1
|
Wang C, Shou Z, Xu C, Huo K, Liu W, Liu H, Zan X, Wang Q, Li L. Enhancing the Implant Osteointegration via Supramolecular Co-Assembly Coating with Early Immunomodulation and Cell Colonization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2410595. [PMID: 39806935 DOI: 10.1002/advs.202410595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 12/30/2024] [Indexed: 01/16/2025]
Abstract
Osteointegration, the effective coupling between an implant and bone tissue, is a highly intricate biological process. The initial stages of bone-related immunomodulation and cellular colonization play crucial roles, but have received limited attention. Herein, a novel supramolecular co-assembled coating of strontium (Sr)-doped metal polyphenol networks (MPN) modified with c(RGDfc) is developed and well-characterized, for eliciting an early immunomodulation and cellular colonization. The results showed that the (Sr-MPN)@RGD coating significantly regulated the polarization of macrophages to the M2 phenotype by controllable release of Sr, and promote the initial adhesion of bone marrow mesenchymal stem cells (BMSCs) by RGD presented on MPN. Notably, the (Sr-MPN)@RGD attenuated osteoclast differentiation and oxidative stress as well as enhanced osteoblast differentiation and angiogenesis due to macrophage polarization toward M2 phenotype, which in turn has a profound effect on neighboring cells through paracrine signaling. In vivo results showed that the (Sr-MPN)@RGD coating manifested superior osseointegration and bone maturation to the bare Ti-rod or Ti-rod coated with MPN and Sr-MPN. This work contributed to the design of multifunctional implant coatings that address the complex biological process of osteointegration from the perspective of orchestrating stem cell recruitment with immunomodulatory strategies.
Collapse
Affiliation(s)
- Chenglong Wang
- Department of Orthopaedics Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Zeyu Shou
- Department of Orthopedics, Zhuji People's Hospital of Zhejiang Province, Zhuji Affiliated Hospital of Wenzhou Medical University, Shaoxing, Zhejiang, 311800, China
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Chengwei Xu
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Kaiyuan Huo
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou, Zhejiang, 325001, China
| | - Wenjie Liu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou, Zhejiang, 325001, China
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, 450001, China
| | - Hao Liu
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, 450001, China
| | - Xingjie Zan
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou, Zhejiang, 325001, China
| | - Qing Wang
- Yongkang First People's Hospital of Wenzhou Medical University, Jinhua, 321300, China
| | - Lianxin Li
- Department of Orthopaedics Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| |
Collapse
|
2
|
Valeo M, Marie S, Rémy M, Menguy T, Le Coz C, Molinari M, Feuillie C, Granier F, Durrieu MC. Bioactive hydrogels based on lysine dendrigrafts as crosslinkers: tailoring elastic properties to influence hMSC osteogenic differentiation. J Mater Chem B 2024; 12:12508-12522. [PMID: 39576239 DOI: 10.1039/d4tb01578a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
Dendrigrafts are multivalent macromolecules with less ordered topology and higher branching than dendrimers. Exhibiting a high density of terminal amines, poly-L-lysine dendrigrafts of the fifth generation (DGL G5) allow hydrogel formation with tailorable crosslinking density and surface modification. This work presents DGL G5 as multifunctional crosslinkers in biomimetic PEG hydrogels to favour the osteogenic differentiation of human mesenchymal stem cells (hMSCs). DGL G5 reaction with dicarboxylic-acid PEG chains yielded amide networks of variable stiffness, measured at the macro and surface nanoscale. Oscillatory rheometry and compression afforded consistent values of Young's modulus, increasing from 8 to more than 30 kPa and correlating with DGL G5 concentration. At the surface level, AFM measurements showed the same tendency but higher E values, from approximately 15 to more than 100 kPa, respectively. To promote cell adhesion and differentiation, the hydrogels were functionalised with a GRGDSPC peptide and a biomimetic of the bone morphogenetic protein 2 (BMP-2), ensuring the same grafting concentrations (between 2.15 ± 0.54 and 2.28 ± 0.23 pmols mm-2) but different hydrogel stiffness. 6 h after seeding on functionalised hydrogels in serum-less media, hMSC showed nascent adhesions on the stiffer gels and greater spreading than on glass controls with serum. After two weeks in osteogenic media, hMSC seeded on the stiffer gels showed greater spreading, more polygonal morphologies and increased levels of osteopontin, an osteoblast marker, compared to controls, which peaked on 22 kPa-gels. Together, these results demonstrate that DGL G5-PEG hydrogel bioactivity can influence the adhesion, spreading and early commitment of hMSCs.
Collapse
Affiliation(s)
- Michele Valeo
- Université de Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | | | - Murielle Rémy
- Université de Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | | | - Cédric Le Coz
- Université de Bordeaux, CNRS, Bordeaux INP, LCPO, ENSMAC, F-33600 Pessac, France
| | - Michael Molinari
- Université de Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | - Cécile Feuillie
- Université de Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | | | | |
Collapse
|
3
|
Łuczak JW, Palusińska M, Matak D, Pietrzak D, Nakielski P, Lewicki S, Grodzik M, Szymański Ł. The Future of Bone Repair: Emerging Technologies and Biomaterials in Bone Regeneration. Int J Mol Sci 2024; 25:12766. [PMID: 39684476 DOI: 10.3390/ijms252312766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Bone defects and fractures present significant clinical challenges, particularly in orthopedic and maxillofacial applications. While minor bone defects may be capable of healing naturally, those of a critical size necessitate intervention through the use of implants or grafts. The utilization of traditional methodologies, encompassing autografts and allografts, is constrained by several factors. These include the potential for donor site morbidity, the restricted availability of suitable donors, and the possibility of immune rejection. This has prompted extensive research in the field of bone tissue engineering to develop advanced synthetic and bio-derived materials that can support bone regeneration. The optimal bone substitute must achieve a balance between biocompatibility, bioresorbability, osteoconductivity, and osteoinductivity while simultaneously providing mechanical support during the healing process. Recent innovations include the utilization of three-dimensional printing, nanotechnology, and bioactive coatings to create scaffolds that mimic the structure of natural bone and enhance cell proliferation and differentiation. Notwithstanding the advancements above, challenges remain in optimizing the controlled release of growth factors and adapting materials to various clinical contexts. This review provides a comprehensive overview of the current advancements in bone substitute materials, focusing on their biological mechanisms, design considerations, and clinical applications. It explores the role of emerging technologies, such as additive manufacturing and stem cell-based therapies, in advancing the field. Future research highlights the need for multidisciplinary collaboration and rigorous testing to develop advanced bone graft substitutes, improving outcomes and quality of life for patients with complex defects.
Collapse
Affiliation(s)
- Julia Weronika Łuczak
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Magdalenka, Poland
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Ciszewskiego 8, Bldg. 23, 02-786 Warsaw, Poland
| | - Małgorzata Palusińska
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Magdalenka, Poland
| | - Damian Matak
- European Biomedical Institute, 05-410 Jozefów, Poland
| | - Damian Pietrzak
- Division of Parasitology and Parasitic Diseases, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Paweł Nakielski
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawińskiego 5B, 02-106 Warsaw, Poland
| | - Sławomir Lewicki
- Institute of Outcomes Research, Maria Sklodowska-Curie Medical Academy, Pl. Żelaznej Bramy 10, 00-136 Warsaw, Poland
| | - Marta Grodzik
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Ciszewskiego 8, Bldg. 23, 02-786 Warsaw, Poland
| | - Łukasz Szymański
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Magdalenka, Poland
- European Biomedical Institute, 05-410 Jozefów, Poland
| |
Collapse
|
4
|
Ramaraju H, Garcia-Gomez E, McAtee AM, Verga AS, Hollister SJ. Shape memory cycle conditions impact human bone marrow stromal cell binding to RGD- and YIGSR-conjugated poly (glycerol dodecanedioate). Acta Biomater 2024; 186:246-259. [PMID: 39111679 DOI: 10.1016/j.actbio.2024.07.057] [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: 05/12/2024] [Revised: 07/21/2024] [Accepted: 07/30/2024] [Indexed: 08/16/2024]
Abstract
Bioresorbable shape memory polymers (SMP) are an emerging class of polymers that can help address several challenges associated with minimally invasive surgery by providing a solution for structural tissue repair. Like most synthetic polymer networks, SMPs require additional biorelevance and modification for biomedical applications. Methodologies used to incorporate bioactive ligands must preserve SMP thermomechanics and ensure biofunctionality following in vivo delivery. We have previously described the development of a novel thermoresponsive bioresorbable SMP, poly (glycerol dodecanedioate) (PGD). In this study, cell-adhesive peptide sequences RGD and YIGSR were conjugated with PGD. We investigated 1) the impact of conjugated peptides on the fixity (Rf), recovery (Rr), and recovery rate (dRr/dT), 2) the impact of conjugated peptides on cell binding, and 3) the impact of the shape memory cycle (Tprog) on conjugated peptide functionality towards binding human bone marrow stromal cells (BMSC). Peptide conjugation conditions impact fixity but not the recovery or recovery rate (p < 0.01). Peptide-conjugated substrates increased cell attachment and proliferation compared with controls (p < 0.001). Using complementary integrin binding cell-adhesive peptides increased proliferation compared with using single peptides (p < 0.05). Peptides bound to PGD substrates exhibited specificity to their respective integrin targets. Following the shape memory cycle, peptides maintained functionality and specificity depending on the shape memory cycle conditions (p < 0.001). The dissipation of strain energy during recovery can drive differential arrangement of conjugated sequences impacting functionality, an important design consideration for functionalized SMPs. STATEMENT OF SIGNIFICANCE: Shape memory elastomers are an emerging class of polymers that are well-suited for minimally invasive repair of soft tissues. Tissue engineering approaches commonly utilize biodegradable scaffolds to deliver instructive cues, including cells and bioactive signals. Delivering these instructive cues on biodegradable shape memory elastomers requires modification with bioactive ligands. Furthermore, it is necessary to ensure the specificity of the ligands to their biological targets when conjugated to the polymer. Moreover, the bioactive ligand functionality must be conserved after completing the shape memory cycle, for applications in tissue engineering.
Collapse
Affiliation(s)
- Harsha Ramaraju
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States.
| | - Elisa Garcia-Gomez
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Annabel M McAtee
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Adam S Verga
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Scott J Hollister
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| |
Collapse
|
5
|
Che Z, Sun Q, Zhao Z, Wu Y, Xing H, Song K, Chen A, Wang B, Cai M. Growth factor-functionalized titanium implants for enhanced bone regeneration: A review. Int J Biol Macromol 2024; 274:133153. [PMID: 38897500 DOI: 10.1016/j.ijbiomac.2024.133153] [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: 02/26/2024] [Revised: 06/02/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024]
Abstract
Titanium and titanium alloys are widely favored materials for orthopedic implants due to their exceptional mechanical properties and biological inertness. The additional benefit of sustained local release of bioactive substances further promotes bone tissue formation, thereby augmenting the osseointegration capacity of titanium implants and attracting increasing attention in bone tissue engineering. Among these bioactive substances, growth factors have shown remarkable osteogenic and angiogenic induction capabilities. Consequently, researchers have developed various physical, chemical, and biological loading techniques to incorporate growth factors into titanium implants, ensuring controlled release kinetics. In contrast to conventional treatment modalities, the localized release of growth factors from functionalized titanium implants not only enhances osseointegration but also reduces the risk of complications. This review provides a comprehensive examination of the types and mechanisms of growth factors, along with a detailed exploration of the methodologies used to load growth factors onto the surface of titanium implants. Moreover, it highlights recent advancements in the application of growth factors to the surface of titanium implants (Scheme 1). Finally, the review discusses current limitations and future prospects for growth factor-functionalized titanium implants. In summary, this paper presents cutting-edge design strategies aimed at enhancing the bone regenerative capacity of growth factor-functionalized titanium implants-a significant advancement in the field of enhanced bone regeneration.
Collapse
Affiliation(s)
- Zhenjia Che
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China.
| | - Qi Sun
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Zhenyu Zhao
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Yanglin Wu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Hu Xing
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Kaihang Song
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Aopan Chen
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Bo Wang
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China.
| | - Ming Cai
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China.
| |
Collapse
|
6
|
López-Serrano C, Côté-Paradis Y, Habenstein B, Loquet A, Le Coz C, Ruel J, Laroche G, Durrieu MC. Integrating Mechanics and Bioactivity: A Detailed Assessment of Elasticity and Viscoelasticity at Different Scales in 2D Biofunctionalized PEGDA Hydrogels for Targeted Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2024; 16:39165-39180. [PMID: 39041490 PMCID: PMC11600396 DOI: 10.1021/acsami.4c10755] [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: 06/28/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/24/2024]
Abstract
Methods for promoting and controlling the differentiation of human mesenchymal stem cells (hMSCs) in vitro before in vivo transplantation are crucial for the advancement of tissue engineering and regenerative medicine. In this study, we developed poly(ethylene glycol) diacrylate (PEGDA) hydrogels with tunable mechanical properties, including elasticity and viscoelasticity, coupled with bioactivity achieved through the immobilization of a mixture of RGD and a mimetic peptide of the BMP-2 protein. Despite the key relevance of hydrogel mechanical properties for cell culture, a standard for its characterization has not been proposed, and comparisons between studies are challenging due to the different techniques employed. Here, a comprehensive approach was employed to characterize the elasticity and viscoelasticity of these hydrogels, integrating compression testing, rheology, and atomic force microscopy (AFM) microindentation. Distinct mechanical behaviors were observed across different PEGDA compositions, and some consistent trends across multiple techniques were identified. Using a photoactivated cross-linker, we controlled the functionalization density independently of the mechanical properties. X-ray photoelectrin spectroscopy and fluorescence microscopy were employed to evaluate the functionalization density of the materials before the culturing of hMSCs on them. The cells cultured on all functionalized hydrogels expressed an early osteoblast marker (Runx2) after 2 weeks, even in the absence of a differentiation-inducing medium compared to our controls. Additionally, after only 1 week of culture with osteogenic differentiation medium, cells showed accelerated differentiation, with clear morphological differences observed among cells in the different conditions. Notably, cells on stiff but stress-relaxing hydrogels exhibited an overexpression of the osteocyte marker E11. This suggests that the combination of the functionalization procedure with the mechanical properties of the hydrogel provides a potent approach to promoting the osteogenic differentiation of hMSCs.
Collapse
Affiliation(s)
- Cristina López-Serrano
- Univ.
Bordeaux, CNRS, Bordeaux INP, CBMN, UMR
5248, Pessac 33600, France
- Laboratoire
d’Ingénierie de Surface, Centre de Recherche sur les
Matériaux Avancés, Département de Génie
des Mines, de la Métallurgie et des Matériaux, Université Laval, Québec, QC G1 V 0A6, Canada
- Axe
médecine régénératrice, Centre de Recherche
du Centre Hospitalier Universitaire de Québec, Hôpital
St-François d’Assise, Québec, QC G1L
3L5, Canada
| | - Yeva Côté-Paradis
- Laboratoire
d’Ingénierie de Surface, Centre de Recherche sur les
Matériaux Avancés, Département de Génie
des Mines, de la Métallurgie et des Matériaux, Université Laval, Québec, QC G1 V 0A6, Canada
- Axe
médecine régénératrice, Centre de Recherche
du Centre Hospitalier Universitaire de Québec, Hôpital
St-François d’Assise, Québec, QC G1L
3L5, Canada
| | - Birgit Habenstein
- Univ.
Bordeaux, CNRS, INSERM, IECB, US1, UAR 3033, F-33600 Pessac, France
| | - Antoine Loquet
- Univ.
Bordeaux, CNRS, INSERM, IECB, US1, UAR 3033, F-33600 Pessac, France
| | - Cédric Le Coz
- Univ.
Bordeaux, CNRS, Bordeaux INP, LCPO, UMR
5629, F-33600 Pessac, France
| | - Jean Ruel
- Département
de Génie Mécanique, Université
Laval, Québec, QC G1V 0A6, Canada
| | - Gaétan Laroche
- Laboratoire
d’Ingénierie de Surface, Centre de Recherche sur les
Matériaux Avancés, Département de Génie
des Mines, de la Métallurgie et des Matériaux, Université Laval, Québec, QC G1 V 0A6, Canada
- Axe
médecine régénératrice, Centre de Recherche
du Centre Hospitalier Universitaire de Québec, Hôpital
St-François d’Assise, Québec, QC G1L
3L5, Canada
| | | |
Collapse
|
7
|
Schuphan J, Stojanović N, Lin YY, Buhl EM, Aveic S, Commandeur U, Schillberg S, Fischer H. A Combination of Flexible Modified Plant Virus Nanoparticles Enables Additive Effects Resulting in Improved Osteogenesis. Adv Healthc Mater 2024; 13:e2304243. [PMID: 38417028 DOI: 10.1002/adhm.202304243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/16/2024] [Indexed: 03/01/2024]
Abstract
Plant virus nanoparticles (VNPs) genetically engineered to present osteogenic cues provide a promising method for biofunctionalizing hydrogels in bone tissue engineering. Flexible Potato virus X (PVX) nanoparticles substantially enhance the attachment and differentiation of human mesenchymal stem cells (hMSCs) by presenting the RGD motif, hydroxyapatite-binding peptide (HABP), or consecutive polyglutamates (E8) in a concentration-dependent manner. Therefore, it is hypothesized that Tobacco mosaic virus nanoparticles, which present 1.6 times more functional peptides than PVX, will meliorate such an impact. This study hypothesizes that cultivating hMSCs on a surface coated with a combination of two VNPs presenting peptides for either cell attachment or mineralization can achieve additionally enhancing effects on osteogenesis. Calcium minerals deposited by differentiating hMSCs increases two to threefold for this combination, while the Alkaline Phosphatase activity of hMSCs grown on the PVX-RGD/PVX-HABP-coated surface significantly surpasses any other VNP combination. Superior additive effects are observed for the first time by employing a combination of VNPs with varying functionalities. It is found that the flexible VNP geometry plays a more critical role than the concentration of functional peptides. In conclusion, various peptide-presenting plant VNPs exhibit an additive enhancing effect offering significant potential for effectively functionalizing cell-containing hydrogels in bone tissue engineering.
Collapse
Affiliation(s)
- Juliane Schuphan
- Institute for Molecular Biotechnology, RWTH Aachen University, Worringerweg 1, 52074, Aachen, Germany
| | - Natalija Stojanović
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Ying-Ying Lin
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Eva Miriam Buhl
- Electron Microscopy Facility, Institute of Pathology, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Sanja Aveic
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Ulrich Commandeur
- Institute for Molecular Biotechnology, RWTH Aachen University, Worringerweg 1, 52074, Aachen, Germany
| | - Stefan Schillberg
- Institute for Molecular Biotechnology, RWTH Aachen University, Worringerweg 1, 52074, Aachen, Germany
| | - Horst Fischer
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| |
Collapse
|
8
|
Beeren IAO, Dos Santos G, Dijkstra PJ, Mota C, Bauer J, Ferreira H, Reis RL, Neves N, Camarero-Espinosa S, Baker MB, Moroni L. A facile strategy for tuning the density of surface-grafted biomolecules for melt extrusion-based additive manufacturing applications. Biodes Manuf 2024; 7:277-291. [PMID: 38818303 PMCID: PMC11133161 DOI: 10.1007/s42242-024-00286-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 04/23/2024] [Indexed: 06/01/2024]
Abstract
Melt extrusion-based additive manufacturing (ME-AM) is a promising technique to fabricate porous scaffolds for tissue engineering applications. However, most synthetic semicrystalline polymers do not possess the intrinsic biological activity required to control cell fate. Grafting of biomolecules on polymeric surfaces of AM scaffolds enhances the bioactivity of a construct; however, there are limited strategies available to control the surface density. Here, we report a strategy to tune the surface density of bioactive groups by blending a low molecular weight poly(ε-caprolactone)5k (PCL5k) containing orthogonally reactive azide groups with an unfunctionalized high molecular weight PCL75k at different ratios. Stable porous three-dimensional (3D) scaffolds were then fabricated using a high weight percentage (75 wt.%) of the low molecular weight PCL5k. As a proof-of-concept test, we prepared films of three different mass ratios of low and high molecular weight polymers with a thermopress and reacted with an alkynated fluorescent model compound on the surface, yielding a density of 201-561 pmol/cm2. Subsequently, a bone morphogenetic protein 2 (BMP-2)-derived peptide was grafted onto the films comprising different blend compositions, and the effect of peptide surface density on the osteogenic differentiation of human mesenchymal stromal cells (hMSCs) was assessed. After two weeks of culturing in a basic medium, cells expressed higher levels of BMP receptor II (BMPRII) on films with the conjugated peptide. In addition, we found that alkaline phosphatase activity was only significantly enhanced on films containing the highest peptide density (i.e., 561 pmol/cm2), indicating the importance of the surface density. Taken together, these results emphasize that the density of surface peptides on cell differentiation must be considered at the cell-material interface. Moreover, we have presented a viable strategy for ME-AM community that desires to tune the bulk and surface functionality via blending of (modified) polymers. Furthermore, the use of alkyne-azide "click" chemistry enables spatial control over bioconjugation of many tissue-specific moieties, making this approach a versatile strategy for tissue engineering applications. Graphic abstract Supplementary Information The online version contains supplementary material available at 10.1007/s42242-024-00286-2.
Collapse
Affiliation(s)
- I. A. O. Beeren
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - G. Dos Santos
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
- 3B’s Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - P. J. Dijkstra
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - C. Mota
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - J. Bauer
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - H. Ferreira
- 3B’s Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Rui L. Reis
- 3B’s Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - N. Neves
- 3B’s Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - S. Camarero-Espinosa
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
- POLYMAT, University of the Basque Country UPV/EHU, 20018 Donostia/San Sebastián, Spain
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
| | - M. B. Baker
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - L. Moroni
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
9
|
Li S, Zhang S, Dong S, Zhao M, Zhang W, Zhang C, Wu Z. Stiffness and BMP-2 Mimetic Peptide Jointly Regulate the Osteogenic Differentiation of Rat Bone Marrow Stromal Cells in a Gelatin Cryogel. Biomacromolecules 2024; 25:890-902. [PMID: 38180887 DOI: 10.1021/acs.biomac.3c01045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2024]
Abstract
Both biochemical and mechanical cues could regulate the function of stem cells, but the interaction mechanism of their signaling pathway remains unclear, especially in the three-dimensional (3D) culture mode. Higher matrix stiffness promotes osteogenic differentiation of stem cells, and bone morphogenic protein-2 (BMP-2) has been clinically applied to promote bone regeneration. Here, the crosstalk of extracellular mechanical signals on BMP-2 signaling was investigated in rat bone marrow stromal cells (rMSCs) cultured inside cryogels with interconnective pores. Stiff cryogel independently promoted osteogenic differentiation and enhanced the autocrine secretion of BMP-2, thus stimulating increased phosphorylation levels of the Smad1/5/8 complex. BMP-2 mimetic peptide (BMMP) and high cryogel stiffness jointly guided the osteogenic differentiation of rMSCs. Inhibition of rho-associated kinase (ROCK) by Y-27632 or inhibition of nonmuscle myosin II (NM II) by blebbistatin showed that osteogenesis induction by BMP-2 signaling, as well as autocrine secretion of BMP-2 and phosphorylation of the Smad complex, requires the involvement of cytoskeletal tension and ROCK pathway signaling. An interconnective microporous cryogel scaffold promoted rMSC osteogenic differentiation by combining matrix stiffness and BMMP, and it accelerated critical cranial defect repair in the rat model.
Collapse
Affiliation(s)
- Sijing Li
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
- Logistics Department, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Shixiong Zhang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Shuao Dong
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Mengen Zhao
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
- Shenzhen Institute for Drug Control, Shenzhen Testing Center of Medical Devices, Shenzhen, Guangdong 518057, China
| | - Wei Zhang
- Department of Outpatient, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Chao Zhang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Zhaoying Wu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| |
Collapse
|
10
|
Kapat K, Kumbhakarn S, Sable R, Gondane P, Takle S, Maity P. Peptide-Based Biomaterials for Bone and Cartilage Regeneration. Biomedicines 2024; 12:313. [PMID: 38397915 PMCID: PMC10887361 DOI: 10.3390/biomedicines12020313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
The healing of osteochondral defects (OCDs) that result from injury, osteochondritis, or osteoarthritis and bear lesions in the cartilage and bone, pain, and loss of joint function in middle- and old-age individuals presents challenges to clinical practitioners because of non-regenerative cartilage and the limitations of current therapies. Bioactive peptide-based osteochondral (OC) tissue regeneration is becoming more popular because it does not have the immunogenicity, misfolding, or denaturation problems associated with original proteins. Periodically, reviews are published on the regeneration of bone and cartilage separately; however, none of them addressed the simultaneous healing of these tissues in the complicated heterogeneous environment of the osteochondral (OC) interface. As regulators of cell adhesion, proliferation, differentiation, angiogenesis, immunomodulation, and antibacterial activity, potential therapeutic strategies for OCDs utilizing bone and cartilage-specific peptides should be examined and investigated. The main goal of this review was to study how they contribute to the healing of OCDs, either alone or in conjunction with other peptides and biomaterials.
Collapse
Affiliation(s)
- Kausik Kapat
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata 700054, West Bengal, India
| | - Sakshi Kumbhakarn
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata 700054, West Bengal, India
| | - Rahul Sable
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata 700054, West Bengal, India
| | - Prashil Gondane
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata 700054, West Bengal, India
| | - Shruti Takle
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata 700054, West Bengal, India
| | - Pritiprasanna Maity
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
11
|
Zhao T, Li X, Li H, Deng H, Li J, Yang Z, He S, Jiang S, Sui X, Guo Q, Liu S. Advancing drug delivery to articular cartilage: From single to multiple strategies. Acta Pharm Sin B 2023; 13:4127-4148. [PMID: 37799383 PMCID: PMC10547919 DOI: 10.1016/j.apsb.2022.11.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/09/2022] [Accepted: 10/28/2022] [Indexed: 11/27/2022] Open
Abstract
Articular cartilage (AC) injuries often lead to cartilage degeneration and may ultimately result in osteoarthritis (OA) due to the limited self-repair ability. To date, numerous intra-articular delivery systems carrying various therapeutic agents have been developed to improve therapeutic localization and retention, optimize controlled drug release profiles and target different pathological processes. Due to the complex and multifactorial characteristics of cartilage injury pathology and heterogeneity of the cartilage structure deposited within a dense matrix, delivery systems loaded with a single therapeutic agent are hindered from reaching multiple targets in a spatiotemporal matched manner and thus fail to mimic the natural processes of biosynthesis, compromising the goal of full cartilage regeneration. Emerging evidence highlights the importance of sequential delivery strategies targeting multiple pathological processes. In this review, we first summarize the current status and progress achieved in single-drug delivery strategies for the treatment of AC diseases. Subsequently, we focus mainly on advances in multiple drug delivery applications, including sequential release formulations targeting various pathological processes, synergistic targeting of the same pathological process, the spatial distribution in multiple tissues, and heterogeneous regeneration. We hope that this review will inspire the rational design of intra-articular drug delivery systems (DDSs) in the future.
Collapse
Affiliation(s)
- Tianyuan Zhao
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Xu Li
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, 999077, Hong Kong, China
| | - Hao Li
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Haoyuan Deng
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Jianwei Li
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Zhen Yang
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing 100044, China
| | - Songlin He
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Shuangpeng Jiang
- Department of Joint Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Xiang Sui
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
| | - Quanyi Guo
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Shuyun Liu
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
12
|
Zhang Y, Rémy M, Apartsin E, Prouvé E, Feuillie C, Labrugère C, Cam N, Durrieu MC. Controlling differentiation of stem cells via bioactive disordered cues. Biomater Sci 2023; 11:6116-6134. [PMID: 37602410 DOI: 10.1039/d3bm00605k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Ideal bone tissue engineering is to induce bone regeneration through the synergistic integration of biomaterial scaffolds, bone progenitor cells, and bone-forming factors. Biomimetic scaffolds imitate the native extracellular matrix (ECM) and are often utilized in vitro as analogues of the natural ECM to facilitate investigations of cell-ECM interactions and processes. In vivo, the cellular microenvironment has a crucial impact on regulating cell behavior and functions. A PET surface was activated and then functionalized with mimetic peptides to promote human mesenchymal stem cell (hMSC) adhesion and differentiation into an osteogenic lineage. Spray technology was used to randomly micropattern peptides (RGD and BMP-2 mimetic peptides) on the PET surface. The distribution of the peptides grafted on the surface, the roughness of the surfaces and the chemistry of the surfaces in each step of the treatment were ascertained by atomic force microscopy, fluorescence microscopy, time-of-flight secondary ion mass spectrometry, Toluidine Blue O assay, and X-ray photoelectron spectroscopy. Subsequently, cell lineage differentiation was evaluated by quantifying the expression of immunofluorescence markers: osteoblast markers (Runx-2, OPN) and osteocyte markers (E11, DMP1, and SOST). In this article, we hypothesized that a unique combination of bioactive micro/nanopatterns on a polymer surface improves the rate of morphology change and enhances hMSC differentiation. In DMEM, after 14 days, disordered micropatterned surfaces with RGD and BMP-2 led to a higher osteoblast marker expression than surfaces with a homogeneous dual peptide conjugation. Finally, hMSCs cultured in osteogenic differentiation medium (ODM) showed accelerated cell differentiation. In ODM, our results highlighted the expression of osteocyte markers when hMSCs were seeded on PET surfaces with random micropatterns.
Collapse
Affiliation(s)
- Yujie Zhang
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | - Murielle Rémy
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | - Evgeny Apartsin
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | - Emilie Prouvé
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | - Cécile Feuillie
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | | | - Nithavong Cam
- Univ. Bordeaux, CNRS, PLACAMAT, UAR 3626, F-33600 Pessac, France
| | | |
Collapse
|
13
|
Ghribi N, Guay-Bégin AA, Bilem I, Chevallier P, Auger FA, Ruel J, Laroche G. Peptide grafting on intraosseous transcutaneous amputation prostheses to promote sealing with skin cells: Potential to limit infections. J Biomed Mater Res A 2023; 111:688-700. [PMID: 36680491 DOI: 10.1002/jbm.a.37505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/22/2023]
Abstract
The long-term success of intraosseous transcutaneous amputation prostheses (ITAPs) mainly relies on dermal attachment of skin cells to the implant. Otherwise, bacteria can easily penetrate through the interface between the implant and the skin. Therefore, infection at this implant/skin interface remains a significant complication in orthopedic surgeries in which these prostheses are required. Two main strategies were investigated to prevent these potential infection problems which consist in either establishing a strong sealing at the skin/implant interface or on eradicating infections by killing bacteria. In this work, two adhesion peptides, either KRGDS or KYIGSR and one antimicrobial peptide, Magainin 2 (Mag 2), were covalently grafted via phosphonate anchor arms to the surface of the Ti6Al4V ELI (extra low interstitials) material, commonly used to manufacture ITAPs. X-ray photoelectron spectroscopy, contact angle, and confocal microscopy analyses enabled to validate the covalent and stable grafting of these three peptides. Dermal fibroblasts cultures on bare Ti6Al4V ELI surfaces and functionalized ones displayed a good cell adhesion and proliferation on all samples. However, cell spreading and viability appeared to be improved on grafted surfaces, especially for those conjugated with KRGDS and Mag 2. Moreover, the dermal sheet attachment, was significantly higher on surfaces functionalized with Mag 2 as compared to the other surfaces. Therefore, the surface functionalization with the antimicrobial peptide Mag 2 seems to be the best approach for the targeted application, as it could play a dual role, inducing a strong skin adhesion while limiting infections on Ti6Al4V ELI materials.
Collapse
Affiliation(s)
- Nawel Ghribi
- Laboratoire d'ingénierie de surface (LIS), Centre de Recherche du CHU de Québec-Université Laval, Hôpital Saint-François d'Assise, Québec, Québec, Canada
- Département de génie des mines, de la métallurgie et des matériaux, Centre de recherche sur les Matériaux Avancés, Université Laval, Québec, Québec, Canada
| | - Andrée-Anne Guay-Bégin
- Laboratoire d'ingénierie de surface (LIS), Centre de Recherche du CHU de Québec-Université Laval, Hôpital Saint-François d'Assise, Québec, Québec, Canada
- Département de génie des mines, de la métallurgie et des matériaux, Centre de recherche sur les Matériaux Avancés, Université Laval, Québec, Québec, Canada
| | - Ibrahim Bilem
- Laboratoire d'ingénierie de surface (LIS), Centre de Recherche du CHU de Québec-Université Laval, Hôpital Saint-François d'Assise, Québec, Québec, Canada
- Département de génie des mines, de la métallurgie et des matériaux, Centre de recherche sur les Matériaux Avancés, Université Laval, Québec, Québec, Canada
| | - Pascale Chevallier
- Laboratoire d'ingénierie de surface (LIS), Centre de Recherche du CHU de Québec-Université Laval, Hôpital Saint-François d'Assise, Québec, Québec, Canada
- Département de génie des mines, de la métallurgie et des matériaux, Centre de recherche sur les Matériaux Avancés, Université Laval, Québec, Québec, Canada
| | - François A Auger
- Centre de Recherche du CHU de Québec-Université Laval, LOEX, Québec, Québec, Canada
| | - Jean Ruel
- Département de Génie mécanique, Université Laval, Québec, Québec, Canada
| | - Gaétan Laroche
- Laboratoire d'ingénierie de surface (LIS), Centre de Recherche du CHU de Québec-Université Laval, Hôpital Saint-François d'Assise, Québec, Québec, Canada
- Département de génie des mines, de la métallurgie et des matériaux, Centre de recherche sur les Matériaux Avancés, Université Laval, Québec, Québec, Canada
| |
Collapse
|
14
|
Oliver‐Cervelló L, Martin‐Gómez H, Mandakhbayar N, Jo Y, Cavalcanti‐Adam EA, Kim H, Ginebra M, Lee J, Mas‐Moruno C. Mimicking Bone Extracellular Matrix: From BMP-2-Derived Sequences to Osteogenic-Multifunctional Coatings. Adv Healthc Mater 2022; 11:e2201339. [PMID: 35941083 PMCID: PMC11468143 DOI: 10.1002/adhm.202201339] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Indexed: 01/28/2023]
Abstract
Cell-material interactions are regulated by mimicking bone extracellular matrix on the surface of biomaterials. In this regard, reproducing the extracellular conditions that promote integrin and growth factor (GF) signaling is a major goal to trigger bone regeneration. Thus, the use of synthetic osteogenic domains derived from bone morphogenetic protein 2 (BMP-2) is gaining increasing attention, as this strategy is devoid of the clinical risks associated with this molecule. In this work, the wrist and knuckle epitopes of BMP-2 are screened to identify peptides with potential osteogenic properties. The most active sequences (the DWIVA motif and its cyclic version) are combined with the cell adhesive RGD peptide (linear and cyclic variants), to produce tailor-made biomimetic peptides presenting the bioactive cues in a chemically and geometrically defined manner. Such multifunctional peptides are next used to functionalize titanium surfaces. Biological characterization with mesenchymal stem cells demonstrates the ability of the biointerfaces to synergistically enhance cell adhesion and osteogenic differentiation. Furthermore, in vivo studies in rat calvarial defects prove the capacity of the biomimetic coatings to improve new bone formation and reduce fibrous tissue thickness. These results highlight the potential of mimicking integrin-GF signaling with synthetic peptides, without the need for exogenous GFs.
Collapse
Affiliation(s)
- Lluís Oliver‐Cervelló
- BiomaterialsBiomechanics and Tissue Engineering GroupDepartment of Materials Science and EngineeringUniversitat Politècnica de Catalunya (UPC)Barcelona08019Spain
- Barcelona Research Center in Multiscale Science and EngineeringUPCBarcelona08019Spain
| | - Helena Martin‐Gómez
- BiomaterialsBiomechanics and Tissue Engineering GroupDepartment of Materials Science and EngineeringUniversitat Politècnica de Catalunya (UPC)Barcelona08019Spain
- Barcelona Research Center in Multiscale Science and EngineeringUPCBarcelona08019Spain
| | - Nandin Mandakhbayar
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan330‐714Republic of Korea
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan330‐714Republic of Korea
- Department of Biomaterials ScienceSchool of DentistryDankook UniversityCheonan330‐714Republic of Korea
| | - Young‐Woo Jo
- Neobiotech Co.Ltd R&D CenterSeoul08381Republic of Korea
| | - Elisabetta Ada Cavalcanti‐Adam
- Department of Cellular BiophysicsGrowth Factor Mechanobiology groupMax Planck Institute for Medical Research Jahnstraße 2969120HeidelbergGermany
| | - Hae‐Won Kim
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan330‐714Republic of Korea
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan330‐714Republic of Korea
- Department of Biomaterials ScienceSchool of DentistryDankook UniversityCheonan330‐714Republic of Korea
| | - Maria‐Pau Ginebra
- BiomaterialsBiomechanics and Tissue Engineering GroupDepartment of Materials Science and EngineeringUniversitat Politècnica de Catalunya (UPC)Barcelona08019Spain
- Barcelona Research Center in Multiscale Science and EngineeringUPCBarcelona08019Spain
- Institute for Bioengineering of CataloniaBarcelona08028Spain
| | - Jung‐Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan330‐714Republic of Korea
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan330‐714Republic of Korea
- Department of Biomaterials ScienceSchool of DentistryDankook UniversityCheonan330‐714Republic of Korea
| | - Carlos Mas‐Moruno
- BiomaterialsBiomechanics and Tissue Engineering GroupDepartment of Materials Science and EngineeringUniversitat Politècnica de Catalunya (UPC)Barcelona08019Spain
- Barcelona Research Center in Multiscale Science and EngineeringUPCBarcelona08019Spain
| |
Collapse
|
15
|
Prouvé E, Rémy M, Feuillie C, Molinari M, Chevallier P, Drouin B, Laroche G, Durrieu MC. Interplay of matrix stiffness and stress relaxation in directing osteogenic differentiation of mesenchymal stem cells. Biomater Sci 2022; 10:4978-4996. [PMID: 35801706 DOI: 10.1039/d2bm00485b] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The aim of this study is to investigate the impact of the stiffness and stress relaxation of poly(acrylamide-co-acrylic acid) hydrogels on the osteogenic differentiation of human mesenchymal stem cells (hMSCs). Varying the amount of the crosslinker and the ratio between the monomers enabled the obtainment of hydrogels with controlled mechanical properties, as characterized using unconfined compression and atomic force microscopy (AFM). Subsequently, the surface of the hydrogels was functionalized with a mimetic peptide of the BMP-2 protein, in order to favor the osteogenic differentiation of hMSCs. Finally, hMSCs were cultured on the hydrogels with different stiffness and stress relaxation: 15 kPa - 15%, 60 kPa - 15%, 140 kPa - 15%, 100 kPa - 30%, and 140 kPa - 70%. The cells on hydrogels with stiffnesses from 60 kPa to 140 kPa presented a star-like shape, typical of osteocytes, which has only been reported by our group for two-dimensional substrates. Then, the extent of hMSC differentiation was evaluated by using immunofluorescence and by quantifying the expression of both osteoblast markers (Runx-2 and osteopontin) and osteocyte markers (E11, DMP1, and sclerostin). It was found that a stiffness of 60 kPa led to a higher expression of osteocyte markers as compared to stiffnesses of 15 and 140 kPa. Finally, the strongest expression of osteoblast and osteocyte differentiation markers was observed for the hydrogel with a high relaxation of 70% and a stiffness of 140 kPa.
Collapse
Affiliation(s)
- Emilie Prouvé
- Laboratoire d'Ingénierie de Surface, Centre de Recherche sur les Matériaux Avancés, Département de Génie des Mines, de la Métallurgie et des Matériaux, Université Laval, 1065 Avenue de la médecine, Québec G1V 0A6, Canada. .,Axe médecine régénératrice, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Hôpital St-François d'Assise, 10 rue de l'Espinay, Québec G1L 3L5, Canada.,Université de Bordeaux, Chimie et Biologie des Membranes et Nano-Objets (UMR5248 CBMN), Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.,CNRS, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.,Bordeaux INP, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.
| | - Murielle Rémy
- Université de Bordeaux, Chimie et Biologie des Membranes et Nano-Objets (UMR5248 CBMN), Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.,CNRS, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.,Bordeaux INP, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.
| | - Cécile Feuillie
- Université de Bordeaux, Chimie et Biologie des Membranes et Nano-Objets (UMR5248 CBMN), Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.,CNRS, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.,Bordeaux INP, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.
| | - Michael Molinari
- Université de Bordeaux, Chimie et Biologie des Membranes et Nano-Objets (UMR5248 CBMN), Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.,CNRS, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.,Bordeaux INP, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.
| | - Pascale Chevallier
- Laboratoire d'Ingénierie de Surface, Centre de Recherche sur les Matériaux Avancés, Département de Génie des Mines, de la Métallurgie et des Matériaux, Université Laval, 1065 Avenue de la médecine, Québec G1V 0A6, Canada. .,Axe médecine régénératrice, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Hôpital St-François d'Assise, 10 rue de l'Espinay, Québec G1L 3L5, Canada
| | - Bernard Drouin
- Axe médecine régénératrice, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Hôpital St-François d'Assise, 10 rue de l'Espinay, Québec G1L 3L5, Canada
| | - Gaétan Laroche
- Laboratoire d'Ingénierie de Surface, Centre de Recherche sur les Matériaux Avancés, Département de Génie des Mines, de la Métallurgie et des Matériaux, Université Laval, 1065 Avenue de la médecine, Québec G1V 0A6, Canada. .,Axe médecine régénératrice, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Hôpital St-François d'Assise, 10 rue de l'Espinay, Québec G1L 3L5, Canada
| | - Marie-Christine Durrieu
- Université de Bordeaux, Chimie et Biologie des Membranes et Nano-Objets (UMR5248 CBMN), Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.,CNRS, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.,Bordeaux INP, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.
| |
Collapse
|
16
|
Fathi-Karkan S, Banimohamad-Shotorbani B, Saghati S, Rahbarghazi R, Davaran S. A critical review of fibrous polyurethane-based vascular tissue engineering scaffolds. J Biol Eng 2022; 16:6. [PMID: 35331305 PMCID: PMC8951709 DOI: 10.1186/s13036-022-00286-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/08/2022] [Indexed: 12/20/2022] Open
Abstract
Certain polymeric materials such as polyurethanes (PUs) are the most prevalent class of used biomaterials in regenerative medicine and have been widely explored as vascular substitutes in several animal models. It is thought that PU-based biomaterials possess suitable hemo-compatibility with comparable performance related to the normal blood vessels. Despite these advantages, the possibility of thrombus formation and restenosis limits their application as artificial functional vessels. In this regard, various surface modification approaches have been developed to enhance both hemo-compatibility and prolong patency. While critically reviewing the recent advances in vascular tissue engineering, mainly PU grafts, this paper summarizes the application of preferred cell sources to vascular regeneration, physicochemical properties, and some possible degradation mechanisms of PU to provide a more extensive perspective for future research.
Collapse
Affiliation(s)
- Sonia Fathi-Karkan
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Nanotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Golgasht St, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behnaz Banimohamad-Shotorbani
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Saghati
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran. .,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Soodabeh Davaran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
17
|
Zhu M, Zhong W, Cao W, Zhang Q, Wu G. Chondroinductive/chondroconductive peptides and their-functionalized biomaterials for cartilage tissue engineering. Bioact Mater 2022; 9:221-238. [PMID: 34820567 PMCID: PMC8585793 DOI: 10.1016/j.bioactmat.2021.07.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/19/2021] [Accepted: 07/05/2021] [Indexed: 02/06/2023] Open
Abstract
The repair of articular cartilage defects is still challenging in the fields of orthopedics and maxillofacial surgery due to the avascular structure of articular cartilage and the limited regenerative capacity of mature chondrocytes. To provide viable treatment options, tremendous efforts have been made to develop various chondrogenically-functionalized biomaterials for cartilage tissue engineering. Peptides that are derived from and mimic the functions of chondroconductive cartilage extracellular matrix and chondroinductive growth factors, represent a unique group of bioactive agents for chondrogenic functionalization. Since they can be chemically synthesized, peptides bear better reproducibility, more stable efficacy, higher modifiability and yielding efficiency in comparison with naturally derived biomaterials and recombinant growth factors. In this review, we summarize the current knowledge in the designs of the chondroinductive/chondroconductive peptides, the underlying molecular mechanisms and their-functionalized biomaterials for cartilage tissue engineering. We also systematically compare their in-vitro and in-vivo efficacies in inducing chondrogenesis. Our vision is to stimulate the development of novel peptides and their-functionalized biomaterials for cartilage tissue engineering.
Collapse
Affiliation(s)
- Mingjing Zhu
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam (VU), Amsterdam Movement Science (AMS), Amsterdam, the Netherlands
| | - Wenchao Zhong
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China
| | - Wei Cao
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam (VU), Amsterdam Movement Science (AMS), Amsterdam, the Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Qingbin Zhang
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China
| | - Gang Wu
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam (VU), Amsterdam Movement Science (AMS), Amsterdam, the Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| |
Collapse
|
18
|
Sun J, Huang Y, Zhao H, Niu J, Ling X, Zhu C, Wang L, Yang H, Yang Z, Pan G, Shi Q. Bio-clickable mussel-inspired peptides improve titanium-based material osseointegration synergistically with immunopolarization-regulation. Bioact Mater 2021; 9:1-14. [PMID: 34820551 PMCID: PMC8586442 DOI: 10.1016/j.bioactmat.2021.10.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/22/2021] [Accepted: 10/03/2021] [Indexed: 12/21/2022] Open
Abstract
Upon the osteoporotic condition, sluggish osteogenesis, excessive bone resorption, and chronic inflammation make the osseointegration of bioinert titanium (Ti) implants with surrounding bone tissues difficult, often lead to prosthesis loosening, bone collapse, and implant failure. In this study, we firstly designed clickable mussel-inspired peptides (DOPA-N3) and grafted them onto the surfaces of Ti materials through robust catechol-TiO2 coordinative interactions. Then, two dibenzylcyclooctyne (DBCO)-capped bioactive peptides RGD and BMP-2 bioactive domain (BMP-2) were clicked onto the DOPA-N3-coated Ti material surfaces via bio-orthogonal reaction. We characterized the surface morphology and biocompatibility of the Ti substrates and optimized the osteogenic capacity of Ti surfaces through adjusting the ideal ratios of BMP-2/RGD at 3:1. In vitro, the dual-functionalized Ti substrates exhibited excellent promotion on adhesion and osteogenesis of mesenchymal stem cells (MSCs), and conspicuous immunopolarization-regulation to shift macrophages to alternative (M2) phenotypes and inhibit inflammation, as well as enhancement of osseointegration and mechanical stability in osteoporotic rats. In summary, our biomimetic surface modification strategy by bio-orthogonal reaction provided a convenient and feasible method to resolve the bioinertia and clinical complications of Ti-based implants, which was conducive to the long-term success of Ti implants, especially in the osteoporotic or inflammatory conditions. A clickable mussel-inspired peptide and two DBCO-capped bioactive peptides for facile decoration of Ti prostheses via robust catechol/TiO2 coordinative interactions and click chemical reaction. Dual functionalized Ti-based surface can improve cell anchoring and osteogenicitity by rationally adjusting the grafting ratio of BMP-2 and RGD peptides. Dual functionalized Ti-based surface synergistically achieve M2 shifting and efficient inflammation inhibition for osseointegration.
Collapse
Affiliation(s)
- Jie Sun
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopaedic Institute of Soochow University, 899 Pinghai, Suzhou, Jiangsu, 215031, China
| | - Yingkang Huang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopaedic Institute of Soochow University, 899 Pinghai, Suzhou, Jiangsu, 215031, China
| | - Huan Zhao
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopaedic Institute of Soochow University, 899 Pinghai, Suzhou, Jiangsu, 215031, China
| | - Junjie Niu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopaedic Institute of Soochow University, 899 Pinghai, Suzhou, Jiangsu, 215031, China
| | - Xuwei Ling
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopaedic Institute of Soochow University, 899 Pinghai, Suzhou, Jiangsu, 215031, China
| | - Can Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopaedic Institute of Soochow University, 899 Pinghai, Suzhou, Jiangsu, 215031, China
| | - Lin Wang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopaedic Institute of Soochow University, 899 Pinghai, Suzhou, Jiangsu, 215031, China
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopaedic Institute of Soochow University, 899 Pinghai, Suzhou, Jiangsu, 215031, China
| | - Zhilu Yang
- Affiliated Dongguan Hospital, Southern Medical University, No. 3 Wandao Road, Dongguan, Guangdong, 523059, China.,Guangdong Provincial Key Laboratory of Shock and Microcirculation, No. 1023 Shatai Road, Guangzhou, Guangdong, 510080, China
| | - Guoqing Pan
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Qin Shi
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopaedic Institute of Soochow University, 899 Pinghai, Suzhou, Jiangsu, 215031, China
| |
Collapse
|
19
|
Krieghoff J, Gronbach M, Schulz-Siegmund M, Hacker MC. Biodegradable macromers for implant bulk and surface engineering. Biol Chem 2021; 402:1357-1374. [PMID: 34433237 DOI: 10.1515/hsz-2021-0161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 08/09/2021] [Indexed: 11/15/2022]
Abstract
Macromers, polymeric molecules with at least two functional groups for cross-polymerization, are interesting materials to tailor mechanical, biochemical and degradative bulk and surface properties of implants for tissue regeneration. In this review we focus on macromers with at least one biodegradable building block. Manifold design options, such as choice of polymeric block(s), optional core molecule and reactive groups, as well as cross-co-polymerization with suitable anchor or linker molecules, allow the adaptation of macromer-based biomaterials towards specific application requirements in both hard and soft tissue regeneration. Implants can be manufactured from macromers using additive manufacturing as well as molding and templating approaches. This review summarizes and discusses the overall concept of biodegradable macromers and recent approaches for macromer processing into implants as well as techniques for surface modification directed towards bone regeneration. These aspects are reviewed including a focus on the authors' contributions to the field through research within the collaborative research project Transregio 67.
Collapse
Affiliation(s)
- Jan Krieghoff
- Medical Faculty, Pharmaceutical Technology, Leipzig University, Eilenburger Str. 15A, D-04317 Leipzig, Germany.,Collaborative Research Center (SFB-TRR67) "Functional Biomaterials for Controlling Healing Processes in Bone and Skin - From Material Science to Clinical Application", Leipzig and Dresden, Germany
| | - Mathis Gronbach
- Medical Faculty, Pharmaceutical Technology, Leipzig University, Eilenburger Str. 15A, D-04317 Leipzig, Germany.,Collaborative Research Center (SFB-TRR67) "Functional Biomaterials for Controlling Healing Processes in Bone and Skin - From Material Science to Clinical Application", Leipzig and Dresden, Germany
| | - Michaela Schulz-Siegmund
- Medical Faculty, Pharmaceutical Technology, Leipzig University, Eilenburger Str. 15A, D-04317 Leipzig, Germany.,Collaborative Research Center (SFB-TRR67) "Functional Biomaterials for Controlling Healing Processes in Bone and Skin - From Material Science to Clinical Application", Leipzig and Dresden, Germany
| | - Michael C Hacker
- Medical Faculty, Pharmaceutical Technology, Leipzig University, Eilenburger Str. 15A, D-04317 Leipzig, Germany.,Collaborative Research Center (SFB-TRR67) "Functional Biomaterials for Controlling Healing Processes in Bone and Skin - From Material Science to Clinical Application", Leipzig and Dresden, Germany.,Institute of Pharmaceutics and Biopharmaceutics, Heinrich Heine University, Universitätsstrasse 1, D-40225 Düsseldorf, Germany
| |
Collapse
|
20
|
Oliver-Cervelló L, Martin-Gómez H, Mas-Moruno C. New trends in the development of multifunctional peptides to functionalize biomaterials. J Pept Sci 2021; 28:e3335. [PMID: 34031952 DOI: 10.1002/psc.3335] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 12/16/2022]
Abstract
Improving cell-material interactions is a major goal in tissue engineering. In this regard, functionalization of biomaterials with cell instructive molecules from the extracellular matrix stands out as a powerful strategy to enhance their bioactivity and achieve optimal tissue integration. However, current functionalization strategies, like the use of native full-length proteins, are associated with drawbacks, thus urging the need of developing new methodologies. In this regard, the use of synthetic peptides encompassing specific bioactive regions of proteins represents a promising alternative. In particular, the combination of peptide sequences with complementary or synergistic effects makes it possible to address more than one biological target at the biomaterial surface. In this review, an overview of the main strategies using peptides to install multifunctionality on biomaterials is presented, mostly focusing on the combination of the RGD motif with other peptides sequences. The evolution of these approaches, starting from simple methods, like using peptide mixtures, to more advanced systems of peptide presentation, with very well defined chemical properties, are explained. For each system of peptide's presentation, three main aspects of multifunctionality-improving receptor selectivity, mimicking the extracellular matrix and preventing bacterial colonization while improving cell adhesion-are highlighted.
Collapse
Affiliation(s)
- Lluís Oliver-Cervelló
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain.,Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, Spain
| | - Helena Martin-Gómez
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain.,Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, Spain
| | - Carlos Mas-Moruno
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain.,Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, Spain
| |
Collapse
|
21
|
Seims KB, Hunt NK, Chow LW. Strategies to Control or Mimic Growth Factor Activity for Bone, Cartilage, and Osteochondral Tissue Engineering. Bioconjug Chem 2021; 32:861-878. [PMID: 33856777 DOI: 10.1021/acs.bioconjchem.1c00090] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Growth factors play a critical role in tissue repair and regeneration. However, their clinical success is limited by their low stability, short half-life, and rapid diffusion from the delivery site. Supraphysiological growth factor concentrations are often required to demonstrate efficacy but can lead to adverse reactions, such as inflammatory complications and increased cancer risk. These issues have motivated the development of delivery systems that enable sustained release and controlled presentation of growth factors. This review specifically focuses on bioconjugation strategies to enhance growth factor activity for bone, cartilage, and osteochondral applications. We describe approaches to localize growth factors using noncovalent and covalent methods, bind growth factors via peptides, and mimic growth factor function with mimetic peptide sequences. We also discuss emerging and future directions to control spatiotemporal growth factor delivery to improve functional tissue repair and regeneration.
Collapse
Affiliation(s)
- Kelly B Seims
- Department of Materials Science and Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Natasha K Hunt
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Lesley W Chow
- Department of Materials Science and Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| |
Collapse
|
22
|
Oliver‐Cervelló L, Martin‐Gómez H, Reyes L, Noureddine F, Ada Cavalcanti‐Adam E, Ginebra M, Mas‐Moruno C. An Engineered Biomimetic Peptide Regulates Cell Behavior by Synergistic Integrin and Growth Factor Signaling. Adv Healthc Mater 2021; 10:e2001757. [PMID: 33336559 DOI: 10.1002/adhm.202001757] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/02/2020] [Indexed: 01/04/2023]
Abstract
Recreating the healing microenvironment is essential to regulate cell-material interactions and ensure the integration of biomaterials. To repair bone, such bioactivity can be achieved by mimicking its extracellular matrix (ECM) and by stimulating integrin and growth factor (GF) signaling. However, current approaches relying on the use of GFs, such as bone morphogenetic protein 2 (BMP-2), entail clinical risks. Here, a biomimetic peptide integrating the RGD cell adhesive sequence and the osteogenic DWIVA motif derived from the wrist epitope of BMP-2 is presented. The approach offers the advantage of having a spatial control over the single binding of integrins and BMP receptors. Such multifunctional platform is designed to incorporate 3,4-dihydroxyphenylalanine to bind metallic oxides with high affinity in a one step process. Functionalization of glass substrates with the engineered peptide is characterized by physicochemical methods, proving a successful surface modification. The biomimetic interfaces significantly improve the adhesion of C2C12 cells, inhibit myotube formation, and activate the BMP-dependent signaling via p38. These effects are not observed on surfaces displaying only one bioactive motif, a mixture of both motifs or soluble DWIVA. These data prove the biological potential of recreating the ECM and engaging in integrin and GF crosstalk via molecular-based mimics.
Collapse
Affiliation(s)
- Lluís Oliver‐Cervelló
- Biomaterials, Biomechanics and Tissue Engineering Group Department of Materials Science and Engineering Universitat Politècnica de Catalunya (UPC) Barcelona 08019 Spain
- Barcelona Research Center in Multiscale Science and Engineering UPC Barcelona 08019 Spain
| | - Helena Martin‐Gómez
- Biomaterials, Biomechanics and Tissue Engineering Group Department of Materials Science and Engineering Universitat Politècnica de Catalunya (UPC) Barcelona 08019 Spain
- Barcelona Research Center in Multiscale Science and Engineering UPC Barcelona 08019 Spain
| | - Leslie Reyes
- Biomaterials, Biomechanics and Tissue Engineering Group Department of Materials Science and Engineering Universitat Politècnica de Catalunya (UPC) Barcelona 08019 Spain
| | - Fatima Noureddine
- Department of Cellular Biophysics Max Planck Institute for Medical Research Jahnstraße 29 Heidelberg 69120 Germany
| | | | - Maria‐Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group Department of Materials Science and Engineering Universitat Politècnica de Catalunya (UPC) Barcelona 08019 Spain
- Barcelona Research Center in Multiscale Science and Engineering UPC Barcelona 08019 Spain
- Institute for Bioengineering of Catalonia Barcelona 08028 Spain
| | - Carlos Mas‐Moruno
- Biomaterials, Biomechanics and Tissue Engineering Group Department of Materials Science and Engineering Universitat Politècnica de Catalunya (UPC) Barcelona 08019 Spain
- Barcelona Research Center in Multiscale Science and Engineering UPC Barcelona 08019 Spain
| |
Collapse
|
23
|
Bullock G, Atkinson J, Gentile P, Hatton P, Miller C. Osteogenic Peptides and Attachment Methods Determine Tissue Regeneration in Modified Bone Graft Substitutes. J Funct Biomater 2021; 12:22. [PMID: 33807267 PMCID: PMC8103284 DOI: 10.3390/jfb12020022] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 01/01/2023] Open
Abstract
The inclusion of biofunctional molecules with synthetic bone graft substitutes has the potential to enhance tissue regeneration during treatment of traumatic bone injuries. The clinical use of growth factors has though been associated with complications, some serious. The use of smaller, active peptides has the potential to overcome these problems and provide a cost-effective, safe route for the manufacture of enhanced bone graft substitutes. This review considers the design of peptide-enhanced bone graft substitutes, and how peptide selection and attachment method determine clinical efficacy. It was determined that covalent attachment may reduce the known risks associated with growth factor-loaded bone graft substitutes, providing a predictable tissue response and greater clinical efficacy. Peptide choice was found to be critical, but even within recognised families of biologically active peptides, the configurations that appeared to most closely mimic the biological molecules involved in natural bone healing processes were most potent. It was concluded that rational, evidence-based design of peptide-enhanced bone graft substitutes offers a pathway to clinical maturity in this highly promising field.
Collapse
Affiliation(s)
- George Bullock
- School of Clinical Dentistry, The University of Sheffield, Sheffield S10 2TA, UK; (G.B.); (J.A.); (C.M.)
| | - Joss Atkinson
- School of Clinical Dentistry, The University of Sheffield, Sheffield S10 2TA, UK; (G.B.); (J.A.); (C.M.)
| | - Piergiorgio Gentile
- School of Engineering, Newcastle University, Stephenson Building, Newcastle upon Tyne NE1 7RU, UK;
| | - Paul Hatton
- School of Clinical Dentistry, The University of Sheffield, Sheffield S10 2TA, UK; (G.B.); (J.A.); (C.M.)
| | - Cheryl Miller
- School of Clinical Dentistry, The University of Sheffield, Sheffield S10 2TA, UK; (G.B.); (J.A.); (C.M.)
| |
Collapse
|
24
|
Preliminary evaluation of BMP-2-derived peptide in repairing a peri-implant critical size defect: A canine model. J Formos Med Assoc 2020; 120:1212-1220. [PMID: 33358040 DOI: 10.1016/j.jfma.2020.07.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/10/2020] [Accepted: 07/17/2020] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND/PURPOSE A synthetic bone morphogenetic protein (BMP)-2-derived peptide has been discovered to promote bone regeneration. The present study investigated the potential of the BMP-2 peptide combined with hydroxyapatite (HAp)/β-tricalcium phosphate (TCP)/collagen (Col) composite in repairing a peri-implant critical size defect. METHODS Twenty-four saddle-type alveolar defects (10 mm mesiodistally and 4 mm apicocoronally) were surgically prepared in edentulous ridges in four male beagle dogs. Following implant placement, the defects with vertically exposed implant fixtures received (a) HAp/TCP/Col composite, (b) HAp/TCP/Col + 4 mg/mL BMP-2 peptide, (c) HAp/TCP/Col + 20 mg/mL BMP-2 peptide, or (d) HAp/TCP/Col + 0.2 mg/mL recombinant human BMP-2 (rhBMP-2). Bone regeneration and mineralization were assessed using radiography, micro-computed tomography (micro-CT), fluorescence labeling, and histologic analyses after healing for 4 or 8 weeks. Implant stability was measured using resonance frequency analysis. RESULTS The 20 mg/mL BMP-2 peptide groups demonstrated a distinguishable advantage in bone regeneration potential over the control groups, as observed on radiographic imaging and histologic examination, although no significant difference was found in implant stability and histomorphometric analysis of mineralization levels. However, the performance of the 20 mg/mL BMP-2 peptide groups were inferior to that of the 0.2 mg/mL rhBMP-2 groups. CONCLUSION The BMP-2 peptide may accelerate peri-implant bone regeneration. The BMP-2 peptide at 20 mg/mL still cannot complete bone repair of peri-implant critical size defect. The BMP-2 peptide at 20 mg/mL has similar osteoinductive performance to the rhBMP-2 at 0.02 mg/mL.
Collapse
|
25
|
Chen G, Kong P, Jiang A, Wang X, Sun Y, Yu T, Chi H, Song C, Zhang H, Subedi D, Ravi Kumar P, Bai K, Liu K, Ji Y, Yan J. A modular programmed biphasic dual-delivery system on 3D ceramic scaffolds for osteogenesis in vitro and in vivo. J Mater Chem B 2020; 8:9697-9717. [PMID: 32789334 DOI: 10.1039/c9tb02127b] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Single-factor delivery is the most common characteristic of bone tissue engineering techniques. However, bone regeneration is a complex process requiring multiple factors and specialized release mechanisms. Therefore, the development of a dual-delivery system allowing for programmed release kinetics would be highly desirable. Improvement of the molarity and versatility of the delivery system has rarely been studied. Herein, we report the development of a novel, modular programmed biphasic dual-release system (SCB), carrying a BMP2 and an engineered collagen I-derived recognition motif (Stath-DGEA), with a self-remodification feature on hydroxyapatite (HA)-based materials. The SCB system was loaded onto an additive manufactured (AM) scaffold in order to evaluate its bifactor osteogenic potential and its biphasic release behavior. Further, the biomechanical properties of the scaffold were studied by using the fluid-structure interaction (FSI) method. Section fluorescent labeling revealed that the HA scaffold has a relatively higher density and efficiency. Additionally, the results of the release and inhibition experiment suggested that the SCB system could facilitate the sustained release of therapeutic levels of two factors during the initial stage of implantation, thereby exhibiting a rapid high-dose release pattern at a specific time point during the second stage. The FSI prediction model indicated that the scaffold provides an excellent biomimetic mechanical and fluid dynamic microenvironment to promote osteogenesis. Our results indicated that incorporation of BMP2 with Stath-DGEA in the biphasic SCB system could have a synergetic effect in promoting the adhesion, proliferation, and differentiation of bone marrow mesenchymal stem cells (BMSCs) in vitro, under staged stimulations. Further, in vivo studies in both ectopic and orthotopic rat models showed that the SCB system loaded onto an AM scaffold could enhance osteointegration and osteoinduction throughout the osteogenic process. Thus, the novel synthetic SCB system described herein used on an AM scaffold provides a biomimetic extracellular environment that enhances bone regeneration and is a promising multifunctional, dual-release platform.
Collapse
Affiliation(s)
- Guanghua Chen
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Jann J, Drevelle O, Lauzon MA, Faucheux N. Adhesion, intracellular signalling and osteogenic differentiation of mesenchymal progenitor cells and preosteoblasts on poly(epsilon)caprolactone films functionalized by peptides derived from fibronectin and/or BMP-9. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 114:111088. [DOI: 10.1016/j.msec.2020.111088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 04/14/2020] [Accepted: 05/08/2020] [Indexed: 12/17/2022]
|
27
|
Fischer NG, He J, Aparicio C. Surface Immobilization Chemistry of a Laminin-Derived Peptide Affects Keratinocyte Activity. COATINGS (BASEL, SWITZERLAND) 2020; 10:560. [PMID: 32855816 PMCID: PMC7448695 DOI: 10.3390/coatings10060560] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Many chemical routes have been proposed to immobilize peptides on biomedical device surfaces, and in particular, on dental implants to prevent peri-implantitis. While a number of factors affect peptide immobilization quality, an easily controllable factor is the chemistry used to immobilize peptides. These factors affect peptide chemoselectivity, orientation, etc., and ultimately control biological activity. Using many different physical and chemical routes for peptide coatings, previous research has intensely focused on immobilizing antimicrobial elements on dental implants to reduce infection rates. Alternatively, our strategy here is different and focused on promoting formation of a long-lasting biological seal between the soft tissue and the implant surface through transmembrane, cell adhesion structures called hemidesmosomes. For that purpose, we used a laminin-derived call adhesion peptide. However, the effect of different immobilization chemistries on cell adhesion peptide activity is vastly unexplored but likely critical. Here, we compared the physiochemical properties and biological responses of a hemidesmosome promoting peptide immobilized using silanization and copper-free click chemistry as a model system for cell adhesion peptides. Successful immobilization was confirmed with water contact angle and X-ray photoelectron spectroscopy. Peptide coatings were retained through 73 days of incubation in artificial saliva. Interestingly, the non-chemoselective immobilization route, silanization, resulted in significantly higher proliferation and hemidesmosome formation in oral keratinocytes compared to chemoselective click chemistry. Our results highlight that the most effective immobilization chemistry for optimal peptide activity is dependent on the specific system (substrate/peptide/cell/biological activity) under study. Overall, a better understanding of the effects immobilization chemistries have on cell adhesion peptide activity may lead to more efficacious coatings for biomedical devices.
Collapse
Affiliation(s)
- Nicholas G. Fischer
- Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 515 Delaware Street S.E., Minneapolis, MN 55455, USA
| | - Jiahe He
- Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 515 Delaware Street S.E., Minneapolis, MN 55455, USA
| | - Conrado Aparicio
- Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 515 Delaware Street S.E., Minneapolis, MN 55455, USA
| |
Collapse
|
28
|
Mertgen AS, Trossmann VT, Guex AG, Maniura-Weber K, Scheibel T, Rottmar M. Multifunctional Biomaterials: Combining Material Modification Strategies for Engineering of Cell-Contacting Surfaces. ACS APPLIED MATERIALS & INTERFACES 2020; 12:21342-21367. [PMID: 32286789 DOI: 10.1021/acsami.0c01893] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
In the human body, cells in a tissue are exposed to signals derived from their specific extracellular matrix (ECM), such as architectural structure, mechanical properties, and chemical composition (proteins, growth factors). Research on biomaterials in tissue engineering and regenerative medicine aims to recreate such stimuli using engineered materials to induce a specific response of cells at the interface. Although traditional biomaterials design has been mostly limited to varying individual signals, increasing interest has arisen on combining several features in recent years to improve the mimicry of extracellular matrix properties. Tremendous progress in combinatorial surface modification exploiting, for example, topographical features or variations in mechanics combined with biochemical cues has enabled the identification of their key regulatory characteristics on various cell fate decisions. Gradients especially facilitated such research by enabling the investigation of combined continuous changes of different signals. Despite unravelling important synergies for cellular responses, challenges arise in terms of fabrication and characterization of multifunctional engineered materials. This review summarizes recent work on combinatorial surface modifications that aim to control biological responses. Modification and characterization methods for enhanced control over multifunctional material properties are highlighted and discussed. Thereby, this review deepens the understanding and knowledge of biomimetic combinatorial material modification, their challenges but especially their potential.
Collapse
Affiliation(s)
- Anne-Sophie Mertgen
- Laboratory for Biointerfaces, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, St. Gallen 9014, Switzerland
- Laboratory for Biomimetic Membranes and Textiles, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, St. Gallen 9014, Switzerland
| | - Vanessa Tanja Trossmann
- Lehrstuhl für Biomaterialien, Universität Bayreuth, Prof.-Rüdiger-Bormann-Strasse 1, Bayreuth 95440, Germany
| | - Anne Géraldine Guex
- Laboratory for Biointerfaces, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, St. Gallen 9014, Switzerland
- Laboratory for Biomimetic Membranes and Textiles, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, St. Gallen 9014, Switzerland
| | - Katharina Maniura-Weber
- Laboratory for Biointerfaces, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, St. Gallen 9014, Switzerland
| | - Thomas Scheibel
- Lehrstuhl für Biomaterialien, Bayerisches Polymerinstitut (BPI), Bayreuther Zentrum für Kolloide und Grenzflächen (BZKG), Bayreuther Zentrum für Molekulare Biowissenschaften (BZMB), Bayreuther Materialzentrum (BayMAT), Universität Bayreuth, Bayreuth 95440, Germany
| | - Markus Rottmar
- Laboratory for Biointerfaces, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, St. Gallen 9014, Switzerland
| |
Collapse
|
29
|
Posa F, Grab AL, Martin V, Hose D, Seckinger A, Mori G, Vukicevic S, Cavalcanti-Adam EA. Copresentation of BMP-6 and RGD Ligands Enhances Cell Adhesion and BMP-Mediated Signaling. Cells 2019; 8:E1646. [PMID: 31847477 PMCID: PMC6953040 DOI: 10.3390/cells8121646] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/09/2019] [Accepted: 12/13/2019] [Indexed: 12/16/2022] Open
Abstract
We report on the covalent immobilization of bone morphogenetic protein 6 (BMP-6) and its co-presentation with integrin ligands on a nanopatterned platform to study cell adhesion and signaling responses which regulate the transdifferentiation of myoblasts into osteogenic cells. To immobilize BMP-6, the heterobifunctional linker MU-NHS is coupled to amine residues of the growth factor; this prevents its internalization while ensuring that its biological activity is maintained. Additionally, to allow cells to adhere to such platform and study signaling events arising from the contact to the surface, we used click-chemistry to immobilize cyclic-RGD carrying an azido group reacting with PEG-alkyne spacers via copper-catalyzed 1,3-dipolar cycloaddition. We show that the copresentation of BMP-6 and RGD favors focal adhesion formation and promotes Smad 1/5/8 phosphorylation. When presented in low amounts, BMP-6 added to culture media of cells adhering to the RGD ligands is less effective than BMP-6 immobilized on the surfaces in inducing Smad complex activation and in inhibiting myotube formation. Our results suggest that a local control of ligand density and cell signaling is crucial for modulating cell response.
Collapse
Affiliation(s)
- Francesca Posa
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstr. 29, 69120 Heidelberg, Germany
- Department of Biophysical Chemistry, Institute of Physical Chemistry, Heidelberg University, Im Neuenheimer Feld 253, 69120 Heidelberg, Germany
- Department of Clinical and Experimental Medicine, University of Foggia, via L. Pinto, 71122 Foggia, Italy
| | - Anna Luise Grab
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstr. 29, 69120 Heidelberg, Germany
- Department of Biophysical Chemistry, Institute of Physical Chemistry, Heidelberg University, Im Neuenheimer Feld 253, 69120 Heidelberg, Germany
- Genome Biology Unit, EMBL, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Volker Martin
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstr. 29, 69120 Heidelberg, Germany
- Department of Biophysical Chemistry, Institute of Physical Chemistry, Heidelberg University, Im Neuenheimer Feld 253, 69120 Heidelberg, Germany
| | - Dirk Hose
- Laboratory for Myeloma Research and Medical Clinic V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Anja Seckinger
- Laboratory for Myeloma Research and Medical Clinic V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Giorgio Mori
- Department of Clinical and Experimental Medicine, University of Foggia, via L. Pinto, 71122 Foggia, Italy
| | - Slobodan Vukicevic
- Laboratory for Mineralized Tissues, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, Šalata 11, 10000 Zagreb, Croatia
| | - Elisabetta Ada Cavalcanti-Adam
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstr. 29, 69120 Heidelberg, Germany
- Department of Biophysical Chemistry, Institute of Physical Chemistry, Heidelberg University, Im Neuenheimer Feld 253, 69120 Heidelberg, Germany
| |
Collapse
|
30
|
Silantyeva EA, Willits RK, Becker ML. Postfabrication Tethering of Molecular Gradients on Aligned Nanofibers of Functional Poly(ε-caprolactone)s. Biomacromolecules 2019; 20:4494-4501. [PMID: 31721566 PMCID: PMC7546418 DOI: 10.1021/acs.biomac.9b01264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Substrates with combinations of topographical and biochemical cues are highly useful for a number of fundamental biological investigations. Tethered molecular concentration gradients in particular are highly desired for a number of biomedical applications including cell migration. Herein, we report a versatile method for the fabrication of aligned nanofiber substrates with a tunable concentration gradient along the fiber direction. 4-Dibenzocyclooctynol (DIBO) was used as an initiator for the ring-opening copolymerization of ε-caprolactone (εCL) and allyl-functionalized ε-caprolactone (AεPCL), which yielded a well-defined polymer with orthogonal functional handles. These materials were fabricated into aligned nanofiber substrates via touch-spinning. Fibers were modified post-spinning with a concentration gradient of fluorescently labeled dye via a light activated thiol-ene reaction through a photomask. As a demonstration, the cell adhesive peptide RGD was chemically tethered to the fiber surface at a second functionalization site via strain-promoted azide-alkyne cycloaddition (SPAAC). This novel approach affords fabrication of dual functional nanofiber substrates.
Collapse
Affiliation(s)
- Elena A. Silantyeva
- Department of Polymer Science, The University of Akron, Akron, OH 44325, USA
| | - Rebecca K. Willits
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| | - Matthew L. Becker
- Department of Polymer Science, The University of Akron, Akron, OH 44325, USA
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
- Department of Chemistry, Duke University, Durham, NC 27708, USA
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| |
Collapse
|
31
|
Motta CMM, Endres KJ, Wesdemiotis C, Willits RK, Becker ML. Enhancing Schwann cell migration using concentration gradients of laminin-derived peptides. Biomaterials 2019; 218:119335. [PMID: 31302351 PMCID: PMC6868524 DOI: 10.1016/j.biomaterials.2019.119335] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 06/29/2019] [Accepted: 07/03/2019] [Indexed: 12/17/2022]
Abstract
Neuroregeneration following peripheral nerve injury is largely mediated by Schwann cells (SC), the principal glial cell that supports neurons in the peripheral nervous system. Axonal regeneration in vivo is limited by the extent of SC migration into the gap between the proximal and distal nerve, however, little is known regarding the principal driving forces for SC migration. Engineered microenvironments, such as molecular and protein gradients, play a role in the migration of many cell types, including cancer cells and fibroblasts. However, haptotactic strategies have not been applied widely to SC. Herein, a series of tethered laminin-derived peptides were analyzed for their influence on SC adhesion, proliferation, and alignment. Concentration gradient substrates were fabricated using a controlled vapor deposition method, followed by covalent peptide attachment via a thiol-ene reaction, and characterized by X-ray photoelectron spectroscopy (XPS) and MALDI-MS imaging. While tethered RGD peptides supported SC adhesion and proliferation, concentration gradients of RGD had little influence on biased SC directional migration. In contrast, YIGSR promoted less SC attachment than RGD, yet YIGSR peptide gradients directed migration with a strong bias to the concentration profile. With YIGSR peptide, overall speed increased with the steepness of the peptide concentration profile. YIGSR gradients had no haptotactic effect on rat dermal fibroblast migration, in contrast to fibroblast migration on RGD gradients. The response of SC to these tethered peptide gradients will guide the development of translationally relevant constructs designed to facilitate endogenous SC infiltration into defects for nerve regeneration.
Collapse
Affiliation(s)
- Cecilia M M Motta
- Department of Polymer Science, The University of Akron, Akron, OH, 44325, United States
| | - Kevin J Endres
- Department of Chemistry, The University of Akron, Akron, OH, 44325, United States
| | - Chrys Wesdemiotis
- Department of Chemistry, The University of Akron, Akron, OH, 44325, United States
| | - Rebecca K Willits
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, United States.
| | - Matthew L Becker
- Department of Polymer Science, The University of Akron, Akron, OH, 44325, United States; Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, United States; Department of Chemistry, Mechanical Engineering and Materials Science, and Orthopaedic Surgery, Duke University, Durham, NC, 27708, United States.
| |
Collapse
|
32
|
Rasi Ghaemi S, Delalat B, Cavallaro A, Mierczynska‐Vasilev A, Vasilev K, Voelcker NH. Differentiation of Rat Mesenchymal Stem Cells toward Osteogenic Lineage on Extracellular Matrix Protein Gradients. Adv Healthc Mater 2019; 8:e1900595. [PMID: 31328896 DOI: 10.1002/adhm.201900595] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/08/2019] [Indexed: 12/15/2022]
Abstract
This report addresses the issue of optimizing extracellular matrix protein density required to support osteogenic lineage differentiation of mesenchymal stem cells (MSCs) by culturing MSCs on surface-bound density gradients of immobilized collagen type I (COL1) and osteopontin (OPN). A chemical surface gradient is prepared by tailoring the surface chemical composition from high hydroxyl groups to aldehyde groups using a diffusion-controlled plasma polymerization technique. Osteogenesis on the gradient surface is determined by immunofluorescence staining against Runx2 as an early marker and by staining of calcium phosphate deposits as a late stage differentiation marker. The Runx2 intensity and calcified area increase with increasing COL1 density up to a critical value corresponding to 124.2 ng cm-2 , above which cell attachment and differentiation do not rise further, while this critical value for OPN is 19.0 ng cm-2 . This gradient approach may facilitate the screening of an optimal biomolecule surface density on tissue-engineered scaffolds, implants, or tissue culture ware to obtain the desired cell response, and may generate opportunities for more cost-effective regenerative medicine.
Collapse
Affiliation(s)
- Soraya Rasi Ghaemi
- Future Industries InstituteUniversity of South Australia Mawson Lakes Adelaide 5095 South Australia Australia
| | - Bahman Delalat
- Future Industries InstituteUniversity of South Australia Mawson Lakes Adelaide 5095 South Australia Australia
- ManufacturingCommonwealth Scientific and Industrial Research Organization (CSIRO) Clayton Melbourne 3168 Victoria Australia
- Drug DeliveryDisposition and DynamicsMonash Institute of Pharmaceutical SciencesMonash University Parkville Melbourne 3052 Victoria Australia
- Department of Mechanical and Aerospace EngineeringMonash University Clayton Melbourne 3168 Victoria Australia
| | - Alex Cavallaro
- Future Industries InstituteUniversity of South Australia Mawson Lakes Adelaide 5095 South Australia Australia
| | - Agnieszka Mierczynska‐Vasilev
- Future Industries InstituteUniversity of South Australia Mawson Lakes Adelaide 5095 South Australia Australia
- School of EngineeringUniversity of South Australia Mawson Lakes Adelaide 5095 South Australia Australia
- The Australian Wine Research InstituteWaite Precinct Hartley Grove cnr Paratoo Road, Urrbrae Adelaide 5064 South Australia Australia
| | - Krasimir Vasilev
- Future Industries InstituteUniversity of South Australia Mawson Lakes Adelaide 5095 South Australia Australia
- School of EngineeringUniversity of South Australia Mawson Lakes Adelaide 5095 South Australia Australia
| | - Nicolas H. Voelcker
- Future Industries InstituteUniversity of South Australia Mawson Lakes Adelaide 5095 South Australia Australia
- ManufacturingCommonwealth Scientific and Industrial Research Organization (CSIRO) Clayton Melbourne 3168 Victoria Australia
- Drug DeliveryDisposition and DynamicsMonash Institute of Pharmaceutical SciencesMonash University Parkville Melbourne 3052 Victoria Australia
- Victorian Node of the Australian National Fabrication FacilityMelbourne Center for Nanofabrication Clayton Melbourne 3168 Victoria Australia
| |
Collapse
|
33
|
Sheyn D, Cohn-Yakubovich D, Ben-David S, De Mel S, Chan V, Hinojosa C, Wen N, Hamilton GA, Gazit D, Gazit Z. Bone-chip system to monitor osteogenic differentiation using optical imaging. MICROFLUIDICS AND NANOFLUIDICS 2019; 23:99. [PMID: 32296299 PMCID: PMC7158882 DOI: 10.1007/s10404-019-2261-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Human organoids and organ-on-chip systems to predict human responses to new therapies and for the understanding of disease mechanisms are being more commonly used in translational research. We have developed a bone-chip system to study osteogenic differentiation in vitro, coupled with optical imaging approach which provides the opportunity of monitoring cell survival, proliferation and differentiation in vitro without the need to terminate the culture. We used the mesenchymal stem cell (MSC) line over-expressing bone morphogenetic protein-2 (BMP-2), under Tet-Off system, and luciferase reporter gene under constitutive promoter. Cells were seeded on chips and supplemented with osteogenic medium. Flow of media was started 24 h later, while static cultures were performed using media reservoirs. Cells grown on the bone-chips under constant flow of media showed enhanced survival/proliferation, comparing to the cells grown in static conditions; luciferase reporter gene expression and activity, reflecting the cell survival and proliferation, was quantified using bioluminescence imaging and a significant advantage to the flow system was observed. In addition, the flow had positive effect on osteogenic differentiation, when compared with static cultures. Quantitative fluorescent imaging, performed using the osteogenic extra-cellular matrix-targeted probes, showed higher osteogenic differentiation of the cells under the flow conditions. Gene expression analysis of osteogenic markers confirmed the osteogenic differentiation of the MSC-BMP2 cells. Immunofluorescent staining performed against the Osteocalcin, Col1, and BSP markers illustrated robust osteogenic differentiation in the flow culture and lessened differentiation in the static culture. To sum, the bone-chip allows monitoring cell survival, proliferation, and osteogenic differentiation using optical imaging.
Collapse
Affiliation(s)
- Dmitriy Sheyn
- Orthopedic Stem Cell Research Lab, Cedars-Sinai Medical Center, AHSP-A8308, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Department of Orthopaedics, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
| | - Doron Cohn-Yakubovich
- Skeletal Biotech Laboratory, Hebrew University of Jerusalem, 91120 Jerusalem, Israel
| | - Shiran Ben-David
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Skeletal Regeneration Program, Cedars-Sinai Medical Center, AHSP-8304, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | - Sandra De Mel
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Skeletal Regeneration Program, Cedars-Sinai Medical Center, AHSP-8304, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | - Virginia Chan
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Skeletal Regeneration Program, Cedars-Sinai Medical Center, AHSP-8304, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | | | | | | | - Dan Gazit
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Department of Orthopaedics, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Skeletal Biotech Laboratory, Hebrew University of Jerusalem, 91120 Jerusalem, Israel
- Skeletal Regeneration Program, Cedars-Sinai Medical Center, AHSP-8304, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | - Zulma Gazit
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Department of Orthopaedics, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Skeletal Biotech Laboratory, Hebrew University of Jerusalem, 91120 Jerusalem, Israel
- Skeletal Regeneration Program, Cedars-Sinai Medical Center, AHSP-8304, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| |
Collapse
|
34
|
Tan GZ, Zhou Y. Electrospinning of biomimetic fibrous scaffolds for tissue engineering: a review. INT J POLYM MATER PO 2019. [DOI: 10.1080/00914037.2019.1636248] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- George Z. Tan
- Department of Industrial, Manufacturing and Systems Engineering, Texas Tech University, Lubbock, TX, USA
| | - Yingge Zhou
- Department of Industrial, Manufacturing and Systems Engineering, Texas Tech University, Lubbock, TX, USA
| |
Collapse
|
35
|
Yassin MA, Fuoco T, Mohamed-Ahmed S, Mustafa K, Finne-Wistrand A. 3D and Porous RGDC-Functionalized Polyester-Based Scaffolds as a Niche to Induce Osteogenic Differentiation of Human Bone Marrow Stem Cells. Macromol Biosci 2019; 19:e1900049. [PMID: 31050389 DOI: 10.1002/mabi.201900049] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/18/2019] [Indexed: 01/05/2023]
Abstract
Polyester-based scaffolds covalently functionalized with arginine-glycine-aspartic acid-cysteine (RGDC) peptide sequences support the proliferation and osteogenic differentiation of stem cells. The aim is to create an optimized 3D niche to sustain human bone marrow stem cell (hBMSC) viability and osteogenic commitment, without reliance on differentiation media. Scaffolds consisting of poly(lactide-co-trimethylene carbonate), poly(LA-co-TMC), and functionalized poly(lactide) copolymers with pendant thiol groups are prepared by salt-leaching technique. The availability of functional groups on scaffold surfaces allows for an easy and straightforward method to covalently attach RGDC peptide motifs without affecting the polymerization degree. The strategy enables the chemical binding of bioactive motifs on the surfaces of 3D scaffolds and avoids conventional methods that require harsh conditions. Gene and protein levels and mineral deposition indicate the osteogenic commitment of hBMSC cultured on the RGDC functionalized surfaces. The osteogenic commitment of hBMSC is enhanced on functionalized surfaces compared with nonfunctionalized surfaces and without supplementing media with osteogenic factors. Poly(LA-co-TMC) scaffolds have potential as scaffolds for osteoblast culture and bone grafts. Furthermore, these results contribute to the development of biomimetic materials and allow a deeper comprehension of the importance of RGD peptides on stem cell transition toward osteoblastic lineage.
Collapse
Affiliation(s)
- Mohammed A Yassin
- Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, Teknikringen, 56-58, SE, 100-44, Stockholm, Sweden.,Department of Clinical Dentistry, Årstadveien 19, 5009 Bergen, Bergen, Norway
| | - Tiziana Fuoco
- Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, Teknikringen, 56-58, SE, 100-44, Stockholm, Sweden
| | - Samih Mohamed-Ahmed
- Department of Clinical Dentistry, Årstadveien 19, 5009 Bergen, Bergen, Norway
| | - Kamal Mustafa
- Department of Clinical Dentistry, Årstadveien 19, 5009 Bergen, Bergen, Norway
| | - Anna Finne-Wistrand
- Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, Teknikringen, 56-58, SE, 100-44, Stockholm, Sweden
| |
Collapse
|
36
|
Zhang Z, Zhang X, Zhao D, Liu B, Wang B, Yu W, Li J, Yu X, Cao F, Zheng G, Zhang Y, Liu Y. TGF‑β1 promotes the osteoinduction of human osteoblasts via the PI3K/AKT/mTOR/S6K1 signalling pathway. Mol Med Rep 2019; 19:3505-3518. [PMID: 30896852 PMCID: PMC6471541 DOI: 10.3892/mmr.2019.10051] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 03/06/2019] [Indexed: 12/22/2022] Open
Abstract
Transforming growth factor β1 (TGF-β1) has been suggested to be a candidate cytokine in the field of bone tissue engineering. Cytokines serve important roles in tissue engineering, particularly in the repair of bone damage; however, the underlying molecular mechanisms remain unclear. In the present study, the effects of TGF-β1 on the osteogenesis and motility of hFOB1.19 human osteoblasts were demonstrated via the phenotype and gene expression of cells. Additionally, the role of the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin/S6 kinase 1 (PI3K/AKT/mTOR/S6K1) signalling pathway in the effects of TGF-β1 on osteoblasts was investigated. It was demonstrated using Cell Counting Kit-8 and flow cytometry assays that the proliferation of human osteoblasts was promoted by 1 ng/ml TGF-β1. In addition, alkaline phosphatase activity, Alizarin red staining, scratch-wound and Transwell assays were conducted. It was revealed that osteogenesis and the migration of cells were regulated by TGF-β1 via the upregulation of osteogenic and migration-associated genes. Alterations in the expression of osteogenesis- and migration-associated genes were evaluated following pre-treatment with a PI3K/AKT inhibitor (LY294002) and an mTOR/S6K1 inhibitor (rapamycin), with or without TGF-β1. The results indicated that TGF-β1 affected the osteogenesis and mineralisation of osteoblasts via the PI3K/AKT signalling pathway. Furthermore, TGF-β1 exhibited effects on mTOR/S6K1 downstream of PI3K/AKT. The present study demonstrated that TGF-β1 promoted the proliferation, differentiation and migration of human hFOB1.19 osteoblasts, and revealed that TGF-β1 affected the biological activity of osteoblasts via the PI3K/AKT/mTOR/S6K1 signalling pathway. Our findings may provide novel insight to aid the development of bone tissue engineering methods for the treatment of bone injury.
Collapse
Affiliation(s)
- Zhaodong Zhang
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Xiuzhi Zhang
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Dewei Zhao
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Baoyi Liu
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Benjie Wang
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Weiting Yu
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Junlei Li
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Xiaobing Yu
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Fang Cao
- Department of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian, Liaoning 116024, P.R. China
| | - Guoshuang Zheng
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Yao Zhang
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Yupeng Liu
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| |
Collapse
|
37
|
Xu R, Zhang Z, Toftdal MS, Møller AC, Dagnaes-Hansen F, Dong M, Thomsen JS, Brüel A, Chen M. Synchronous delivery of hydroxyapatite and connective tissue growth factor derived osteoinductive peptide enhanced osteogenesis. J Control Release 2019; 301:129-139. [PMID: 30880079 DOI: 10.1016/j.jconrel.2019.02.037] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/21/2019] [Accepted: 02/25/2019] [Indexed: 10/27/2022]
Abstract
In bone tissue engineering, electrospun fibrous scaffolds can provide excellent mechanical support, extracellular matrix mimicking components, such as 3D spacial fibrous environment for cell growth and controlled release of signaling molecules for osteogenesis. Here, a facile strategy comprising the incorporation of an osteogenic inductive peptide H1, derived from the cysteine knot (CT) domain of connective tissue growth factor (CTGF), in the core of Silk Fibroin (SF) was developed for osteogenic induction, synergistically with co-delivering hydroxyapatite (HA) from the shell of poly(l-lactic acid-co-ε-caprolactone) (PLCL). The core-shell nanofibrous structure was confirmed by transmission electron microscopy (TEM). Furthermore, the sustained released H1 has effectively promoted proliferation and osteoblastic differentiation of human induced pluripotent stem cells-derived mesenchymal stem cells (hiPS-MSCs). Moreover, after 8 weeks implantation in mice, this SF-H1/PLCL-HA composite induced bone tissue formation significantly faster than SF/PLCL as indicated by μCT. The present study is the first to demonstrate that release of short hydrophilic peptides derived from CTGF combined with HA potentiated the regenerative capacity for healing critical sized calvarial defect in vivo.
Collapse
Affiliation(s)
- Ruodan Xu
- Department of Engineering, Aarhus University, DK-8000 Aarhus C, Denmark; Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medicine Science, China
| | - Zhongyang Zhang
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, DK-8000 Aarhus C, Denmark
| | | | | | - Frederik Dagnaes-Hansen
- Institute of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, DK-8000 Aarhus C, Denmark
| | - Mingdong Dong
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, DK-8000 Aarhus C, Denmark.
| | - Jesper Skovhus Thomsen
- Institute of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, DK-8000 Aarhus C, Denmark
| | - Annemarie Brüel
- Institute of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, DK-8000 Aarhus C, Denmark
| | - Menglin Chen
- Department of Engineering, Aarhus University, DK-8000 Aarhus C, Denmark; Interdisciplinary Nanoscience Center (iNANO), Aarhus University, DK-8000 Aarhus C, Denmark.
| |
Collapse
|
38
|
Mas-Moruno C, Su B, Dalby MJ. Multifunctional Coatings and Nanotopographies: Toward Cell Instructive and Antibacterial Implants. Adv Healthc Mater 2019; 8:e1801103. [PMID: 30468010 DOI: 10.1002/adhm.201801103] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/15/2018] [Indexed: 01/02/2023]
Abstract
In biomaterials science, it is nowadays well accepted that improving the biointegration of dental and orthopedic implants with surrounding tissues is a major goal. However, implant surfaces that support osteointegration may also favor colonization of bacterial cells. Infection of biomaterials and subsequent biofilm formation can have devastating effects and reduce patient quality of life, representing an emerging concern in healthcare. Conversely, efforts toward inhibiting bacterial colonization may impair biomaterial-tissue integration. Therefore, to improve the long-term success of medical implants, biomaterial surfaces should ideally discourage the attachment of bacteria without affecting eukaryotic cell functions. However, most current strategies seldom investigate a combined goal. This work reviews recent strategies of surface modification to simultaneously address implant biointegration while mitigating bacterial infections. To this end, two emerging solutions are considered, multifunctional chemical coatings and nanotopographical features.
Collapse
Affiliation(s)
- Carlos Mas-Moruno
- Biomaterials, Biomechanics and Tissue Engineering Group; Department of Materials Science and Engineering & Center in Multiscale Science and Engineering; Universitat Politècnica de Catalunya (UPC); Barcelona 08019 Spain
| | - Bo Su
- Bristol Dental School; University of Bristol; Bristol BS1 2LY UK
| | - Matthew J. Dalby
- Centre for Cell Engineering; University of Glasgow; Glasgow G12 UK
| |
Collapse
|
39
|
Padiolleau L, Chanseau C, Durrieu S, Chevallier P, Laroche G, Durrieu MC. Single or Mixed Tethered Peptides To Promote hMSC Differentiation toward Osteoblastic Lineage. ACS APPLIED BIO MATERIALS 2018; 1:1800-1809. [DOI: 10.1021/acsabm.8b00236] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Laurence Padiolleau
- Chimie et Biologie des Membranes et Nano-Objets (UMR5248 CBMN), Université de Bordeaux, Pessac, France
- Laboratoire d’Ingénierie de Surface (LIS), Département de Génie des Mines, de la Métallurgie et des Matériaux, Centre de Recherche sur les Matériaux Avancés (CERMA), Université Laval, Québec G1V 0A6, Canada
- Hôpital St-François d’Assise, Centre de Recherche du Centre Hospitalier Universitaire de Québec (CRCHUQ), Québec G1L 3L5, Canada
| | - Christel Chanseau
- Chimie et Biologie des Membranes et Nano-Objets (UMR5248 CBMN), Université de Bordeaux, Pessac, France
| | - Stéphanie Durrieu
- ARNA Laboratory, Université de Bordeaux, 33076 Bordeaux, France
- ARNA Laboratory, INSERM, U1212 − CNRS UMR 5320, 33000 Bordeaux, France
| | - Pascale Chevallier
- Laboratoire d’Ingénierie de Surface (LIS), Département de Génie des Mines, de la Métallurgie et des Matériaux, Centre de Recherche sur les Matériaux Avancés (CERMA), Université Laval, Québec G1V 0A6, Canada
- Hôpital St-François d’Assise, Centre de Recherche du Centre Hospitalier Universitaire de Québec (CRCHUQ), Québec G1L 3L5, Canada
| | - Gaétan Laroche
- Laboratoire d’Ingénierie de Surface (LIS), Département de Génie des Mines, de la Métallurgie et des Matériaux, Centre de Recherche sur les Matériaux Avancés (CERMA), Université Laval, Québec G1V 0A6, Canada
- Hôpital St-François d’Assise, Centre de Recherche du Centre Hospitalier Universitaire de Québec (CRCHUQ), Québec G1L 3L5, Canada
| | - Marie-Christine Durrieu
- Chimie et Biologie des Membranes et Nano-Objets (UMR5248 CBMN), Université de Bordeaux, Pessac, France
| |
Collapse
|
40
|
Charoenlarp P, Rajendran AK, Fujihara R, Kojima T, Nakahama KI, Sasaki Y, Akiyoshi K, Takechi M, Iseki S. The improvement of calvarial bone healing by durable nanogel-crosslinked materials. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2018; 29:1876-1894. [DOI: 10.1080/09205063.2018.1517403] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Pornkawee Charoenlarp
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
- Department of Radiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Arun Kumar Rajendran
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Rie Fujihara
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Nishikyo-ku, Kyoto, Japan
| | - Taisei Kojima
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Nishikyo-ku, Kyoto, Japan
| | - Ken-ichi Nakahama
- Department of Cellular Physiological Chemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Yoshihiro Sasaki
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Nishikyo-ku, Kyoto, Japan
| | - Kazunari Akiyoshi
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Nishikyo-ku, Kyoto, Japan
| | - Masaki Takechi
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Sachiko Iseki
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
41
|
Gan D, Liu M, Xu T, Wang K, Tan H, Lu X. Chitosan/biphasic calcium phosphate scaffolds functionalized with BMP-2-encapsulated nanoparticles and RGD for bone regeneration. J Biomed Mater Res A 2018; 106:2613-2624. [PMID: 29790251 DOI: 10.1002/jbm.a.36453] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/19/2018] [Accepted: 05/11/2018] [Indexed: 12/24/2022]
Abstract
Advancements in bone tissue engineering require the improvement of tissue scaffolds, which should not only exhibit suitable mechanical properties and highly porous structures, but also effectively carry signaling molecules that can mediate bone formation and tissue regeneration. In the present study, we established chitosan/biphasic calcium phosphate (CS/BCP) scaffolds functionalized with Arg-Gly-Asp (RGD) and BMP-2-loaded nanoparticles. The resulting scaffolds were highly similar to natural bone extracellular matrix (ECM) in terms of composition and structural properties. First, we synthesized CS/BCP composite bionic scaffolds via the freeze-drying method. Then, RGD peptides were covalently conjugated onto the scaffolds via the EDC/NHS method. The BMP-2-encapsulated BSA nanoparticles were prepared via a desolvation method and then coated with CS and oxidized alginate to achieve sustained release of BMP-2. In vitro cell culture and in vivo implantation tests confirmed that RGD and BMP-2 synergistically enhanced cell attachment and spreading by providing integrin binding surface and facilitating osteogenic differentiation. In summary, the bioceramic/biopolymer scaffolds functionalized with signaling biomolecules successfully provided a favorable microenvironment for bone formation and thus serve as potential candidates for use in bone tissue engineering. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 2613-2624, 2018.
Collapse
Affiliation(s)
- Donglin Gan
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Min Liu
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Tong Xu
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Kefeng Wang
- National Engineering Research Center for Biomaterials, Genome Research Center for Biomaterials Sichuan University, Chengdu, Sichuan, 610064, China
| | - Hui Tan
- Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 518035, China
| | - Xiong Lu
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| |
Collapse
|
42
|
Hu S, Chen H, Zhou X, Chen G, Hu K, Cheng Y, Wang L, Zhang F. Thermally induced self-agglomeration 3D scaffolds with BMP-2-loaded core-shell fibers for enhanced osteogenic differentiation of rat adipose-derived stem cells. Int J Nanomedicine 2018; 13:4145-4155. [PMID: 30046239 PMCID: PMC6054293 DOI: 10.2147/ijn.s167035] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Introduction Scaffold structure plays a vital role in cell behaviors. Compared with two-dimensional structure, 3D scaffolds can mimic natural extracellular matrix (ECM) and promote cell–cell and cell–matrix interactions. The combination of osteoconductive scaffolds and osteoinductive growth factors is considered to have synergistic effects on bone regeneration. Materials and methods In this study, core–shell poly(lactide-co-glycolide) (PLGA)/polycaprolactone (PCL)–BMP-2 (PP–B) fibrous scaffolds were prepared through coaxial electrospinning. Next, we fabricated 3D scaffolds based on PP–B fibers with thermally induced self-agglomeration (TISA) method and compared with conventional PLGA/PCL scaffolds in terms of scaffold morphology and BMP-2 release behaviors. Then, rat adipose-derived stem cells (rADSCs) were seeded on the scaffolds, and the effects on cell proliferation, cell morphology, and osteogenic differentiation of rADSCs were detected. Results The results demonstrated that 3D scaffold incorporated with BMP-2 significantly increased proliferation and osteogenic differentiation of rADSCs, followed by PP–B group. Conclusion Our findings indicate that scaffolds with 3D structure and osteoinductive growth factors have great potential in bone tissue engineering.
Collapse
Affiliation(s)
- Shuying Hu
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China,
| | - Hanbang Chen
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China,
| | - Xuefeng Zhou
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Gang Chen
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China,
| | - Ke Hu
- Key Laboratory of Clinical and Medical Engineering, Department of Biomedical Engineering, School of Basic Medical Science, Nanjing Medical University, Nanjing 210000, China
| | - Yi Cheng
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China,
| | - Lili Wang
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China,
| | - Feimin Zhang
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China,
| |
Collapse
|
43
|
Wang J, Meng F, Song W, Jin J, Ma Q, Fei D, Fang L, Chen L, Wang Q, Zhang Y. Nanostructured titanium regulates osseointegration via influencing macrophage polarization in the osteogenic environment. Int J Nanomedicine 2018; 13:4029-4043. [PMID: 30022825 PMCID: PMC6045901 DOI: 10.2147/ijn.s163956] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Introduction Fabricating nanostructured surface topography represents the mainstream approach to induce osteogenesis for the next-generation bone implant. In the past, the bone implant was designed to minimize host repulsive reactions in order to acquire biocompatibility. However, increasing reports indicate that the absence of an appropriate immune response cannot acquire adequate osseointegration after implantation in vivo. Materials and methods We prepared different topographies on the surface of titanium (Ti) specimens by grinding, etching and anodizing, and they were marked as polished specimen (P), specimen with nanotubes (NTs) in small diameters (NT-30) and specimen with NTs in large diameters (NT-100). We evaluated the ability of different topographies of the specimen to induce osteogenic differentiation of mice bone marrow mesenchymal stem cells (BMSCs) in vitro and to induce osseointegration in vivo. Furthermore, we investigated the effect of different topographies on the polarization and secretion of macrophages, and the effect of macrophage polarization on topography-induced osteogenic differentiation of mice BMSCs. Finally, we verified the effect of macrophage polarization on topography-induced osseointegration in vivo by using Cre*RBP-Jfl/fl mice in which classically activated macrophage was restrained. Results The osteogenic differentiation of mice BMSCs induced by specimen with different topographies was NT-100>NT-30>P, while the osseointegration induced by specimen with different topographies in vivo was NT-30>NT-100>P. In addition, specimen of NT-30 could induce more macrophages to M2 polarization, while specimen of P and NT-100 could induce more macrophages to M1 polarization. When co-culture mice BMSCs and macrophages on specimen with different topographies, the osteogenic differentiation of mice BMSCs was NT-30>NT-100≥P. The osseointegration induced by NT-100 in Cre*RBP-Jfl/fl mice was much better than that of wild type mice. Conclusion It is suggested that the intrinsic immunomodulatory effects of nanomaterials are not only crucial to evaluate the in vivo biocompatibility but also required to determine the final osseointegration. To clarify the immune response and osseointegration may be beneficial for the designation and optimization of the bone implant.
Collapse
Affiliation(s)
- Jinjin Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shannxi Province, China, .,Department of Immunology, School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shannxi Province, China
| | - Fanhui Meng
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shannxi Province, China,
| | - Wen Song
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shannxi Province, China,
| | - Jingyi Jin
- Department of Immunology, School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shannxi Province, China
| | - Qianli Ma
- Department of Immunology, School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shannxi Province, China
| | - Dongdong Fei
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shannxi Province, China,
| | - Liang Fang
- Department of Immunology, School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shannxi Province, China
| | - Lihua Chen
- Department of Immunology, School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shannxi Province, China
| | - Qintao Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shannxi Province, China,
| | - Yumei Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shannxi Province, China,
| |
Collapse
|
44
|
Bilem I, Plawinski L, Chevallier P, Ayela C, Sone ED, Laroche G, Durrieu MC. The spatial patterning of RGD and BMP-2 mimetic peptides at the subcellular scale modulates human mesenchymal stem cells osteogenesis. J Biomed Mater Res A 2017; 106:959-970. [DOI: 10.1002/jbm.a.36296] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/06/2017] [Accepted: 11/10/2017] [Indexed: 11/10/2022]
Affiliation(s)
- I. Bilem
- Laboratoire d'Ingénierie de Surface, Centre de Recherche sur les Matériaux Avancés, Département de Génie des Mines, de la Métallurgie et des Matériaux; Université Laval, 1065 Avenue de la médecine; Québec G1V 0A6 Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, Hôpital St-François d'Assise, 10 rue de l'Espinay; Québec G1L 3L5 Canada
- CNRS, Institute of Chemistry & Biology of Membranes & Nanoobjects (CBMN 5248); Pessac F-33600 France
- Bordeaux INP, CBMN, UMR 5248; Pessac F-33600 France
| | - L. Plawinski
- CNRS, Institute of Chemistry & Biology of Membranes & Nanoobjects (CBMN 5248); Pessac F-33600 France
- Bordeaux INP, CBMN, UMR 5248; Pessac F-33600 France
| | - P. Chevallier
- Laboratoire d'Ingénierie de Surface, Centre de Recherche sur les Matériaux Avancés, Département de Génie des Mines, de la Métallurgie et des Matériaux; Université Laval, 1065 Avenue de la médecine; Québec G1V 0A6 Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, Hôpital St-François d'Assise, 10 rue de l'Espinay; Québec G1L 3L5 Canada
| | - C. Ayela
- Université de Bordeaux, IMS, UMR CNRS 5218; Talence F-33400 France
| | - E. D. Sone
- Institute of Biomaterials and Biomedical Engineering, Department of Materials Science and Engineering, and Faculty of Dentistry; University of Toronto; Toronto ON M5S 3G9 Canada
| | - G. Laroche
- Laboratoire d'Ingénierie de Surface, Centre de Recherche sur les Matériaux Avancés, Département de Génie des Mines, de la Métallurgie et des Matériaux; Université Laval, 1065 Avenue de la médecine; Québec G1V 0A6 Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, Hôpital St-François d'Assise, 10 rue de l'Espinay; Québec G1L 3L5 Canada
| | - M. C. Durrieu
- CNRS, Institute of Chemistry & Biology of Membranes & Nanoobjects (CBMN 5248); Pessac F-33600 France
- Bordeaux INP, CBMN, UMR 5248; Pessac F-33600 France
| |
Collapse
|
45
|
Li S, Xu Y, Yu J, Becker ML. Enhanced osteogenic activity of poly(ester urea) scaffolds using facile post-3D printing peptide functionalization strategies. Biomaterials 2017; 141:176-187. [DOI: 10.1016/j.biomaterials.2017.06.038] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 06/05/2017] [Accepted: 06/27/2017] [Indexed: 12/28/2022]
|
46
|
Sun T, Qu Y, Cui W, Yang L, Ji Y, Yu W, Navinduth R, Shao Z, Yang H, Guo X. Evaluation of osteogenic inductivity of a novel BMP2-mimicking peptide P28 and P28-containing bone composite. J Biomed Mater Res A 2017; 106:210-220. [PMID: 28884509 DOI: 10.1002/jbm.a.36228] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 08/01/2017] [Accepted: 08/07/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Tingfang Sun
- Department of Orthopedics; Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Wuhan 430022 China
| | - Yanzhen Qu
- Department of Orthopedics; Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Wuhan 430022 China
| | - Wei Cui
- Department of Orthopedics; Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Wuhan 430022 China
| | - Liang Yang
- Department of Orthopedics; Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Wuhan 430022 China
| | - Yanhui Ji
- Department of Orthopedics; Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Wuhan 430022 China
| | - Wei Yu
- Department of Orthopedics; Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Wuhan 430022 China
| | - Ramphul Navinduth
- Department of Orthopedics; Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Wuhan 430022 China
| | - Zengwu Shao
- Department of Orthopedics; Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Wuhan 430022 China
| | - Hu Yang
- Department of Chemical and Life Science Engineering; Virginia Commonwealth University; Richmond Virginia 23219
- Department of Pharmaceutics; Virginia Commonwealth University; Richmond Virginia 23298
- Massey Cancer Center; Virginia Commonwealth University; Richmond Virginia 23298
| | - Xiaodong Guo
- Department of Orthopedics; Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Wuhan 430022 China
| |
Collapse
|
47
|
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: 1.6] [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
|
48
|
Li X, Jia J, Mei Y, Latour RA. Molecular modeling to predict peptide accessibility for peptide-functionalized hydrogels. Biointerphases 2017; 12:031008. [PMID: 28821213 PMCID: PMC5562506 DOI: 10.1116/1.4992101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 07/28/2017] [Accepted: 08/07/2017] [Indexed: 12/16/2022] Open
Abstract
Peptide-functionalized (PF) hydrogels are being widely investigated by the tissue engineering and regenerative medicine communities for a broad range of applications because of their unique potential to mimic the natural extracellular matrix and promote tissue regeneration. In order for these complex material systems to perform their intended bioactive function (e.g., cell signaling), the peptides that are tethered to the hydrogel matrix must be accessible at the hydrogel surface for cell-receptor binding. The factors influencing the surface accessibility of the tethered peptide mainly include the length of the tethers, the loading (i.e., concentration) of the peptide, and the association between the tethered peptide and the hydrogel matrix. In the present work, the authors developed coarse-grained molecular models based on the all-atom polymer consistent force field for a type of poly(ethylene glycol)-based PF hydrogel and conducted molecular simulations to investigate the distribution of the peptide within the hydrogel and its surface accessibility as a function of tether length and peptide concentration. The calculated results of the effects of these design parameters on the surface accessibility of the peptide agree very well with corresponding experimental measurements in which peptide accessibility was quantified by the number of cells attached to the hydrogel surface per unit area. The developed modeling methods are able to provide unique insights into the molecular behavior of PF hydrogels and the distribution of the tethered peptides, which can serve as a guide for hydrogel design optimization.
Collapse
Affiliation(s)
- Xianfeng Li
- Department of Bioengineering, Clemson University, Clemson, South Carolina 29634
| | - Jia Jia
- Department of Bioengineering, Clemson University, Clemson, South Carolina 29634
| | - Ying Mei
- Department of Bioengineering, Clemson University, Clemson, South Carolina 29634
| | - Robert A Latour
- Department of Bioengineering, Clemson University, Clemson, South Carolina 29634
| |
Collapse
|
49
|
Surface biofunctionalization of three-dimensional porous poly(lactic acid) scaffold using chitosan/OGP coating for bone tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 77:92-101. [DOI: 10.1016/j.msec.2017.03.220] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 01/09/2017] [Accepted: 03/24/2017] [Indexed: 11/24/2022]
|
50
|
Camarero-Espinosa S, Cooper-White J. Tailoring biomaterial scaffolds for osteochondral repair. Int J Pharm 2017; 523:476-489. [DOI: 10.1016/j.ijpharm.2016.10.035] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/11/2016] [Accepted: 10/17/2016] [Indexed: 12/11/2022]
|