, Hugstetter Str.55, D-79106, Freiburg, Germany.
| | - Michael Seidenstuecker
- Center for Surgery, Department of Orthopedics and Trauma Surgery, Medical Center - University of Freiburg, Hugstetter Str.55, D-79106, Freiburg, Germany
| | - Ingo H Pilz
- Center for Surgery, Department of Orthopedics and Trauma Surgery, Medical Center - University of Freiburg, Hugstetter Str.55, D-79106, Freiburg, Germany
| | - Norbert P Suedkamp
- Center for Surgery, Department of Orthopedics and Trauma Surgery, Medical Center - University of Freiburg, Hugstetter Str.55, D-79106, Freiburg, Germany
| | - Hermann O Mayr
- Center for Surgery, Department of Orthopedics and Trauma Surgery, Medical Center - University of Freiburg, Hugstetter Str.55, D-79106, Freiburg, Germany
| | - Anke Bernstein
- Center for Surgery, Department of Orthopedics and Trauma Surgery, Medical Center - University of Freiburg, Hugstetter Str.55, D-79106, Freiburg, Germany
| |
Collapse
|
36
|
Kim HY, Lee JH, Yun JW, Park JH, Park BW, Rho GJ, Jang SJ, Park JS, Lee HC, Yoon YM, Hwang TS, Lee DH, Byun JH, Oh SH. Development of Porous Beads to Provide Regulated BMP-2 Stimulation for Varying Durations: In Vitro and In Vivo Studies for Bone Regeneration. Biomacromolecules 2016; 17:1633-42. [PMID: 27068184 DOI: 10.1021/acs.biomac.6b00009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
It is commonly accepted that the sustained release of bone morphogenetic protein-2 (BMP-2) can enhance bone regeneration and minimize its safety issues. However, little is known regarding the appropriate duration of BMP-2 stimulation for sufficient osteogenic differentiation and new bone formation because of the short half-life of BMP-2 in the physiological environment and the lack of a well-defined delivery matrix that can regulate the release period of BMP-2. In this study, we prepared porous poly(lactic-co-glycolic acid) (PLGA) beads with different surface pore sizes that can regulate the release period of BMP-2 (i.e., 7, 17, and 30 days) while providing the BMP-2 concentration required for bone regeneration. Our findings in both in vitro cell culture and in vivo animal studies using these BMP-2-loaded beads demonstrate that release of BMP-2 within 7 days affects only the initial differentiation of human periosteum-derived cells (hPDCs) and does not significantly enhance their subsequent differentiation into mature functional cells. However, extending the duration of BMP-2 stimulation over 17 days can provide a suitable environment for osteogenic differentiation of hPDCs and new bone formation.
Collapse
Affiliation(s)
- Ho Yong Kim
- Department of Nanobiomedical Science, Dankook University , Cheonan 330-714, Korea
| | - Jin Ho Lee
- Department of Advanced Materials, Hannam University , Daejeon 305-811, Korea
| | - Jeong-Won Yun
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University , Jinju 660-702, Korea
| | - Jin-Ho Park
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University , Jinju 660-702, Korea
| | - Bong-Wook Park
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University , Jinju 660-702, Korea
| | - Gyu-Jin Rho
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University , Jinju 660-701, Korea
| | - Si-Jung Jang
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University , Jinju 660-701, Korea
| | - Ji-Sung Park
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University , Jinju 660-701, Korea
| | - Hee-Chun Lee
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Gyeongsang National University , Jinju 660-701, Korea
| | - Young Min Yoon
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Gyeongsang National University , Jinju 660-701, Korea
| | - Tae Sung Hwang
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Gyeongsang National University , Jinju 660-701, Korea
| | - Dong Hoon Lee
- Department of Anatomy, Gyeongsang National University School of Medicine, Institute of Health Sciences, Gyeongsang National University , Jinju 660-702, Korea
| | - June-Ho Byun
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University , Jinju 660-702, Korea
| | - Se Heang Oh
- Department of Nanobiomedical Science, Dankook University , Cheonan 330-714, Korea
| |
Collapse
|
37
|
Chen MH, Weng JJ, Cheng CT, Wu RC, Huang SC, Wu CE, Chung YH, Liu CY, Chang MH, Chen MH, Chiang KC, Yeh TS, Su Y, Yeh CN. ALDH1A3, the Major Aldehyde Dehydrogenase Isoform in Human Cholangiocarcinoma Cells, Affects Prognosis and Gemcitabine Resistance in Cholangiocarcinoma Patients. Clin Cancer Res 2016; 22:4225-35. [PMID: 27076629 DOI: 10.1158/1078-0432.ccr-15-1800] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 03/15/2016] [Indexed: 12/20/2022]
Abstract
PURPOSE Intrahepatic cholangiocarcinoma is a fatal primary liver cancer resulting from diagnosis at an advanced stage. Understanding the mechanisms of drug resistance and metastasis of cholangiocarcinoma may improve the disease prognosis. Enhanced aldehyde dehydrogenase (ALDH) activity is suggested to be associated with increased drug resistance and the metastasis. This study aims to investigate the roles of the ALDH isoforms in cholangiocarcinoma. EXPERIMENTAL DESIGN Aldefluor assays, RT-PCR, and Western blot analysis were used to identify the major ALDH isoforms contributing to Aldefluor activity in human cholangiocarcinoma cell lines. We manipulated isoform expression in HuCCT1 cells to elucidate the role of ALDH1A3 in the malignant progression of these cells. Finally, we used immunohistochemical staining to evaluate the clinical significance of ALDH1A3 in 77 hepatectomized cholangiocarcinoma patients and an additional 31 patients with advanced cholangiocarcinoma who received gemcitabine-based therapy. RESULTS ALDH(high) cholangiocarcinoma cells not only migrated faster but were more resistant to gemcitabine. Among the 19 ALDH isoforms studied, ALDH1A3 was found to be the main contributor to Aldefluor activity. In addition, we also found that knockdown of ALDH1A3 expression in HuCCT1 cells markedly reduced not only their sensitivity to gemcitabine, which might be attributed to a decreased expression of ribonucleotide reductase M1, but also their migration. Most importantly, this enzyme was also identified as an independent poor prognostic factor for patients with intrahepatic cholangiocarcinoma, as well as a prognostic biomarker of gemcitabine-treated patients. CONCLUSIONS ALDH1A3 plays an important role in enhancing malignant behavior of cholangiocarcinoma and serves as a new therapeutic target. Clin Cancer Res; 22(16); 4225-35. ©2016 AACR.
Collapse
Affiliation(s)
- Ming-Huang Chen
- School of Medicine, National Yang-Ming University, Taipei, Taiwan. Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jing-Jie Weng
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan. Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Chi-Tung Cheng
- Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| | - Ren-Chin Wu
- Department of Pathology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| | - Shih-Chiang Huang
- Department of Pathology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| | - Chiao-En Wu
- Department of Hematology-Oncology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Hsiu Chung
- Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chun-Yu Liu
- School of Medicine, National Yang-Ming University, Taipei, Taiwan. Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Mu-Hsin Chang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan. Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ming-Han Chen
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Kun-Chun Chiang
- Department of Surgery, Chang Gung Memorial Hospital, Keelung; Chang Gung University, Taoyuan, Taiwan
| | - Ta-Sen Yeh
- Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| | - Yeu Su
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan.
| | - Chun-Nan Yeh
- Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
38
|
Balmayor ER, Geiger JP, Aneja MK, Berezhanskyy T, Utzinger M, Mykhaylyk O, Rudolph C, Plank C. Chemically modified RNA induces osteogenesis of stem cells and human tissue explants as well as accelerates bone healing in rats. Biomaterials 2016; 87:131-146. [PMID: 26923361 DOI: 10.1016/j.biomaterials.2016.02.018] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 02/10/2016] [Accepted: 02/16/2016] [Indexed: 01/24/2023]
Abstract
Limitations associated to the use of growth factors represent a major hurdle to musculoskeletal regeneration. On the one hand, they are needed to induce neo-tissue formation for the substitution of a necrotic or missing tissue. On the other hand, these factors are used in supraphysiological concentrations, are short lived and expensive and result in many side effects. Here we develop a gene transfer strategy based on the use of chemically modified mRNA (cmRNA) coding for human bone morphogenetic protein 2 (hBMP-2) that is non-immunogenic and highly stable when compared to unmodified mRNA. Transfected stem cells secrete hBMP-2, show elevated alkaline phosphatase levels and upregulated expression of RunX2, ALP, Osterix, Osteocalcin, Osteopontin and Collagen Type I genes. Mineralization was induced as seen by positive Alizarin red staining. hBMP-2 cmRNA transfected human fat tissue also yielded an osteogenic response in vitro as indicated by expression of hBMP-2, RunX2, ALP and Collagen Type I. Delivering hBMP-2 cmRNA to a femur defect in a rat model results in new bone tissue formation as early as 2 weeks after application of very low doses. Overall, our studies demonstrate the feasibility and therapeutic potential of a new cmRNA-based gene therapy strategy that is safe and efficient. When applied clinically, this approach could overcome BMP-2 growth factor associated limitations in bone regeneration.
Collapse
Affiliation(s)
- Elizabeth R Balmayor
- Institute of Molecular Immunology and Experimental Oncology, Technical University Munich, Ismaninger Str. 22, 81675 Munich, Germany; Ethris GmbH, Semmelweisstr. 3, 82152 Planegg, Germany; Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University Munich, Ismaninger Str. 22, 81675 Munich, Germany.
| | | | | | - Taras Berezhanskyy
- Institute of Molecular Immunology and Experimental Oncology, Technical University Munich, Ismaninger Str. 22, 81675 Munich, Germany; Ethris GmbH, Semmelweisstr. 3, 82152 Planegg, Germany
| | | | - Olga Mykhaylyk
- Institute of Molecular Immunology and Experimental Oncology, Technical University Munich, Ismaninger Str. 22, 81675 Munich, Germany; Ethris GmbH, Semmelweisstr. 3, 82152 Planegg, Germany
| | | | - Christian Plank
- Institute of Molecular Immunology and Experimental Oncology, Technical University Munich, Ismaninger Str. 22, 81675 Munich, Germany; Ethris GmbH, Semmelweisstr. 3, 82152 Planegg, Germany.
| |
Collapse
|
39
|
Gothard D, Smith EL, Kanczler JM, Black CR, Wells JA, Roberts CA, White LJ, Qutachi O, Peto H, Rashidi H, Rojo L, Stevens MM, El Haj AJ, Rose FRAJ, Shakesheff KM, Oreffo ROC. In Vivo Assessment of Bone Regeneration in Alginate/Bone ECM Hydrogels with Incorporated Skeletal Stem Cells and Single Growth Factors. PLoS One 2015; 10:e0145080. [PMID: 26675008 PMCID: PMC4684226 DOI: 10.1371/journal.pone.0145080] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 11/27/2015] [Indexed: 12/21/2022] Open
Abstract
The current study has investigated the use of decellularised, demineralised bone extracellular matrix (ECM) hydrogel constructs for in vivo tissue mineralisation and bone formation. Stro-1-enriched human bone marrow stromal cells were incorporated together with select growth factors including VEGF, TGF-β3, BMP-2, PTHrP and VitD3, to augment bone formation, and mixed with alginate for structural support. Growth factors were delivered through fast (non-osteogenic factors) and slow (osteogenic factors) release PLGA microparticles. Constructs of 5 mm length were implanted in vivo for 28 days within mice. Dense tissue assessed by micro-CT correlated with histologically assessed mineralised bone formation in all constructs. Exogenous growth factor addition did not enhance bone formation further compared to alginate/bone ECM (ALG/ECM) hydrogels alone. UV irradiation reduced bone formation through degradation of intrinsic growth factors within the bone ECM component and possibly also ECM cross-linking. BMP-2 and VitD3 rescued osteogenic induction. ALG/ECM hydrogels appeared highly osteoinductive and delivery of angiogenic or chondrogenic growth factors led to altered bone formation. All constructs demonstrated extensive host tissue invasion and vascularisation aiding integration and implant longevity. The proposed hydrogel system functioned without the need for growth factor incorporation or an exogenous inducible cell source. Optimal growth factor concentrations and spatiotemporal release profiles require further assessment, as the bone ECM component may suffer batch variability between donor materials. In summary, ALG/ECM hydrogels provide a versatile biomaterial scaffold for utilisation within regenerative medicine which may be tailored, ultimately, to form the tissue of choice through incorporation of select growth factors.
Collapse
Affiliation(s)
- David Gothard
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD, United Kingdom
- * E-mail: (DG); (ROCO)
| | - Emma L. Smith
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD, United Kingdom
| | - Janos M. Kanczler
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD, United Kingdom
| | - Cameron R. Black
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD, United Kingdom
| | - Julia A. Wells
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD, United Kingdom
| | - Carol A. Roberts
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD, United Kingdom
| | - Lisa J. White
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling, School of Pharmacy, University of Nottingham, Centre for Biomolecular Sciences, University Park, Nottingham, NG7 2RD, United Kingdom
| | - Omar Qutachi
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling, School of Pharmacy, University of Nottingham, Centre for Biomolecular Sciences, University Park, Nottingham, NG7 2RD, United Kingdom
| | - Heather Peto
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling, School of Pharmacy, University of Nottingham, Centre for Biomolecular Sciences, University Park, Nottingham, NG7 2RD, United Kingdom
| | - Hassan Rashidi
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling, School of Pharmacy, University of Nottingham, Centre for Biomolecular Sciences, University Park, Nottingham, NG7 2RD, United Kingdom
| | - Luis Rojo
- Department of Materials, Imperial College London, Royal School of Mines, London, SW7 2AZ, United Kingdom
- Department of Bioengineering, Imperial College London, South Kensington Campus, London, SW7 2AZ, United Kingdom
- Institute for Biomedical Engineering, Imperial College London, South Kensington Campus, London, SW7 2AZ, United Kingdom
- Biomaterials, Biomimetics, Biophotonics Research Division, King's College London, Dental Institute, Guy's Hospital, Tower Wing, London Bridge, London SE1 9RT, United Kingdom
| | - Molly M. Stevens
- Department of Materials, Imperial College London, Royal School of Mines, London, SW7 2AZ, United Kingdom
- Department of Bioengineering, Imperial College London, South Kensington Campus, London, SW7 2AZ, United Kingdom
- Institute for Biomedical Engineering, Imperial College London, South Kensington Campus, London, SW7 2AZ, United Kingdom
| | - Alicia J. El Haj
- Institute for Science and Technology in Medicine, Keele University, Guy Hilton Research Centre, Stoke-on-Trent, ST4 7BQ, United Kingdom
| | - Felicity R. A. J. Rose
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling, School of Pharmacy, University of Nottingham, Centre for Biomolecular Sciences, University Park, Nottingham, NG7 2RD, United Kingdom
| | - Kevin M. Shakesheff
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling, School of Pharmacy, University of Nottingham, Centre for Biomolecular Sciences, University Park, Nottingham, NG7 2RD, United Kingdom
- Locate Therapeutics Limited, MediCity, Nottingham, NG90 6BH, United Kingdom
| | - Richard O. C. Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD, United Kingdom
- * E-mail: (DG); (ROCO)
| |
Collapse
|
40
|
Migliorini E, Valat A, Picart C, Cavalcanti-Adam EA. Tuning cellular responses to BMP-2 with material surfaces. Cytokine Growth Factor Rev 2015; 27:43-54. [PMID: 26704296 DOI: 10.1016/j.cytogfr.2015.11.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 11/13/2015] [Indexed: 02/08/2023]
Abstract
Bone morphogenetic protein 2 (BMP-2) has been known for decades as a strong osteoinductive factor and for clinical applications is combined solely with collagen as carrier material. The growing concerns regarding side effects and the importance of BMP-2 in several developmental and physiological processes have raised the need to improve the design of materials by controlling BMP-2 presentation. Inspired by the natural cell environment, new material surfaces have been engineered and tailored to provide both physical and chemical cues that regulate BMP-2 activity. Here we describe surfaces designed to present BMP-2 to cells in a spatially and temporally controlled manner. This is achieved by trapping BMP-2 using physicochemical interactions, either covalently grafted or combined with other extracellular matrix components. In the near future, we anticipate that material science and biology will integrate and further develop tools for in vitro studies and potentially bring some of them toward in vivo applications.
Collapse
Affiliation(s)
- Elisa Migliorini
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, D-70569 Stuttgart, Germany
- Department of Biophysical Chemistry, University of Heidelberg, INF 253, D-69120 Heidelberg, Germany, Tel: +49-6221-54 5064
| | - Anne Valat
- CNRS-UMR 5628, LMGP, 3 parvis L.Néel, F-38 016 Grenoble, France
- University Grenoble Alpes, Grenoble Institute of Technology, LMGP, 3 parvis Louis Néel, F-28016 Grenoble, France
- INSERM U823, ERL CNRS5284, Université de Grenoble Alpes, Institut Albert Bonniot, Site Santé, BP170, 38042 Grenoble cedex 9, France, Tel: +33-04-56529311
| | - Catherine Picart
- CNRS-UMR 5628, LMGP, 3 parvis L.Néel, F-38 016 Grenoble, France
- University Grenoble Alpes, Grenoble Institute of Technology, LMGP, 3 parvis Louis Néel, F-28016 Grenoble, France
| | - Elisabetta Ada Cavalcanti-Adam
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, D-70569 Stuttgart, Germany
- Department of Biophysical Chemistry, University of Heidelberg, INF 253, D-69120 Heidelberg, Germany, Tel: +49-6221-54 5064
| |
Collapse
|
41
|
Repair of segmental ulna defects using a β-TCP implant in combination with a heparan sulfate glycosaminoglycan variant. Acta Biomater 2015; 28:193-204. [PMID: 26384700 DOI: 10.1016/j.actbio.2015.09.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 08/06/2015] [Accepted: 09/07/2015] [Indexed: 10/23/2022]
Abstract
Given the wide spread clinical use of ceramic-based bone void fillers, we sought to determine the efficacy of an FDA-approved β-tricalcium phosphate bone graft substitute (JAX™) in combination with a carboxymethyl cellulose (CMC) handling agent that included a particular heparan glycosaminoglycan (GAG) variant, herein referred to as HS3. Having recently demonstrated efficacy of a combination collagen/HS3 device, we further aimed to determine the support that HS3 could offer a handling agent used to administer a more tissue-relevant bone void filler. This study evaluated the JAX™-HS3 combination device in 1.5 cm critical-sized defects in the ulna bones of 27 male New Zealand White rabbits. Treatment groups consisted of JAX™ applied with CMC alone, or JAX™ with CMC containing either 30 μg or 100 μg of the HS3 GAG. Data based on radiographic, μCT, mechanical, and histological analyses at 4 and 8 weeks post-surgery, clearly demonstrate enhanced new bone formation in the JAX™-HS3 combination treated defects compared to treatment with JAX™ alone. The efficacy of such a combination advocates for inclusion of HS3 in handling agents used in the preparation of various bone void fillers being used in orthopaedic surgery. STATEMENT OF SIGNIFICANCE Synthetic bone grafts and demineralized bone matrices are gaining prominence as alternatives to autologous and allogeneic bone grafts and are frequently administered in granular form, necessitating their combination with a handling agent. Typical handling agents include glycerol, gelatin, cellulose, hyaluronic acid and lecithin, formulated as hydrogels, which can be further enhanced by the addition of heparan sulfate (HS) glycosaminoglycans that augment the osteostimulatory properties of the graft. Here we assessed the efficacy of β-TCP granules combined with a hydrogel consisting of carboxymethyl cellulose and the HS variant (HS3) previously shown to enhance osteogenic healing. The data advocates for HS3 to be included during the formulation of hydrogel-based carriers that support the various bone void fillers being used in orthopaedic surgery.
Collapse
|
42
|
Mode of heparin attachment to nanocrystalline hydroxyapatite affects its interaction with bone morphogenetic protein-2. Biointerphases 2015; 10:04A308. [DOI: 10.1116/1.4933109] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
43
|
Sánchez-Duffhues G, Hiepen C, Knaus P, Ten Dijke P. Bone morphogenetic protein signaling in bone homeostasis. Bone 2015; 80:43-59. [PMID: 26051467 DOI: 10.1016/j.bone.2015.05.025] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 04/11/2015] [Accepted: 05/20/2015] [Indexed: 01/06/2023]
Abstract
Bone morphogenetic proteins (BMPs) are cytokines belonging to the transforming growth factor-β (TGF-β) superfamily. They play multiple functions during development and tissue homeostasis, including regulation of the bone homeostasis. The BMP signaling pathway consists in a well-orchestrated manner of ligands, membrane receptors, co-receptors and intracellular mediators, that regulate the expression of genes controlling the normal functioning of the bone tissues. Interestingly, BMP signaling perturbation is associated to a variety of low and high bone mass diseases, including osteoporosis, bone fracture disorders and heterotopic ossification. Consistent with these findings, in vitro and in vivo studies have shown that BMPs have potent effects on the activity of cells regulating bone function, suggesting that manipulation of the BMP signaling pathway may be employed as a therapeutic approach to treat bone diseases. Here we review the recent advances on BMP signaling and bone homeostasis, and how this knowledge may be used towards improved diagnosis and development of novel treatment modalities. This article is part of a Special Issue entitled "Muscle Bone Interactions".
Collapse
Affiliation(s)
- Gonzalo Sánchez-Duffhues
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Center, The Netherlands
| | - Christian Hiepen
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany; Berlin Brandenburg School of Regenerative Therapies (BSRT), Charité Universitätsmedizin, Berlin, Germany
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany; Berlin Brandenburg School of Regenerative Therapies (BSRT), Charité Universitätsmedizin, Berlin, Germany.
| | - Peter Ten Dijke
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Center, The Netherlands.
| |
Collapse
|
44
|
Balmayor ER. Targeted delivery as key for the success of small osteoinductive molecules. Adv Drug Deliv Rev 2015; 94:13-27. [PMID: 25959428 DOI: 10.1016/j.addr.2015.04.022] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 04/20/2015] [Accepted: 04/29/2015] [Indexed: 02/08/2023]
Abstract
Molecules such as growth factors, peptides and small molecules can guide cellular behavior and are thus important for tissue engineering. They are rapidly emerging as promising compounds for the regeneration of tissues of the musculoskeletal system. Growth factors have disadvantages such as high cost, short half-life, supraphysiological amounts needed, etc. Therefore, small molecules may be an alternative. These molecules have been discovered using high throughput screening. Small osteoinductive molecules exhibit several advantages over growth factors owing to their small sizes, such as high stability and non-immunogenicity. These molecules may stimulate directly signaling pathways that are important for osteogenesis. However, systemic application doesn't induce osteogenesis in most cases. Therefore, local administration is needed. This may be achieved by using a bone graft material providing additional osteoconductive properties. These graft materials can also act by themselves as a delivery matrix for targeted and local delivery. Furthermore, vascularization is necessary in the process of osteogenesis. Many of the small molecules are also capable of promoting vascularization of the tissue to be regenerated. Thus, in this review, special attention is given to molecules that are capable of inducing both angiogenesis and osteogenesis simultaneously. Finally, more recent preclinical and clinical uses in bone regeneration of those molecules are described, highlighting the needs for the clinical translation of these promising compounds.
Collapse
|
45
|
Surface-mediated delivery of siRNA from fibrin hydrogels for knockdown of the BMP-2 binding antagonist noggin. Acta Biomater 2015; 25:109-20. [PMID: 26234488 DOI: 10.1016/j.actbio.2015.07.045] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 06/28/2015] [Accepted: 07/29/2015] [Indexed: 12/26/2022]
Abstract
Antagonists and inhibitory molecules responsible for maintaining tissue homeostasis can present a significant barrier to healing when tissue engineering/regenerative medicine strategies are employed. One example of this situation is the up-regulation of antagonists such as noggin in response to increasing concentrations of bone morphogenetic protein-2 (BMP-2) present from endogenous bone repair processes or delivered exogenously from biomaterials (synthetic bone grafts). While recombinant human (rh)BMP-2 delivered from synthetic bone grafts has been shown to be an effective alternative to autografts and allografts, the supraphysiological doses of rhBMP-2 have led to clinically-adverse side effects. The high rhBMP-2 dosage may be required, in part, to overcome the presence of antagonists such as noggin. Small interfering RNA (siRNA) is an appealing approach to overcome this problem because it can knock-down antagonists or inhibitory molecules in a temporary manner. Here, we conducted fundamental studies on the delivery of siRNA from material surfaces as a means to knock-down antagonists like noggin. Non-viral cationic lipid (Lipofectamine)-siRNA complexes were delivered from a fibrin hydrogel surface to MC3T3-E1 preosteoblasts that were treated with a supraphysiological dose of rhBMP-2 to achieve noggin mRNA expression levels higher than cells naïve to rhBMP-2. Confocal microscopy and flow cytometry showed intracellular uptake of siRNA in over 98% of MC3T3-E1 cells after 48 h. Doses of 0.5 and 1 μg noggin siRNA were able to significantly reduce noggin mRNA to levels equivalent to those in MC3T3-E1 cells not exposed to rhBMP-2 with no effects on cell viability. STATEMENT OF SIGNIFICANCE Small interfering RNA (siRNA) has been considered for treatment of diseases ranging from Alzheimer's to cancer. However, the ability to use siRNA in conjunction with biomaterials to direct tissue regeneration processes has received relatively little attention. Using the bone morphogenetic protein 2 antagonist, noggin, as a model, this research describes an approach to knock-down molecules that are inhibitory to desired regenerative pathways at the mRNA level via siRNA delivery from a hydrogel surface. Interactions between the material (fibrin) surface and polycation-siRNA complexes, release of the siRNA from the material surface, high levels of cellular uptake/internalization of siRNA, and significant knockdown of the targeting (noggin) mRNA are demonstrated. Broader future applications include those to nerve regeneration, cardiovascular tissue engineering, directing (stem) cell behavior, and mitigating inflammatory responses to materials.
Collapse
|
46
|
Polyelectrolyte Complex Carrier Enhances Therapeutic Efficiency and Safety Profile of Bone Morphogenetic Protein-2 in Porcine Lumbar Interbody Fusion Model. Spine (Phila Pa 1976) 2015; 40:964-73. [PMID: 25893351 DOI: 10.1097/brs.0000000000000935] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN Porcine lumbar interbody fusion model. OBJECTIVE This study evaluates the effect of polyelectrolyte complex (PEC) carrier in enhancing the therapeutic efficiency and safety profile of bone morphogenetic protein-2 (BMP-2) in a large animal model. SUMMARY OF BACKGROUND DATA Extremely large amounts of BMP-2 are administered to achieve consistent spinal fusion, which has led to complications. Heparin-modified PEC carrying reduced BMP-2 doses of 0.5 μg was demonstrated to achieve consistent spinal fusion with reduction of complications in rodent model. The purpose of this study was to evaluate whether PEC could improve the therapeutic efficiency of BMP-2 in porcine model. METHODS Three-segment (L3-L6) anterior lumbar interbody fusions with instrumentation were performed on 6 pigs using 3 different doses of BMP-2, namely, (1) 50 μg, (2) 150 μg, and (3) 300 μg. The BMP-2 was delivered using heparin-modified alginate microbeads loaded into biodegradable cage. Fusion performance was evaluated after 3 months. RESULTS Manual palpation and micro-computed tomography showed consistent fusion in all experimental groups. Heterotopic bone formation beyond the cage implant area was more evident in group 2 and group 3 than in group 1. Similarly, superior bone microstructure was observed in the new bone with the lowered BMP-2 dose. Biomechanical evaluation revealed enhanced stiffness of the operated segments compared with nonoperated segments (P < 0.05). Mechanical stability was maintained despite dose reduction of BMP-2. Although the mineral apposition rate was higher in group 3, unsatisfactory bony microstructure with decreased trabecular number was observed in group 3 compared with group 1. CONCLUSION PEC carrying low doses of BMP-2 achieved consistent interbody fusion. We observed dose-related reduction in heterotopic ossification without compromising the stability of the fused segments. PEC carrier reduces the efficacious doses of BMP-2. This could enhance the safety profile of BMP-2 and reduce dose- and carrier-related complications. LEVEL OF EVIDENCE N/A.
Collapse
|
47
|
Gohil SV, Brittain SB, Kan HM, Drissi H, Rowe DW, Nair LS. Evaluation of enzymatically crosslinked injectable glycol chitosan hydrogel. J Mater Chem B 2015; 3:5511-5522. [PMID: 32262522 DOI: 10.1039/c5tb00663e] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Enzymatically cross-linkable phenol-conjugated glycol chitosan was prepared by reacting glycol chitosan with 3-(4-hydroxyphenyl)propionic acid (HPP). The chemical modification was confirmed by FTIR, 1H-NMR and UV spectroscopy. Glycol chitosan hydrogels (HPP-GC) with or without rhBMP-2 were prepared by the oxidative coupling of the substituted phenol groups in the presence of hydrogen peroxide and horse radish peroxidase. Rheological characterization demonstrated the feasibility of developing hydrogels with varying storage moduli by changing the polymer concentration. The gel presented a microporous structure with pore sizes ranging from 50-350 μm. The good viability of encapsulated 7F2 osteoblasts indicated non-toxicity of the gelation conditions. In vitro release of rhBMP-2 in phosphate buffer solution showed ∼11% release in 360 h. The ability of the hydrogel to maintain the in vivo bioactivity of rhBMP-2 was evaluated in a bilateral critical size calvarial bone defect model in Col3.6 transgenic fluorescent reporter mice. The presence of fluorescent green osteoblast cells with overlying red alizarin complexone and yellow stain indicating osteoclast TRAP activity confirmed active cell-mediated mineralization and remodelling process at the implantation site. The complete closure of the defect site at 4 and 8 weeks post implantation demonstrated the potent osteoinductivity of the rhBMP-2 containing gel.
Collapse
Affiliation(s)
- Shalini V Gohil
- Department of Orthopaedic Surgery, UConn Health, E-7041, MC-3711, 263 Farmington Avenue, Farmington, Connecticut 06030, USA.
| | | | | | | | | | | |
Collapse
|
48
|
Quinlan E, Thompson EM, Matsiko A, O'Brien FJ, López-Noriega A. Long-term controlled delivery of rhBMP-2 from collagen–hydroxyapatite scaffolds for superior bone tissue regeneration. J Control Release 2015; 207:112-9. [DOI: 10.1016/j.jconrel.2015.03.028] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/19/2015] [Accepted: 03/23/2015] [Indexed: 01/11/2023]
|
49
|
Novel Protamine-Based Polyelectrolyte Carrier Enhances Low-Dose rhBMP-2 in Posterolateral Spinal Fusion. Spine (Phila Pa 1976) 2015; 40:613-21. [PMID: 25705961 DOI: 10.1097/brs.0000000000000841] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN A rodent posterolateral spinal fusion model. OBJECTIVE This study evaluated a protamine-based polyelectrolyte complex (PEC) developed to use heparin in enhancing the biological activity of low-dose recombinant human bone morphogenetic protein-2 (rhBMP-2) in spinal fusion. SUMMARY OF BACKGROUND DATA rhBMP-2 is commonly regarded as the most potent bone-inducing molecule. However, poor pharmacokinetics and short in vivo half-life means that large amounts of the bioactive growth factor are required for consistent clinical outcomes. This has been associated with a number of adverse tissue reactions including seroma and heterotopic ossification. Glycosaminoglycans including heparin are known to stabilize rhBMP-2 bioactivity. Previous studies with poly-L-lysine (PLL) and heparin-based PEC carriers amplified the therapeutic efficacy of low-dose BMP-2. However, questions remained on the eventual clinical applicability of relatively cytotoxic PLL. In the present study, a protamine-based PEC carrier was designed to further enhance the safety and efficacy of BMP-2 by delivering lower dose within the therapeutic window. METHODS A polyelectrolyte shell was deposited on the surface of alginate microbead templates using the polycation (protamine)/polyanion (heparin) layer-by-layer polyelectrolyte self-assembly protocol. rhBMP-2 was loaded onto the outermost layer via heparin affinity binding. Loading and release of rhBMP-2 were evaluated in vitro. The bone-inductive ability of 20-fold reduction of rhBMP-2 with the different carrier vehicle was evaluated using a posterolateral spinal fusion model in rats. RESULTS In vitro uptake and release analysis, protamine-based PEC showed higher uptake and significantly enhanced control release than PLL-based PEC (P < 0.05). In vivo implantation with protamine-based and PLL-based PEC showed better fusion performances than absorbable collagen sponge-delivered same dose of rhBMP-2, and negative control group through manual palpation, micro-computed tomography, and histological analyses. CONCLUSION Solid posterolateral spinal fusion was achieved with 20-fold reduction of rhBMP-2 when delivered using protamine-based PEC carrier in the rat posterolateral spinal fusion model. LEVEL OF EVIDENCE N/A.
Collapse
|
50
|
Müller CW, Hildebrandt K, Gerich T, Krettek C, van Griensven M, Rosado Balmayor E. BMP-2-transduced human bone marrow stem cells enhance neo-bone formation in a rat critical-sized femur defect. J Tissue Eng Regen Med 2015; 11:1122-1131. [PMID: 25783748 DOI: 10.1002/term.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 01/08/2015] [Accepted: 01/15/2015] [Indexed: 12/15/2022]
Abstract
Synthetic graft materials are considered as possible substitutes for cancellous bone, but lack osteogenic and osteoinductive properties. In this study, we investigated how composite scaffolds of βTCP containing osteogenic human bone marrow mesenchymal stem cells (hBMSCs) and osteoinductive bone morphogenetic protein-2 (BMP-2) influenced the process of fracture healing. hBMSCs were loaded into βTCP scaffolds 24 h before implantation in a rat critical-sized bone defect. hBMSCs were either stimulated with rhBMP-2 or transduced with BMP-2 by gene transfer. The effect of both protein stimulation and gene transfer was compared for osteogenic outcome. X-rays were conducted at weeks 0, 1, 3, 6, 9 and 12 post-operatively. In addition, bone-labelling fluorochromes were applied at 0, 3, 6 and 9 weeks. Histological analysis was performed for the amount of callus tissue and cartilage formation. At 6 weeks, the critical-sized defect in 33% of the rats treated with the Ad-BMP-2-transduced hBMSCs/βTCP scaffolds was radiographically bridged. In contrast, in only 10% of the rats treated with rhBMP2/hBMSCs, 12 weeks post-treatment, the bone defect was closed in all treated rats of the Ad-BMP-2 group except for one. Histology showed significantly higher amounts of callus formation in both Ad-BMP-2- and rhBMP-2-treated rats. The amount of neocartilage was less pronounced in both BMP-2-related groups. In summary, scaffolds with BMP-2-transduced hBMSCs performed better than those with the rhBMP2/hBMSCs protein. These results suggest that combinations of osteoconductive biomaterials with genetically modified MSCs capable of secreting osteoinductive proteins may represent a promising alternative for bone regeneration. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
| | | | - Torsten Gerich
- Department of Trauma Surgery, Centre Hospitalier de Luxembourg (CHL), Luxembourg
| | | | - Martijn van Griensven
- Department of Experimental Trauma Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany
| | - Elizabeth Rosado Balmayor
- Department of Experimental Trauma Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany
| |
Collapse
|