1
|
Zhou T, C. Cavalcante R, Ge C, Franceschi RT, Ma PX. Synthetic helical peptides on nanofibers to activate cell-surface receptors and synergistically enhance critical-sized bone defect regeneration. Bioact Mater 2025; 43:98-113. [PMID: 39381328 PMCID: PMC11458538 DOI: 10.1016/j.bioactmat.2024.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/15/2024] [Accepted: 08/17/2024] [Indexed: 10/10/2024] Open
Abstract
More than 500,000 bone grafting procedures are performed annually in the USA. Considering the significant limitations of available bone grafts, we previously invented a phase-separation technology to generate nanofibrous poly(l-lactic acid) (PLLA) scaffolds that mimic the bone matrix collagen in nanofiber geometry and enhance bone regeneration. Here we report the development of nanofibrous scaffolds with covalently attached synthetic peptides that mimic native collagen peptides to activate the two main collagen receptors in bone cells, discoidin domain receptor 2 (DDR2) and β1 integrins. We synthesized a PLLA-based graft-copolymer to enable covalent peptide conjugation via a click reaction. Using PLLA and the graft-copolymer, we developed 3D scaffolds with interconnected pores and peptides-containing nanofibers to activate DDR2 and β1 integrins of osteogenic cells. The degradation rate and mechanical properties of the scaffolds are tunable. The peptides-decorated nanofibrous scaffolds demonstrated 7.8 times more mineralized bone regeneration over the control scaffolds without the peptides in a critical-sized bone defect regeneration model after 8 weeks of implantation, showing a synergistic effect of the two peptides. This study demonstrates the power of scaffolds to mimic ECM at both nanometer and molecular levels, activating cell surface receptors to liberate the innate regenerative potential of host stem/progenitor cells.
Collapse
Affiliation(s)
- Tongqing Zhou
- Macromolecular Science and Engineering Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rafael C. Cavalcante
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Chunxi Ge
- Department of Periodontics and Oral Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Renny T. Franceschi
- Department of Periodontics and Oral Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Peter X. Ma
- Macromolecular Science and Engineering Center, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
2
|
Rambhia KJ, Sun H, Feng K, Kannan R, Doleyres Y, Holzwarth JM, Doepker M, Franceschi RT, Ma PX. Nanofibrous 3D scaffolds capable of individually controlled BMP and FGF release for the regulation of bone regeneration. Acta Biomater 2024:S1742-7061(24)00636-6. [PMID: 39486780 DOI: 10.1016/j.actbio.2024.10.044] [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: 04/13/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
The current clinical applications of bone morphogenetic proteins (BMPs) are limited to only a few specific indications. Locally controlled delivery of combinations of growth factors can be a promising strategy to improve BMP-based bone repair. However, the success of this approach requires the development of an effective release system and the correct choice of growth factors capable of enhancing BMP activity. Basic fibroblast growth factor (bFGF, also known as FGF-2) has shown promise in promoting bone repair, although conflicting results have been reported. Considering the complex biological activities of FGF-2, we hypothesized that FGF-2 can promote BMP-induced bone regeneration only if the dosage and kinetic parameters of the two factors are individually tailored. In this study, we conducted systematic in vitro studies on cell proliferation, differentiation, and mineralization in response to factor dose, delivery mode (sequential or simultaneous), and release rate. Subsequently, we designed individually controlled BMP-7 and FGF-2 release poly(lactide-co-glycolide) (PLGA) nanospheres attached to the poly(l-lactic acid) (PLLA) nanofibrous scaffolds. The data showed that BMP-7-induced bone formation was accelerated by a relatively higher FGF-2 dose (100 ng/scaffold) delivered at a faster release rate, or by a relatively lower FGF-2 dose (10 ng/scaffold) at a slower release rate in an in vivo bone regeneration model. In contrast, a very high dose of FGF-2 (1000 ng/scaffold) inhibited bone regeneration under all conditions. In vitro and in vivo data suggest that FGF-2 improved BMP-7-induced bone regeneration by coordinating FGF-2 dosage and release kinetics to enhance stem cell migration, proliferation, and angiogenesis. STATEMENT OF SIGNIFICANCE: Bone morphogenetic proteins (BMPs) are the most potent growth/differentiation factors in bone development and regeneration. However, the clinical applications of BMPs have been limited to only a few specific indications due to the required supraphysiological dosages with the current BMP products and their side effects. Locally controlled delivery of BMPs and additional growth factors that can enhance their osteogenic potency are highly desired. However, different growth factors act with different mechanisms. Here we report a nanofibrous scaffold that mimics collagen in size and geometry and is immobilized with biodegradable nanospheres to achieve local and distinct release profiles of BMP7 and FGF2. Systematic studies demonstrated low dose BMP7 and FGF2 with different temporal release profiles can optimally enhance bone regeneration.
Collapse
Affiliation(s)
- Kunal J Rambhia
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hongli Sun
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kai Feng
- Macromolecular Science and Engineering Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rahasudha Kannan
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yasmine Doleyres
- Macromolecular Science and Engineering Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jeremy M Holzwarth
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mikayla Doepker
- Department of Biology, Kalamazoo College, Kalamazoo, MI 49006, USA
| | - Renny T Franceschi
- Department of Periodontics and Oral Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Peter X Ma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Macromolecular Science and Engineering Center, University of Michigan, Ann Arbor, MI 48109, USA; Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
3
|
Nelogi SY, Patil AK, Chowdhary R. Enhancing bone tissue engineering using iron nanoparticles and magnetic fields: A focus on cytomechanics and angiogenesis in the chicken egg chorioallantoic membrane model. J Indian Prosthodont Soc 2024; 24:175-185. [PMID: 38650343 PMCID: PMC11129814 DOI: 10.4103/jips.jips_440_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 02/22/2024] [Accepted: 03/03/2024] [Indexed: 04/25/2024] Open
Abstract
AIM To evaluate the potential of iron nanoparticles (FeNPs) in conjunction with magnetic fields (MFs) to enhance osteoblast cytomechanics, promote cell homing, bone development activity, and antibacterial capabilities, and to assess their in vivo angiogenic viability using the chicken egg chorioallantoic membrane (CAM) model. SETTINGS AND DESIGN Experimental study conducted in a laboratory setting to investigate the effects of FeNPs and MFs on osteoblast cells and angiogenesis using a custom titanium (Ti) substrate coated with FeNPs. MATERIALS AND METHODS A custom titanium (Ti) was coated with FeNPs. Evaluations were conducted to analyze the antibacterial properties, cell adhesion, durability, physical characteristics, and nanoparticle absorption associated with FeNPs. Cell physical characteristics were assessed using protein markers, and microscopy, CAM model, was used to quantify blood vessel formation and morphology to assess the FeNP-coated Ti's angiogenic potential. This in vivo study provided critical insights into tissue response and regenerative properties for biomedical applications. STATISTICAL ANALYSIS Statistical analysis was performed using appropriate tests to compare experimental groups and controls. Significance was determined at P < 0.05. RESULTS FeNPs and MFs notably improved osteoblast cell mechanical properties facilitated the growth and formation of new blood vessels and bone tissue and promoted cell migration to targeted sites. In the group treated with FeNPs and exposed to MFs, there was a significant increase in vessel percentage area (76.03%) compared to control groups (58.11%), along with enhanced mineralization and robust antibacterial effects (P < 0.05). CONCLUSION The study highlights the promising potential of FeNPs in fostering the growth of new blood vessels, promoting the formation of bone tissue, and facilitating targeted cell migration. These findings underscore the importance of further investigating the mechanical traits of FeNPs, as they could significantly advance the development of effective bone tissue engineering techniques, ultimately enhancing clinical outcomes in the field.
Collapse
Affiliation(s)
- Santosh Yamanappa Nelogi
- Department of Prosthodontics, KLEVK Institute of Dental Sciences, KLE Academy of Higher Education and Research, Belgavi, Karnataka, India
| | - Anand Kumar Patil
- Department of Prosthodontics, KLEVK Institute of Dental Sciences, KLE Academy of Higher Education and Research, Belgavi, Karnataka, India
| | - Ramesh Chowdhary
- Department of Prosthodontics, Siddhartha Institute of Dental Sciences, Tumakuru, Karnataka, India
| |
Collapse
|
4
|
Zhao P, Yang F, Jia X, Xiao Y, Hua C, Xing M, Lyu G. Extracellular Matrices as Bioactive Materials for In Situ Tissue Regeneration. Pharmaceutics 2023; 15:2771. [PMID: 38140112 PMCID: PMC10747903 DOI: 10.3390/pharmaceutics15122771] [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: 08/19/2023] [Revised: 10/28/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Bioactive materials based on a nature-derived extracellular matrix (NECM) represent a category of biomedical devices with versatile therapeutic applications in the realms of tissue repair and engineering. With advancements in decellularization technique, the inherent bioactive molecules and the innate nano-structural and mechanical properties are preserved in three-dimensional scaffolds mainly composed of collagens. Techniques such as electrospinning, three-dimensional printing, and the intricate fabrication of hydrogels are developed to mimic the physical structures, biosignalling and mechanical cues of ECM. Until now, there has been no approach that can fully account for the multifaceted properties and diverse applications of NECM. In this review, we introduce the main proteins composing NECMs and explicate the importance of them when used as therapeutic devices in tissue repair. Nano-structural features of NECM and their applications regarding tissue repair are summarized. The origins, degradability, and mechanical property of and immune responses to NECM are also introduced. Furthermore, we review their applications, and clinical features thereof, in the repair of acute and chronic wounds, abdominal hernia, breast deformity, etc. Some typical marketed devices based on NECM, their indications, and clinical relevance are summarized.
Collapse
Affiliation(s)
- Peng Zhao
- Burn & Trauma Treatment Center, Affiliated Hospital of Jiangnan University, Wuxi 214122, China
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi 214000, China; (F.Y.); (Y.X.)
| | - Fengbo Yang
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi 214000, China; (F.Y.); (Y.X.)
| | - Xiaoli Jia
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi 214000, China; (F.Y.); (Y.X.)
| | - Yuqin Xiao
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi 214000, China; (F.Y.); (Y.X.)
| | - Chao Hua
- Burn & Trauma Treatment Center, Affiliated Hospital of Jiangnan University, Wuxi 214122, China
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Guozhong Lyu
- Burn & Trauma Treatment Center, Affiliated Hospital of Jiangnan University, Wuxi 214122, China
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi 214000, China; (F.Y.); (Y.X.)
| |
Collapse
|
5
|
Bello SA, Cruz-Lebrón J, Rodríguez-Rivera OA, Nicolau E. Bioactive Scaffolds as a Promising Alternative for Enhancing Critical-Size Bone Defect Regeneration in the Craniomaxillofacial Region. ACS APPLIED BIO MATERIALS 2023; 6:4465-4503. [PMID: 37877225 DOI: 10.1021/acsabm.3c00432] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
Reconstruction of critical-size bone defects (CSDs) in the craniomaxillofacial (CMF) region remains challenging. Scaffold-based bone-engineered constructs have been proposed as an alternative to the classical treatments made with autografts and allografts. Scaffolds, a key component of engineered constructs, have been traditionally viewed as biologically passive temporary replacements of deficient bone lacking intrinsic cues to promote osteogenesis. Nowadays, scaffolds are functionalized, giving rise to bioactive scaffolds promoting bone regeneration more effectively than conventional counterparts. This review focuses on the three approaches most used to bioactivate scaffolds: (1) conferring microarchitectural designs or surface nanotopography; (2) loading bioactive molecules; and (3) seeding stem cells on scaffolds, providing relevant examples of in vivo (preclinical and clinical) studies where these methods are employed to enhance CSDs healing in the CMF region. From these, adding bioactive molecules (specifically bone morphogenetic proteins or BMPs) to scaffolds has been the most explored to bioactivate scaffolds. Nevertheless, the downsides of grafting BMP-loaded scaffolds in patients have limited its successful translation into clinics. Despite these drawbacks, scaffolds containing safer, cheaper, and more effective bioactive molecules, combined with stem cells and topographical cues, remain a promising alternative for clinical use to treat CSDs in the CMF complex replacing autografts and allografts.
Collapse
Affiliation(s)
- Samir A Bello
- Department of Chemistry, University of Puerto Rico, Rio Piedras Campus, PO Box 23346, San Juan, Puerto Rico 00931, United States
- Molecular Sciences Research Center, University of Puerto Rico, 1390 Ponce De León Ave, Suite 1-7, San Juan, Puerto Rico 00926, United States
| | - Junellie Cruz-Lebrón
- Department of Chemistry, University of Puerto Rico, Rio Piedras Campus, PO Box 23346, San Juan, Puerto Rico 00931, United States
- Molecular Sciences Research Center, University of Puerto Rico, 1390 Ponce De León Ave, Suite 1-7, San Juan, Puerto Rico 00926, United States
| | - Osvaldo A Rodríguez-Rivera
- Department of Chemistry, University of Puerto Rico, Rio Piedras Campus, PO Box 23346, San Juan, Puerto Rico 00931, United States
- Molecular Sciences Research Center, University of Puerto Rico, 1390 Ponce De León Ave, Suite 1-7, San Juan, Puerto Rico 00926, United States
| | - Eduardo Nicolau
- Department of Chemistry, University of Puerto Rico, Rio Piedras Campus, PO Box 23346, San Juan, Puerto Rico 00931, United States
- Molecular Sciences Research Center, University of Puerto Rico, 1390 Ponce De León Ave, Suite 1-7, San Juan, Puerto Rico 00926, United States
| |
Collapse
|
6
|
Doostmohammadi M, Niknezhad SV, Forootanfar H, Ghasemi Y, Jafari E, Adeli-Sardou M, Amirsadeghi A, Ameri A. Development of Ag NPs/allantoin loaded PCL/GEL electrospun nanofibers for topical wound treatment. J Biomater Appl 2023; 38:692-706. [PMID: 37905355 DOI: 10.1177/08853282231212605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
In the present study, the allantoin and silver nanoparticle (Ag NPs) loaded poly caprolactone/gelatin (PCL/GEL) nanofibers produced using electrospinning technique and their cyto-compatibility and wound healing activity were evaluated in vitro and in vivo. The SEM imaging revealed diameters of 278.8 ± 10 and 240.6 ± 12 nm for PCL/GEL/Ag NPs and PCL/GEL/Ag NPs/allantoin scaffolds. The Ag NPs entrapment into scaffolds was evaluated by FTIR analysis and EDX mapping. Both scaffolds containing Ag NPs and Ag NPs/allantoin exhibited valuable wound healing activity in Wistar rat animal model. The profound granulation tissue formation, high collagen deposition in coordination with low level of edema and inflammatory cells in Ag NPs/allantoin loaded scaffolds resulted in complete and mature re-epithelialization in giving the healing score (12 out of 12) equal to positive control group to the wounds treated with these scaffolds. It was concluded that the Ag NPs/allantoin loaded scaffolds regarding to their good antibacterial activity and excellent wound healing activity could be introduced as new effective wound dressing materials.
Collapse
Affiliation(s)
- Mohsen Doostmohammadi
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Seyyed Vahid Niknezhad
- Department of Cell and Tissue Biology, Program in Craniofacial Biology, University of California, San Francisco, CA, USA
| | - Hamid Forootanfar
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Younes Ghasemi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elham Jafari
- Department of Pathology, Pathology and Stem Cell Research Center, School of Medicine, Kerman University of Medical Science, Kerman, Iran
| | - Mahboubeh Adeli-Sardou
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Armin Amirsadeghi
- Department of Cell and Tissue Biology, Program in Craniofacial Biology, University of California, San Francisco, CA, USA
| | - Alieh Ameri
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
7
|
Liu J, Xie X, Wang T, Chen H, Fu Y, Cheng X, Wu J, Li G, Liu C, Liimatainen H, Zheng Z, Wang X, Kaplan DL. Promotion of Wound Healing Using Nanoporous Silk Fibroin Sponges. ACS APPLIED MATERIALS & INTERFACES 2023; 15:12696-12707. [PMID: 36855948 DOI: 10.1021/acsami.2c20274] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Wound dressings are important for wound repair. The morphology of the biomaterials used in these dressings, and in particular, the pore structure affects tissue regeneration by facilitating attachment and proliferation of cells due to the hierarchical multiscale, water absorbance, and nutrient transport. In the present study, silk fibroin (SF) sponges with walls containing nanopores (SFNS) were prepared from SF nanoparticles generated during the autoclaving of SF solutions, followed by leaching the SF nanoparticles from the freeze-dried sponges of SF. The nano/microporous structure, biofluid absorbance, and porosity of the SF sponges with and without nanopores were characterized. In vitro cell proliferation, in vivo biocompatibility, and wound healing were evaluated with the sponges. The results demonstrated that SFNS had significantly increased porosity and water permeability, as well as cell attachment and proliferation when compared with SF sponges without the nanopores (SFS). Wound dressings were assessed in a rat skin wound model, and SFNS was superior to SFS in accelerating wound healing, supported by vascularization, deposition of collagen, and increased epidermal thickness over 21 days. Hence, such a dressing material with a hierarchical multiscale pore structure could promote cell migration, vascularization, and tissue regeneration independently without adding any growth factor, which would offer a new strategy to design and engineer better-performed wound dressing.
Collapse
Affiliation(s)
- Jian Liu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
- China National Textile and Apparel Council Key Laboratory for Silk Functional Materials and Technology, Soochow University, Suzhou 215123, China
| | - Xusheng Xie
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Tao Wang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Hao Chen
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Yuhang Fu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Xinyu Cheng
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Jianbing Wu
- College of Textile, Garment and Design, Changshu Institute of Technology, Suzhou 215500, China
| | - Gang Li
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Chenming Liu
- Fiber and Particle Engineering Research Unit, University of Oulu, Oulu 90014, Finland
| | - Henrikki Liimatainen
- Fiber and Particle Engineering Research Unit, University of Oulu, Oulu 90014, Finland
| | - Zhaozhu Zheng
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
- Simatech Incorporation, Suzhou 215123, China
| | - Xiaoqin Wang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| |
Collapse
|
8
|
Halperin-Sternfeld M, Pokhojaev A, Ghosh M, Rachmiel D, Kannan R, Grinberg I, Asher M, Aviv M, Ma PX, Binderman I, Sarig R, Adler-Abramovich L. Immunomodulatory fibrous hyaluronic acid-Fmoc-diphenylalanine-based hydrogel induces bone regeneration. J Clin Periodontol 2023; 50:200-219. [PMID: 36110056 PMCID: PMC10086858 DOI: 10.1111/jcpe.13725] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 08/30/2022] [Accepted: 09/09/2022] [Indexed: 01/18/2023]
Abstract
AIM To investigate the potential of an ultrashort aromatic peptide hydrogelator integrated with hyaluronic acid (HA) to serve as a scaffold for bone regeneration. MATERIALS AND METHODS Fluorenylmethyloxycarbonyl-diphenylalanine (FmocFF)/HA hydrogel was prepared and characterized using microscopy and rheology. Osteogenic differentiation of MC3T3-E1 preosteoblasts was investigated using Alizarin red, alkaline phosphatase and calcium deposition assays. In vivo, 5-mm-diameter calvarial critical-sized defects were prepared in 20 Sprague-Dawley rats and filled with either FmocFF/HA hydrogel, deproteinized bovine bone mineral, FmocFF/Alginate hydrogel or left unfilled. Eight weeks after implantation, histology and micro-computed tomography analyses were performed. Immunohistochemistry was performed in six rats to assess the hydrogel's immunomodulatory effect. RESULTS A nanofibrous FmocFF/HA hydrogel with a high storage modulus of 46 KPa was prepared. It supported osteogenic differentiation of MC3T3-E1 preosteoblasts and facilitated calcium deposition. In vivo, the hydrogel implantation resulted in approximately 93% bone restoration. It induced bone deposition not only around the margins, but also generated bony islets along the defect. Elongated M2 macrophages lining at the periosteum-hydrogel interface were observed 1 week after implantation. After 3 weeks, these macrophages were dispersed through the regenerating tissue surrounding the newly formed bone. CONCLUSIONS FmocFF/HA hydrogel can serve as a cell-free, biomimetic, immunomodulatory scaffold for bone regeneration.
Collapse
Affiliation(s)
- Michal Halperin-Sternfeld
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel.,The Center for the Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel
| | - Ariel Pokhojaev
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Moumita Ghosh
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel.,The Center for the Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel.,Department of Chemistry, Techno India University, Kolkata, West Bengal, India
| | - Dana Rachmiel
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel.,The Center for the Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel
| | - Raha Kannan
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Itzhak Grinberg
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel.,The Center for the Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel
| | - Moshe Asher
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Moran Aviv
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel.,The Center for the Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel.,School of Mechanical Engineering, Afeka Tel Aviv Academic College of Engineering, Tel Aviv, Israel
| | - Peter X Ma
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Itzhak Binderman
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Rachel Sarig
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,The Dan David Center for Human Evolution and Biohistory Research, Tel Aviv University, Tel Aviv, Israel
| | - Lihi Adler-Abramovich
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel.,The Center for the Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
9
|
Qu HL, Sun LJ, Li X, Liu F, Sun HH, He XT, Gan D, Yin Y, Tian BM, Chen FM, Wu RX. Long non-coding RNA AC018926.2 regulates palmitic acid exposure-compromised osteogenic potential of periodontal ligament stem cells via the ITGA2/FAK/AKT pathway. Cell Prolif 2023:e13411. [PMID: 36720715 PMCID: PMC10392068 DOI: 10.1111/cpr.13411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/06/2023] [Accepted: 01/18/2023] [Indexed: 02/02/2023] Open
Abstract
Although obesity has been proposed as a risk factor for periodontitis, the influence of excessive fat accumulation on the development of periodontitis and periodontal recovery from disease remains largely unknown. This study investigated the cellular response of periodontal ligament stem cells (PDLSCs) to elevated levels of a specific fatty acid, namely, palmitic acid (PA). The mechanism by which PA exposure compromises the osteogenic potential of cells was also explored. It was found that exposure of PDLSCs to abundant PA led to decreased cell osteogenic differentiation. Given that long non-coding RNAs (lncRNAs) play a key role in the stem cell response to adverse environmental stimuli, we screened the lncRNAs that were differentially expressed in PDLSCs following PA exposure using lncRNA microarray analysis, and AC018926.2 was identified as the lncRNA that was most sensitive to PA. Next, gain/loss-of-function studies illustrated that AC018926.2 was an important regulator in PA-mediated osteogenic differentiation of PDLSCs. Mechanistically, AC018926.2 upregulated integrin α2 (ITGA2) expression and therefore activated ITGA2/FAK/AKT signalling. Further functional studies revealed that inactivation of ITGA2/FAK/AKT signalling by silencing ITGA2 counteracted the pro-osteogenic effect induced by AC018926.2 overexpression. Moreover, the results of bioinformatics analysis and RNA immunoprecipitation assay suggested that AC018926.2 might transcriptionally regulate ITGA2 expression by binding to PARP1 protein. Our data suggest that AC018926.2 may serve as a therapeutic target for the management of periodontitis in obese patients.
Collapse
Affiliation(s)
- Hong-Lei Qu
- Department of Periodontology, State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Li-Juan Sun
- Department of Periodontology, State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Xuan Li
- Department of Periodontology, State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Fen Liu
- Department of Pediatric Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Hai-Hua Sun
- Department of General Dentistry and Emergency, State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Xiao-Tao He
- Department of Periodontology, State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Dian Gan
- Department of Periodontology, State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Yuan Yin
- Department of Periodontology, State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Bei-Min Tian
- Department of Periodontology, State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Fa-Ming Chen
- Department of Periodontology, State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Rui-Xin Wu
- Department of Periodontology, State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| |
Collapse
|
10
|
The effect of external magnetic field on osteogenic and antimicrobial behaviour of surface-functionalized custom titanium chamber with iron nanoparticles. A preliminary research. Odontology 2022:10.1007/s10266-022-00769-7. [DOI: 10.1007/s10266-022-00769-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 11/15/2022] [Indexed: 11/30/2022]
|
11
|
Extracellular matrix-mimicking nanofibrous chitosan microspheres as cell micro-ark for tissue engineering. Carbohydr Polym 2022; 292:119693. [PMID: 35725181 DOI: 10.1016/j.carbpol.2022.119693] [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: 01/22/2022] [Revised: 05/23/2022] [Accepted: 06/01/2022] [Indexed: 11/20/2022]
Abstract
In the present study, extracellular matrix (ECM)-mimicking nanofibrous chitosan microspheres (NCM) were developed via thermal induction of chitosan molecular chain from alkaline/urea aqueous solution. The regeneration of NCM from chitosan was proved to be physical process. The morphology of NCM could be precisely controlled by adjusting the initial solution concentration and the reaction temperature. The NCM possessed desirable in vitro/vivo biocompatibility and biodegradability. The excellent cell adhesion capability of NCM facilitated the formation of large-sized 3D geometric constructs in vitro. The NCM promoted in vitro osteogenic differentiation of rat bone marrow stem cells (rMSCs). Finally, pre-differentiated rMSCs-NCM constructs obviously enhanced in vivo bone healing of rat calvarial defects. This work opened up a new avenue for the construction of chitosan microspheres with ECM-like nanofibrous structure, indicated the great potential of the NCM as micro-Noah's Ark for stem cells to anchor, proliferate, and pre-differentiate for tissue engineering.
Collapse
|
12
|
Chakraborty J, Roy S, Ghosh S. 3D Printed Hydroxyapatite Promotes Congruent Bone Ingrowth in Rat Load Bearing Defects. Biomed Mater 2022; 17. [PMID: 35381582 DOI: 10.1088/1748-605x/ac6471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/05/2022] [Indexed: 11/11/2022]
Abstract
3D porous hydroxyapatite scaffolds produced by conventional foaming processes have limited control over the scaffold's pore size, geometry, and pore interconnectivity. In addition, random internal pore architecture often results in limited clinical success. Imitating the intricate 3D architecture and the functional dynamics of skeletal deformations is a difficult task, highlighting the necessity for a custom-made, on-demand tissue replacement, for which 3D printing is a potential solution. To combat these problems, here we report the ability of 3D printed hydroxyapatite scaffolds for in vivo bone regeneration in a rat tibial defect model. Rapid prototyping using the direct-write technique to fabricate 25 mm2 hydroxyapatite scaffolds were employed for precise control over geometry (both external and internal) and scaffold chemistry. Bone ingrowth was determined using histomorphometry and a novel micro-CT image analysis. Substantial bone ingrowth was observed in implants that filled the defect site. Further validating this quantitatively by micro-CT, the Bone mineral density of the implant at the defect site was 1024 mgHA/ccm, which was approximately 61.5% more than the bone mineral density found with the sham control at the defect site. In addition, no evident immunoinflammatory response was observed in the H&E micrographs. Interestingly, the present study showed a positive correlation with the outcomes obtained in our previous in vitro study. Overall, the results suggest that 3D printed hydroxyapatite scaffolds developed in this study offer a suitable matrix for rendering patient-specific and defect-specific bone formation and warrant further testing for clinical application.
Collapse
Affiliation(s)
- Juhi Chakraborty
- Indian Institute of Technology Delhi, Hauz Khas, New Delhi, Delhi, 110016, INDIA
| | - Subhadeep Roy
- Indian Institute of Technology Delhi, Hauz Khas, New Delhi, Delhi, 110016, INDIA
| | - Sourabh Ghosh
- Department of Textile Technology, Indian Institute of Technology Delhi, TX-110C, Hauz Khas, New Delhi, 110016, New Delhi, 110016, INDIA
| |
Collapse
|
13
|
Pathmanapan S, Sekar M, Pandurangan AK, Anandasadagopan SK. Fabrication of Mesoporous Silica Nanoparticle-Incorporated Coaxial Nanofiber for Evaluating the In Vitro Osteogenic Potential. Appl Biochem Biotechnol 2021; 194:302-322. [PMID: 34762271 DOI: 10.1007/s12010-021-03741-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/21/2021] [Indexed: 11/25/2022]
Abstract
The most important role of tissue engineering is to develop a biomaterial with a property that mimics the extracellular matrix (ECM) by enhancing the lineage-specific proliferation and differentiation with favorable regeneration property to aid in new tissue formation. Thus, to develop an ideal scaffold for bone repair, we have fabricated a composite nanofiber by the coaxial electrospinning technique. The coaxial electrospun nanofiber contains the core layer, consisting of polyvinyl alcohol (PVA) blended with oregano extract and mesoporous silica nanoparticles (PVA-OE-MSNPs), and the shell layer, consisting of poly-ε-caprolactone blended with collagen and hydroxyapatite (PCL-collagen-HAP). We evaluated the physicochemical properties of the nanofibers using X-ray diffraction (XRD), thermogravimetric analysis (TGA), scanning electron microscopy (SEM), and Fourier transform infrared spectroscopy (FTIR). In vitro biocompatibility, cell adhesion, cell viability, and osteogenic potential were evaluated by 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenlytetrazolium bromide (MTT), calcein AM, and alkaline phosphatase (ALP) activity and Alizarin Red staining in NIH 3T3/MG-63 cells. The results showed that the nanoparticle-incorporated coaxial nanofiber was observed with bead-free, continuous, and uniform fiber morphology with a mean diameter in the range of 310 ± 125 nm. From the biochemical studies, it is observed that the incorporation of nanofiber with HAP and MSNPs shows good swelling property with ideal porosity, biodegradation, and enhanced biomineralization property. In vitro results showed that the scaffolds with nanoparticles have higher cell adhesion, cell viability, ALP activity, and mineralization potential. Thus, the fabricated nanofiber could be an appropriate implantable biomaterial for bone tissue engineering.
Collapse
Affiliation(s)
- Srinivetha Pathmanapan
- Biochemistry and Biotechnology Laboratory, Central Leather Research Institute, Council of Scientific and Industrial Research (CSIR), Adyar, Chennai, 600020, India
- Department of Leather Technology, Housed at CSIR-Central Leather Research Institute, Alagappa College of Technology, Anna University, Chennai, 600020, India
| | - Mythrehi Sekar
- Biochemistry and Biotechnology Laboratory, Central Leather Research Institute, Council of Scientific and Industrial Research (CSIR), Adyar, Chennai, 600020, India
| | - Ashok Kumar Pandurangan
- School of Life Science, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Suresh Kumar Anandasadagopan
- Biochemistry and Biotechnology Laboratory, Central Leather Research Institute, Council of Scientific and Industrial Research (CSIR), Adyar, Chennai, 600020, India.
- Academy of Scientific and Innovative Research (AcSIR), CSIR-CLRI Campus, Chennai, 600020, India.
| |
Collapse
|
14
|
Zhang J, Zhu SS, Jiang N. Effect of micro/nanoscaled Ti phosphate/Ti oxide hybrid coating on the osseointegration of Ti implants. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2021; 39:531-539. [PMID: 34636200 DOI: 10.7518/hxkq.2021.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
OBJECTIVES This study was performed to fabricate a bionic coating with titanium (Ti) phosphate to promote the osseointegration of Ti substrate implants. METHODS Phosphorylated micro/nanocoating was prepared on the surface of pure titanium (i.e., TiP-Ti) by hydrothermal process under special pressure, and the untreated smooth pure titanium (cp-Ti) was selected as the control. To evaluate the characteristics of the coating surface, scanning electron microscopy, X-ray diffraction, atomic force microscopy, and contact-angle measurement were performed. In addition, the effects of TiP-Ti on the proliferation, adhesion, and differentiation of rat bone marrow mesenchymal stem cells (BMSCs) were investigated by using in vitro cytology. Finally, TiP-Ti implants were implanted into the rat tibia, and the effect of TiP-Ti on the osseointegration in the host was evaluated after 12 weeks. RESULTS The TiP-Ti surface presented a bionic structure with coexisting nanoscale 3D spatial structure and microscale pores. In vitro experiments showed that the BMSCs had enhanced adhesion, proliferation, and osteogenic differentiation on the TiP-Ti surface. Furthermore, in vivo, TiP-Ti showed considerably stronger osseointegration compared with pure titanium, and the ultimate shear strength and maximum pushing force were significantly improved. CONCLUSIONS A bionic structure with TiP-Ti micro/nanoscale coating was successfully fabricated, indicating a promising method for modifying the surface of implants.
Collapse
Affiliation(s)
- Jie Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Orthognathic and Temporomandibular Joint Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Song-Song Zhu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Orthognathic and Temporomandibular Joint Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Nan Jiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Orthognathic and Temporomandibular Joint Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
15
|
Zhang Z, Gong L, Li M, Wei G, Liu Y. The osteogenic differentiation of human bone marrow stromal cells induced by nanofiber scaffolds using bioinformatics. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166245. [PMID: 34391896 DOI: 10.1016/j.bbadis.2021.166245] [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: 04/17/2021] [Revised: 07/26/2021] [Accepted: 08/09/2021] [Indexed: 11/16/2022]
Abstract
This article aims to investigate the mechanism of behaviors of human bone marrow stromal cells (hBMSCs) affected by scaffold structure combining Monte Carlo feature selection (MFCS), incremental feature selection (IFS) and support vector machine (SVM). The specific differentially expressed genes (DEGs) of hBMSCs cultured on nanofiber (NF) scaffolds and freeform fabrication (FFF) scaffolds were obtained. Key genes were screened from common genes between osteogenic DEGs and NF specific DEGs with MFCS, IFS and SVM. The results demonstrated that NF scaffolds induced hBMSCs to express more genes related to osteogenic differentiation. Finally, 16 key genes were identified among the common genes. The common genes were significantly enriched in Rap1 signaling pathway, extracellular matrix and ossification. The results in this study suggested that the gene expression of hBMSCs was sensitive to NF scaffolds and FFF scaffolds, and the osteogenic differentiation of hBMSCs could be enhanced by NF scaffolds.
Collapse
Affiliation(s)
- Zhenghai Zhang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Lulu Gong
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| | - Min Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Guoshuai Wei
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yan Liu
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| |
Collapse
|
16
|
Lin Z, Nica C, Sculean A, Asparuhova MB. Positive Effects of Three-Dimensional Collagen-Based Matrices on the Behavior of Osteoprogenitors. Front Bioeng Biotechnol 2021; 9:708830. [PMID: 34368101 PMCID: PMC8334008 DOI: 10.3389/fbioe.2021.708830] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/05/2021] [Indexed: 12/22/2022] Open
Abstract
Recent research has demonstrated that reinforced three-dimensional (3D) collagen matrices can provide a stable scaffold for restoring the lost volume of a deficient alveolar bone. In the present study, we aimed to comparatively investigate the migratory, adhesive, proliferative, and differentiation potential of mesenchymal stromal ST2 and pre-osteoblastic MC3T3-E1 cells in response to four 3D collagen-based matrices. Dried acellular dermal matrix (DADM), hydrated acellular dermal matrix (HADM), non-crosslinked collagen matrix (NCM), and crosslinked collagen matrix (CCM) did all enhance the motility of the osteoprogenitor cells. Compared to DADM and NCM, HADM and CCM triggered stronger migratory response. While cells grown on DADM and NCM demonstrated proliferative rates comparable to control cells grown in the absence of a biomaterial, cells grown on HADM and CCM proliferated significantly faster. The pro-proliferative effects of the two matrices were supported by upregulated expression of genes regulating cell division. Increased expression of genes encoding the adhesive molecules fibronectin, vinculin, CD44 antigen, and the intracellular adhesive molecule-1 was detected in cells grown on each of the scaffolds, suggesting excellent adhesive properties of the investigated biomaterials. In contrast to genes encoding the bone matrix proteins collagen type I (Col1a1) and osteopontin (Spp1) induced by all matrices, the expression of the osteogenic differentiation markers Runx2, Alpl, Dlx5, Ibsp, Bglap2, and Phex was significantly increased in cells grown on HADM and CCM only. Short/clinically relevant pre-coating of the 3D biomaterials with enamel matrix derivative (EMD) or recombinant bone morphogenetic protein-2 (rBMP-2) significantly boosted the osteogenic differentiation of both osteoprogenitor lines on all matrices, including DADM and NCM, indicating that EMD and BMP-2 retained their biological activity after being released from the matrices. Whereas EMD triggered the expression of all osteogenesis-related genes, rBMP-2 upregulated early, intermediate, and late osteogenic differentiation markers except for Col1a1 and Spp1. Altogether, our results support favorable influence of HADM and CCM on the recruitment, growth, and osteogenic differentiation of the osteoprogenitor cell types. Furthermore, our data strongly support the biofunctionalization of the collagen-based matrices with EMD or rBMP-2 as a potential treatment modality for bone defects in the clinical practice.
Collapse
Affiliation(s)
- Zhikai Lin
- Laboratory of Oral Cell Biology, Dental Research Center, School of Dental Medicine, University of Bern, Bern, Switzerland.,Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland.,Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Cristina Nica
- Laboratory of Oral Cell Biology, Dental Research Center, School of Dental Medicine, University of Bern, Bern, Switzerland.,Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland
| | - Anton Sculean
- Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland
| | - Maria B Asparuhova
- Laboratory of Oral Cell Biology, Dental Research Center, School of Dental Medicine, University of Bern, Bern, Switzerland.,Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland
| |
Collapse
|
17
|
Li Z, Du T, Ruan C, Niu X. Bioinspired mineralized collagen scaffolds for bone tissue engineering. Bioact Mater 2021; 6:1491-1511. [PMID: 33294729 PMCID: PMC7680706 DOI: 10.1016/j.bioactmat.2020.11.004] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/20/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022] Open
Abstract
Successful regeneration of large segmental bone defects remains a major challenge in clinical orthopedics, thus it is of important significance to fabricate a suitable alternative material to stimulate bone regeneration. Due to their excellent biocompatibility, sufficient mechanical strength, and similar structure and composition of natural bone, the mineralized collagen scaffolds (MCSs) have been increasingly used as bone substitutes via tissue engineering approaches. Herein, we thoroughly summarize the state of the art of MCSs as tissue-engineered scaffolds for acceleration of bone repair, including their fabrication methods, critical factors for osteogenesis regulation, current opportunities and challenges in the future. First, the current fabrication methods for MCSs, mainly including direct mineral composite, in-situ mineralization and 3D printing techniques, have been proposed to improve their biomimetic physical structures in this review. Meanwhile, three aspects of physical (mechanics and morphology), biological (cells and growth factors) and chemical (composition and cross-linking) cues are described as the critical factors for regulating the osteogenic feature of MCSs. Finally, the opportunities and challenges associated with MCSs as bone tissue-engineered scaffolds are also discussed to point out the future directions for building the next generation of MCSs that should be endowed with satisfactorily mimetic structures and appropriately biological characters for bone regeneration.
Collapse
Affiliation(s)
- Zhengwei Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Tianming Du
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
| | - Changshun Ruan
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Xufeng Niu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100083, PR China
- Research Institute of Beihang University in Shenzhen, Shenzhen, 518057, PR China
| |
Collapse
|
18
|
Manipulating Air-Gap Electrospinning to Create Aligned Polymer Nanofiber-Wrapped Glass Microfibers for Cortical Bone Tissue Engineering. Bioengineering (Basel) 2020; 7:bioengineering7040165. [PMID: 33419239 PMCID: PMC7766430 DOI: 10.3390/bioengineering7040165] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/10/2020] [Accepted: 12/15/2020] [Indexed: 11/17/2022] Open
Abstract
Osteons are the repeating unit throughout cortical bone, consisting of canals filled with blood and nerve vessels surrounded by concentric lamella of hydroxyapatite-containing collagen fibers, providing mechanical strength. Creating a biodegradable scaffold that mimics the osteon structure is crucial for optimizing cellular infiltration and ultimately the replacement of the scaffold with native cortical bone. In this study, a modified air-gap electrospinning setup was exploited to continuously wrap highly aligned polycaprolactone polymer nanofibers around individual 1393 bioactive glass microfibers, resulting in a synthetic structure similar to osteons. By varying the parameters of the device, scaffolds with polymer fibers wrapped at angles between 5-20° to the glass fiber were chosen. The scaffold indicated increased cell migration by demonstrating unidirectional cell orientation along the fibers, similar to recent work regarding aligned nerve and muscle regeneration. The wrapping decreased the porosity from 90% to 80%, which was sufficient for glass conversion through ion exchange validated by inductively coupled plasma. Scaffold degradation was not cytotoxic. Encapsulating the glass with polymer nanofibers caused viscoelastic deformation during three-point bending, preventing typical brittle glass fracture, while maintaining cell migration. This scaffold design structurally mimics the osteon, with the intent to replace its material compositions for better regeneration.
Collapse
|
19
|
Application of platelet-rich plasma (PRP) improves self-renewal of human spermatogonial stem cells in two-dimensional and three-dimensional culture systems. Acta Histochem 2020; 122:151627. [PMID: 33002788 DOI: 10.1016/j.acthis.2020.151627] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/17/2020] [Accepted: 09/14/2020] [Indexed: 12/20/2022]
Abstract
Spermatogonial stem cells (SSCs) are very sensitive to chemotherapy and radiotherapy, so male infertility is a great challenge for prepubertal cancer survivors. Cryoconservation of testicular cells before cancer treatment can preserve SSCs from treatment side effects. Different two-dimensional (2D) and three-dimensional (3D) culture systems of SSCs have been used in many species as a useful technique to in vitro spermatogenesis. We evaluated the proliferation of SSCs in 2D and 3D culture systems of platelet-rich plasma (PRP). testicular cells of four brain-dead patients cultivated in 2D pre-culture system, characterization of SSCs performed by RT-PCR, flow cytometry, immunocytochemistry and their functionality assessed by xenotransplantation to azoospermia mice. PRP prepared and dosimetry carried out to determine the optimized dose of PRP. After preparation of PRP scaffold, cytotoxic and histological evaluation performed and SSCs cultivated into three groups: control, 2D culture by optimized dose of PRP and PRP scaffold. The diameter and number of colonies measured and relative expression of GFRa1 and c-KIT evaluated by real-time PCR. Results indicated the expression of PLZF, VASA, OCT4, GFRa1 and vimentin in colonies after 2D pre-culture, xenotransplantation demonstrated proliferated SSCs have proper functionality to homing in mouse testes. The relative expression of c-KIT showed a significant increase as compared to the control group (*: p < 0.05) in PRP- 2D group, expression of GFRa1 and c-KIT in PRP scaffold group revealed a significant increase as compared to other groups (***: p < 0.001). The number and diameter of colonies in the PRP-2D group showed a considerable increase (p < 0.01) as compared to the control group. In PRP- scaffold group, a significant increase (p < 0.01) was seen only in the number of colonies related to the control group. Our results suggested that PRP scaffold can reconstruct a suitable structure to the in vitro proliferation of SSCs.
Collapse
|
20
|
Kumar P, Saini M, Dehiya BS, Sindhu A, Kumar V, Kumar R, Lamberti L, Pruncu CI, Thakur R. Comprehensive Survey on Nanobiomaterials for Bone Tissue Engineering Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E2019. [PMID: 33066127 PMCID: PMC7601994 DOI: 10.3390/nano10102019] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023]
Abstract
One of the most important ideas ever produced by the application of materials science to the medical field is the notion of biomaterials. The nanostructured biomaterials play a crucial role in the development of new treatment strategies including not only the replacement of tissues and organs, but also repair and regeneration. They are designed to interact with damaged or injured tissues to induce regeneration, or as a forest for the production of laboratory tissues, so they must be micro-environmentally sensitive. The existing materials have many limitations, including impaired cell attachment, proliferation, and toxicity. Nanotechnology may open new avenues to bone tissue engineering by forming new assemblies similar in size and shape to the existing hierarchical bone structure. Organic and inorganic nanobiomaterials are increasingly used for bone tissue engineering applications because they may allow to overcome some of the current restrictions entailed by bone regeneration methods. This review covers the applications of different organic and inorganic nanobiomaterials in the field of hard tissue engineering.
Collapse
Affiliation(s)
- Pawan Kumar
- Department of Materials Science and Nanotechnology, Deenbandhu Chhotu Ram University of Science and Technology, Murthal 131039, India; (M.S.); (B.S.D.)
| | - Meenu Saini
- Department of Materials Science and Nanotechnology, Deenbandhu Chhotu Ram University of Science and Technology, Murthal 131039, India; (M.S.); (B.S.D.)
| | - Brijnandan S. Dehiya
- Department of Materials Science and Nanotechnology, Deenbandhu Chhotu Ram University of Science and Technology, Murthal 131039, India; (M.S.); (B.S.D.)
| | - Anil Sindhu
- Department of Biotechnology, Deenbandhu Chhotu Ram University of Science and Technology, Murthal 131039, India;
| | - Vinod Kumar
- Department of Bio and Nanotechnology, Guru Jambheshwar University of Science and Technology, Hisar 125001, India; (V.K.); (R.T.)
| | - Ravinder Kumar
- School of Mechanical Engineering, Lovely Professional University, Phagwara 144411, India
| | - Luciano Lamberti
- Dipartimento di Meccanica, Matematica e Management, Politecnico di Bari, 70125 Bari, Italy;
| | - Catalin I. Pruncu
- Department of Design, Manufacturing & Engineering Management, University of Strathclyde, Glasgow G1 1XJ, UK
- Department of Mechanical Engineering, Imperial College London, London SW7 2AZ, UK
| | - Rajesh Thakur
- Department of Bio and Nanotechnology, Guru Jambheshwar University of Science and Technology, Hisar 125001, India; (V.K.); (R.T.)
| |
Collapse
|
21
|
Stutz C, Strub M, Clauss F, Huck O, Schulz G, Gegout H, Benkirane-Jessel N, Bornert F, Kuchler-Bopp S. A New Polycaprolactone-Based Biomembrane Functionalized with BMP-2 and Stem Cells Improves Maxillary Bone Regeneration. NANOMATERIALS 2020; 10:nano10091774. [PMID: 32911737 PMCID: PMC7558050 DOI: 10.3390/nano10091774] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/05/2020] [Accepted: 09/06/2020] [Indexed: 12/12/2022]
Abstract
Oral diseases have an impact on the general condition and quality of life of patients. After a dento-alveolar trauma, a tooth extraction, or, in the case of some genetic skeletal diseases, a maxillary bone defect, can be observed, leading to the impossibility of placing a dental implant for the restoration of masticatory function. Recently, bone neoformation was demonstrated after in vivo implantation of polycaprolactone (PCL) biomembranes functionalized with bone morphogenic protein 2 (BMP-2) and ibuprofen in a mouse maxillary bone lesion. In the present study, human bone marrow derived mesenchymal stem cells (hBM-MSCs) were added on BMP-2 functionalized PCL biomembranes and implanted in a maxillary bone lesion. Viability of hBM-MSCs on the biomembranes has been observed using the "LIVE/DEAD" viability test and scanning electron microscopy (SEM). Maxillary bone regeneration was observed for periods ranging from 90 to 150 days after implantation. Various imaging methods (histology, micro-CT) have demonstrated bone remodeling and filling of the lesion by neoformed bone tissue. The presence of mesenchymal stem cells and BMP-2 allows the acceleration of the bone remodeling process. These results are encouraging for the effectiveness and the clinical use of this new technology combining growth factors and mesenchymal stem cells derived from bone marrow in a bioresorbable membrane.
Collapse
Affiliation(s)
- Céline Stutz
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative NanoMedicine (RNM), FMTS, 67000 Strasbourg, France; (C.S.); (M.S.); (F.C.); (O.H.); (H.G.); (N.B.-J.); (F.B.)
| | - Marion Strub
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative NanoMedicine (RNM), FMTS, 67000 Strasbourg, France; (C.S.); (M.S.); (F.C.); (O.H.); (H.G.); (N.B.-J.); (F.B.)
- Faculté de Chirurgie Dentaire, Université de Strasbourg (UDS), 8 rue Ste Elisabeth, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaires, Pediatric Dentistry, Hôpitaux Universitaires de Strasbourg (HUS), 1 place de l’Hôpital, 67000 Strasbourg, France
| | - François Clauss
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative NanoMedicine (RNM), FMTS, 67000 Strasbourg, France; (C.S.); (M.S.); (F.C.); (O.H.); (H.G.); (N.B.-J.); (F.B.)
- Faculté de Chirurgie Dentaire, Université de Strasbourg (UDS), 8 rue Ste Elisabeth, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaires, Pediatric Dentistry, Hôpitaux Universitaires de Strasbourg (HUS), 1 place de l’Hôpital, 67000 Strasbourg, France
| | - Olivier Huck
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative NanoMedicine (RNM), FMTS, 67000 Strasbourg, France; (C.S.); (M.S.); (F.C.); (O.H.); (H.G.); (N.B.-J.); (F.B.)
- Faculté de Chirurgie Dentaire, Université de Strasbourg (UDS), 8 rue Ste Elisabeth, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaires, Periodontology, Hôpitaux Universitaires de Strasbourg (HUS), 1 place de l’Hôpital, 67000 Strasbourg, France
| | - Georg Schulz
- Core Facility Micro- and Nanotomography, Biomaterials Science Center (BMC), Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, 4123 Allschwil, Switzerland;
| | - Hervé Gegout
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative NanoMedicine (RNM), FMTS, 67000 Strasbourg, France; (C.S.); (M.S.); (F.C.); (O.H.); (H.G.); (N.B.-J.); (F.B.)
- Faculté de Chirurgie Dentaire, Université de Strasbourg (UDS), 8 rue Ste Elisabeth, 67000 Strasbourg, France
| | - Nadia Benkirane-Jessel
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative NanoMedicine (RNM), FMTS, 67000 Strasbourg, France; (C.S.); (M.S.); (F.C.); (O.H.); (H.G.); (N.B.-J.); (F.B.)
- Faculté de Chirurgie Dentaire, Université de Strasbourg (UDS), 8 rue Ste Elisabeth, 67000 Strasbourg, France
| | - Fabien Bornert
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative NanoMedicine (RNM), FMTS, 67000 Strasbourg, France; (C.S.); (M.S.); (F.C.); (O.H.); (H.G.); (N.B.-J.); (F.B.)
- Pôle de Médecine et Chirurgie Bucco-Dentaires, Pediatric Dentistry, Hôpitaux Universitaires de Strasbourg (HUS), 1 place de l’Hôpital, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaires, Oral Medicine and Oral Surgery, Hôpitaux Universitaires de Strasbourg (HUS), 1 place de l’Hôpital, 67000 Strasbourg, France
| | - Sabine Kuchler-Bopp
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative NanoMedicine (RNM), FMTS, 67000 Strasbourg, France; (C.S.); (M.S.); (F.C.); (O.H.); (H.G.); (N.B.-J.); (F.B.)
- Correspondence: ; Tel.: +33-619610523
| |
Collapse
|
22
|
Henn D, Chen K, Fischer K, Rauh A, Barrera JA, Kim YJ, Martin RA, Hannig M, Niedoba P, Reddy SK, Mao HQ, Kneser U, Gurtner GC, Sacks JM, Schmidt VJ. Tissue Engineering of Axially Vascularized Soft-Tissue Flaps with a Poly-(ɛ-Caprolactone) Nanofiber-Hydrogel Composite. Adv Wound Care (New Rochelle) 2020; 9:365-377. [PMID: 32587789 DOI: 10.1089/wound.2019.0975] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 12/18/2019] [Indexed: 11/12/2022] Open
Abstract
Objective: To develop a novel approach for tissue engineering of soft-tissue flaps suitable for free microsurgical transfer, using an injectable nanofiber hydrogel composite (NHC) vascularized by an arteriovenous (AV) loop. Approach: A rat AV loop model was used for tissue engineering of vascularized soft-tissue flaps. NHC or collagen-elastin (CE) scaffolds were implanted into isolation chambers together with an AV loop and explanted after 15 days. Saphenous veins were implanted into the scaffolds as controls. Neoangiogenesis, ultrastructure, and protein expression of SYNJ2BP, EPHA2, and FOXC1 were analyzed by immunohistochemistry and compared between the groups. Rheological properties were compared between the two scaffolds and native human adipose tissue. Results: A functional neovascularization was evident in NHC flaps with its amount being comparable with CE flaps. Scanning electron microscopy revealed a strong mononuclear cell infiltration along the nanofibers in NHC flaps and a trend toward higher fiber alignment compared with CE flaps. SYNJ2BP and EPHA2 expression in endothelial cells (ECs) was lower in NHC flaps compared with CE flaps, whereas FOXC1 expression was increased in NHC flaps. Compared with the stiffer CE flaps, the NHC flaps showed similar rheological properties to native human adipose tissue. Innovation: This is the first study to demonstrate the feasibility of tissue engineering of soft-tissue flaps with similar rheological properties as human fat, suitable for microsurgical transfer using an injectable nanofiber hydrogel composite. Conclusions: The injectable NHC scaffold is suitable for tissue engineering of axially vascularized soft-tissue flaps with a solid neovascularization, strong cellular infiltration, and biomechanical properties similar to human fat. Our data indicate that SYNJ2BP, EPHA2, and FOXC1 are involved in AV loop-associated angiogenesis and that the scaffold material has an impact on protein expression in ECs.
Collapse
Affiliation(s)
- Dominic Henn
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California
- Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen, Ruprecht-Karls-University of Heidelberg, Heidelberg, Germany
| | - Kellen Chen
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California
| | - Katharina Fischer
- Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen, Ruprecht-Karls-University of Heidelberg, Heidelberg, Germany
| | - Annika Rauh
- Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen, Ruprecht-Karls-University of Heidelberg, Heidelberg, Germany
| | - Janos A. Barrera
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California
| | - Yoo-Jin Kim
- Institute of Pathology, Kaiserslautern, Germany
| | - Russell A. Martin
- Department of Materials Science and Engineering, Whiting School of Engineering, and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
- Translational Tissue Engineering Center and Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Matthias Hannig
- Clinic of Operative Dentistry, Periodontology and Preventive Dentistry, Saarland University, Homburg, Germany
| | - Patricia Niedoba
- Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen, Ruprecht-Karls-University of Heidelberg, Heidelberg, Germany
| | - Sashank K. Reddy
- Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Hai-Quan Mao
- Department of Materials Science and Engineering, Whiting School of Engineering, and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
- Translational Tissue Engineering Center and Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ulrich Kneser
- Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen, Ruprecht-Karls-University of Heidelberg, Heidelberg, Germany
| | - Geoffrey C. Gurtner
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California
| | - Justin M. Sacks
- Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Volker J. Schmidt
- Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen, Ruprecht-Karls-University of Heidelberg, Heidelberg, Germany
- Department for Plastic and Breast Surgery, Zealand University Hospital Roskilde, Roskilde, Denmark
| |
Collapse
|
23
|
Nanofibrous spongy microspheres to deliver rabbit mesenchymal stem cells and anti-miR-199a to regenerate nucleus pulposus and prevent calcification. Biomaterials 2020; 256:120213. [PMID: 32736170 DOI: 10.1016/j.biomaterials.2020.120213] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/29/2020] [Accepted: 06/17/2020] [Indexed: 12/15/2022]
Abstract
Lower back pain is mainly caused by intervertebral disc degeneration, in which calcification is frequently involved. Here novel nanofibrous spongy microspheres (NF-SMS) are used to carry rabbit bone marrow mesenchymal stromal cells (MSCs) to regenerate nucleus pulposus tissues. NF-SMS are shown to significantly enhance the MSC seeding, proliferation and differentiation over control microcarriers. Furthermore, a hyperbranched polymer (HP) with negligible cytotoxicity and high microRNA (miRNAs) binding affinity is synthesized. The HP can complex with anti-miR-199a and self-assemble into "double shell" polyplexes which are able to achieve high transfection efficiency into MSCs. A double-emulsion technique is used to encapsulate these polyplexes in biodegradable nanospheres (NS) to enable sustained anti-miR-199 delivery. Our results demonstrate that MSC/HP-anti-miR-199a/NS/NF-SMS constructs can promote the nucleus pulposus (NP) phenotype and resist calcification in vitro and in a subcutaneous environment. Furthermore, injection of MSC/HP-anti-miR-199a/NS/NF-SMS can stay in place, produce functional extracellular matrix, maintain disc height and prevent intervertebral disc (IVD) calcification in a rabbit lumbar degeneration model.
Collapse
|
24
|
Schüttler KF, Bauhofer MW, Ketter V, Giese K, Eschbach DA, Yenigün M, Fuchs-Winkelmann S, Paletta JRJ. Direct incorporation of mesenchymal stem cells into a Nanofiber scaffold - in vitro and in vivo analysis. Sci Rep 2020; 10:9557. [PMID: 32533010 PMCID: PMC7293317 DOI: 10.1038/s41598-020-66281-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 05/13/2020] [Indexed: 11/08/2022] Open
Abstract
Bony defects are a common problem in musculoskeletal surgery. Replacement with autologous bone grafts is limited by availability of transplant material. Sterilized cancellous bone, while being osteoconductive, has limited osteoinductivity. Nanofiber scaffolds are currently used for several purposes due to their capability of imitating the extracellular matrix. Furthermore, they allow modification to provide functional properties. Previously we showed that electrospun nanofiber scaffolds can be used for bone tissue regeneration. While aiming to use the osteoinductive capacities of collagen type-I nanofibers we saw reduced scaffold pore sizes that limited cellular migration and thus colonization of the scaffolds. Aim of the present study was the incorporation of mesenchymal stem cells into the electrospinning process of a nanofiber scaffold to produce cell-seeded nanofiber scaffolds for bone replacement. After construction of a suitable spinning apparatus for simultaneous electrospinning and spraying with independently controllable spinning and spraying devices and extensive optimization of the spinning process, in vitro and in vivo evaluation of the resulting scaffolds was conducted. Stem cells isolated from rat femora were incorporated into PLLA (poly-l-lactide acid) and PLLA-collagen type-I nanofiber scaffolds (PLLA Col I Blend) via simultaneous electrospinning and -spraying. Metabolic activity, proliferation and osteoblastic differentiation were assessed in vitro. For in vivo evaluation scaffolds were implanted into critical size defects of the rat scull. After 4 weeks, animals were sacrificed and bone healing was analyzed using CT-scans, histological, immunhistochemical and fluorescence evaluation. Successful integration of mesenchymal stem cells into the scaffolds was achieved by iteration of spinning and spraying conditions regarding polymer solvent, spinning distance, the use of a liquid counter-electrode, electrode voltage and spinning duration. In vivo formation of bone tissue was achieved. Using a PLLA scaffold, comparable results for the cell-free and cell-seeded scaffolds were found, while the cell-seeded PLLA-collagen scaffolds showed significantly better bone formation when compared to the cell-free PLLA-collagen scaffolds. These results provide support for the future use of cell-seeded nanofiber scaffolds for large bony defects.
Collapse
Affiliation(s)
- Karl F Schüttler
- Center for Orthopedics and Trauma Surgery, University Hospital Giessen and Marburg, Location Marburg, Marburg, Germany
| | - Michael W Bauhofer
- Center for Orthopedics and Trauma Surgery, University Hospital Giessen and Marburg, Location Marburg, Marburg, Germany
| | - Vanessa Ketter
- Center for Orthopedics and Trauma Surgery, University Hospital Giessen and Marburg, Location Marburg, Marburg, Germany
| | - Katja Giese
- Center for Orthopedics and Trauma Surgery, University Hospital Giessen and Marburg, Location Marburg, Marburg, Germany
| | - Daphne A Eschbach
- Center for Orthopedics and Trauma Surgery, University Hospital Giessen and Marburg, Location Marburg, Marburg, Germany
| | - Mesut Yenigün
- Department of Neurology, University Hospital Giessen and Marburg, Location Giessen, Giessen, Germany
| | - Susanne Fuchs-Winkelmann
- Center for Orthopedics and Trauma Surgery, University Hospital Giessen and Marburg, Location Marburg, Marburg, Germany
| | - Jürgen R J Paletta
- Center for Orthopedics and Trauma Surgery, University Hospital Giessen and Marburg, Location Marburg, Marburg, Germany.
| |
Collapse
|
25
|
PDMS Nano-Modified Scaffolds for Improvement of Stem Cells Proliferation and Differentiation in Microfluidic Platform. NANOMATERIALS 2020; 10:nano10040668. [PMID: 32252384 PMCID: PMC7221996 DOI: 10.3390/nano10040668] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/14/2020] [Accepted: 03/31/2020] [Indexed: 02/07/2023]
Abstract
Microfluidics cell-based assays require strong cell-substrate adhesion for cell viability, proliferation, and differentiation. The intrinsic properties of PDMS, a commonly used polymer in microfluidics systems, regarding cell-substrate interactions have limited its application for microfluidics cell-based assays. Various attempts by previous researchers, such as chemical modification, plasma-treatment, and protein-coating of PDMS revealed some improvements. These strategies are often reversible, time-consuming, short-lived with either cell aggregates formation, not cost-effective as well as not user- and eco-friendly too. To address these challenges, cell-surface interaction has been tuned by the modification of PDMS doped with different biocompatible nanomaterials. Gold nanowires (AuNWs), superparamagnetic iron oxide nanoparticles (SPIONs), graphene oxide sheets (GO), and graphene quantum dot (GQD) have already been coupled to PDMS as an alternative biomaterial enabling easy and straightforward integration during microfluidic fabrication. The synthesized nanoparticles were characterized by corresponding methods. Physical cues of the nanostructured substrates such as Young’s modulus, surface roughness, and nanotopology have been carried out using atomic force microscopy (AFM). Initial biocompatibility assessment of the nanocomposites using human amniotic mesenchymal stem cells (hAMSCs) showed comparable cell viabilities among all nanostructured PDMS composites. Finally, osteogenic stem cell differentiation demonstrated an improved differentiation rate inside microfluidic devices. The results revealed that the presence of nanomaterials affected a 5- to 10-fold increase in surface roughness. In addition, the results showed enhancement of cell proliferation from 30% (pristine PDMS) to 85% (nano-modified scaffolds containing AuNWs and SPIONs), calcification from 60% (pristine PDMS) to 95% (PDMS/AuNWs), and cell surface marker expression from 40% in PDMS to 77% in SPION- and AuNWs-PDMS scaffolds at 14 day. Our results suggest that nanostructured composites have a very high potential for stem cell studies and future therapies.
Collapse
|
26
|
Yao Q, Liu Y, Pan Y, Miszuk JM, Sun H. One-pot porogen free method fabricated porous microsphere-aggregated 3D PCL scaffolds for bone tissue engineering. J Biomed Mater Res B Appl Biomater 2020; 108:2699-2710. [PMID: 32154997 DOI: 10.1002/jbm.b.34601] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/13/2020] [Accepted: 03/01/2020] [Indexed: 12/12/2022]
Abstract
Three-dimensional (3D) scaffolds with interconnected, hierarchically structured pores, and biomimetic nanostructures are desirable for tissue engineering, where preparation with a facile and biocompatible strategy remains challenging. In the present work, an innovative porous microspheres-aggregated 3D PCL scaffold with macropores, micropores, and nanofibrous-like structures was fabricated through a one-pot thermally induced phase separation (TIPS) method without the use of any porogen or specific instruments. Importantly, the porosity, pore size, and mechanical properties of our scaffolds were tailorable through tuning of the polymer concentration. Interestingly, the bioactivity of our 3D PCL scaffolds was significantly improved, as abundant apatite-like layers were formed on the 3D porous scaffolds, while no obvious apatite was observed on the 2D flat PCL film. Moreover, the high surface area attributed to the hierarchical macro/micro/nanostructure enabled our 3D porous scaffold to serve as a drug delivery depot for sustained release of both small molecule drug (phenamil) and protein (BMP2). In addition to sustained drug release, the hierarchical structure and high mechanical properties also contribute to significantly improving BMP2-induced osteogenic differentiation. In summary, we developed a novel PCL porous scaffold through a facile, one-pot TIPS method and demonstrated its promising potential application in large bone defect repair.
Collapse
Affiliation(s)
- Qingqing Yao
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Institute of Advanced Materials for Nano-Bio Applications, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Department of Biomedical Engineering, University of South Dakota, Sioux Falls, South Dakota, USA
| | - Yu Liu
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Institute of Advanced Materials for Nano-Bio Applications, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yining Pan
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Institute of Advanced Materials for Nano-Bio Applications, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jacob M Miszuk
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, South Dakota, USA
| | - Hongli Sun
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, South Dakota, USA
| |
Collapse
|
27
|
Yang N, Tan RP, Chan AHP, Lee BSL, Santos M, Hung J, Liao Y, Bilek MMM, Fei J, Wise SG, Bao S. Immobilized Macrophage Colony-Stimulating Factor (M-CSF) Regulates the Foreign Body Response to Implanted Materials. ACS Biomater Sci Eng 2020; 6:995-1007. [PMID: 33464851 DOI: 10.1021/acsbiomaterials.9b01887] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The functionality and durability of implanted biomaterials are often compromised by an exaggerated foreign body reaction (FBR). M1/M2 polarization of macrophages is a critical regulator of scaffold-induced FBR. Macrophage colony-stimulating factor (M-CSF), a hematopoietic growth factor, induces macrophages into an M2-like polarized state, leading to immunoregulation and promoting tissue repair. In the present study, we explored the immunomodulatory effects of surface bound M-CSF on poly-l-lactic acid (PLLA)-induced FBR. M-CSF was immobilized on the surface of PLLA via plasma immersion ion implantation (PIII). M-CSF functionalized PLLA, PLLA-only, and PLLA+PIII were assessed in an IL-1β luciferase reporter mouse to detect real-time levels of IL-1β expression, reflecting acute inflammation in vivo. Additionally, these different treated scaffolds were implanted subcutaneously into wild-type mice to explore the effect of M-CSF in polarization of M2-like macrophages (CD68+/CD206+), related cytokines (pro-inflammatory: IL-1β, TNF and MCP-1; anti-inflammatory: IL-10 and TGF-β), and angiogenesis (CD31) by immunofluorescent staining. Our data demonstrated that IL-1β activity in M-CSF functionalized scaffolds was ∼50% reduced compared to PLLA-only at day 1 (p < 0.01) and day 2 (p < 0.05) post-implantation. There were >2.6-fold more CD206+ macrophages in M-CSF functionalized PLLA compared to PLLA-only at day 7 (p < 0.001), along with higher levels of IL-10 at both day 7 (p < 0.05) and day 14 (p < 0.01), and TGF-β at day 3 (p < 0.05), day 7 (p < 0.05), and day 14 (p < 0.001). Lower levels of pro-inflammatory cytokines were also detected in M-CSF functionalized PLLA in the early phase of the immune response compared to PLLA-only: a ∼58% decrease at day 3 in IL-1β; a ∼91% decrease at day 3 and a ∼66% decrease at day 7 in TNF; and a ∼60% decrease at day 7 in MCP-1. Moreover, enhanced angiogenesis inside and on/near the scaffold was observed in M-CSF functionalized PLLA compared to PLLA-only at day 3 (p < 0.05) and day 7 (p < 0.05), respectively. Overall, M-CSF functionalized PLLA enhanced CD206+ macrophage polarization and angiogenesis, consistent with lower levels of pro-inflammatory cytokines and higher levels of anti-inflammatory cytokines in early stages of the host response, indicating potential immunoregulatory functions on the local environment.
Collapse
Affiliation(s)
- Nianji Yang
- Discipline of Pathology, University of Sydney, Sydney, New South Wales 2006, Australia.,Discipline of Physiology, University of Sydney, Sydney, New South Wales 2006, Australia.,The Heart Research Institute, Sydney, Australia.,Charles Perkins Centre, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Richard P Tan
- Discipline of Physiology, University of Sydney, Sydney, New South Wales 2006, Australia.,The Heart Research Institute, Sydney, Australia.,Charles Perkins Centre, University of Sydney, Sydney, New South Wales 2006, Australia
| | | | - Bob S L Lee
- Discipline of Physiology, University of Sydney, Sydney, New South Wales 2006, Australia.,The Heart Research Institute, Sydney, Australia.,Charles Perkins Centre, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Miguel Santos
- Discipline of Physiology, University of Sydney, Sydney, New South Wales 2006, Australia.,The Heart Research Institute, Sydney, Australia.,Charles Perkins Centre, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Juichien Hung
- Discipline of Physiology, University of Sydney, Sydney, New South Wales 2006, Australia.,The Heart Research Institute, Sydney, Australia.,Charles Perkins Centre, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Yun Liao
- Department of Pharmacy, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Marcela M M Bilek
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales 2006, Australia.,School of Physics, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Jian Fei
- School of Life Science and Technology, Shanghai Tongji University, Shanghai, China.,Research Centre for Model Organism, Shanghai, China
| | - Steven G Wise
- Discipline of Physiology, University of Sydney, Sydney, New South Wales 2006, Australia.,The Heart Research Institute, Sydney, Australia.,Charles Perkins Centre, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Shisan Bao
- Discipline of Pathology, University of Sydney, Sydney, New South Wales 2006, Australia.,Charles Perkins Centre, University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
28
|
KOZUMA W, KON K, KAWAKAMI S, BOBOTHIKE A, IIJIMA H, SHIOTA M, KASUGAI S. Osteoconductive potential of a hydroxyapatite fiber material with magnesium: In vitro and in vivo studies. Dent Mater J 2019; 38:771-778. [DOI: 10.4012/dmj.2018-333] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Wataru KOZUMA
- Department of Oral Implantology and Regenerative Dental Medicine, Tokyo Medical and Dental University
| | - Kazuhiro KON
- Department of Oral Implantology and Regenerative Dental Medicine, Tokyo Medical and Dental University
| | - Sawako KAWAKAMI
- Department of Oral Implantology and Regenerative Dental Medicine, Tokyo Medical and Dental University
| | - Aung BOBOTHIKE
- Department of Oral Implantology and Regenerative Dental Medicine, Tokyo Medical and Dental University
| | - Hajime IIJIMA
- Department of Oral Implantology and Regenerative Dental Medicine, Tokyo Medical and Dental University
| | - Makoto SHIOTA
- Department of Oral Implantology and Regenerative Dental Medicine, Tokyo Medical and Dental University
| | - Shohei KASUGAI
- Department of Oral Implantology and Regenerative Dental Medicine, Tokyo Medical and Dental University
| |
Collapse
|
29
|
Yahia S, Khalil IA, El-Sherbiny IM. Sandwich-Like Nanofibrous Scaffolds for Bone Tissue Regeneration. ACS APPLIED MATERIALS & INTERFACES 2019; 11:28610-28620. [PMID: 31328910 DOI: 10.1021/acsami.9b06359] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Advanced bone healing approaches included a wide range of biomaterials that mainly mimic the composition, structure, and properties of bone extracellular matrix with osteogenic activity. The present study aimed to develop a sandwich-like structure of electrospun nanofibers (NFs) based on polycaprolactone (PCL) and chitosan/polyethylene oxide (CS/PEO) composite to stimulate bone fracture healing. The morphology of the fabricated scaffolds was examined using scanning electron microscopy (SEM). Apatite deposition was evaluated using simulated body fluid (SBF). The physicochemical and mechanical properties of samples were analyzed by Fourier transform infrared, differential scanning calorimetry (DSC), thermogravimetric analysis (TGA), and universal testing machine. SEM images exhibited a porous three-dimensional structure with NF diameters of 514-4745 nm and 68-786 nm for PCL NFs layer and the sandwich-like NFs scaffolds, respectively. Deposition of apatite crystal on scaffolds started at week 2 followed by heavy deposition at week 8. This was confirmed by measuring the consumption of calcium and phosphorous ions from SBF. Thermal stability of scaffolds was confirmed using DSC and TGA. Moreover, the PCL NF layer in the middle of the developed sandwich structure reinforced the scaffolds with bear load up to 12.224 ± 1.12 MPa and Young's modulus of 17.53 ± 3.24 MPa. The scaffolds' porous structure enhanced both cell propagation and proliferation. Besides, the presence of CS in the outer NF layers of the scaffolds increased the hydrophilicity, as evidenced by the reduction of contact angle from 116.6 to 57.6°, which is essential for cell attachment. Cell viability study on mesenchymal stem cells proved the cytocompatibility of the fabricated scaffolds. Finally, in vivo mandibular bone defect rabbit model was used to confirm the regeneration of a new healthy bone within 28 days. In conclusion, the developed scaffolds could be a promising solution to stimulate bone regeneration.
Collapse
Affiliation(s)
- Sarah Yahia
- Nanomedicine Lab, Center of Materials Sciences (CMS) , Zewail City of Science and Technology , 6th of October, Giza 12578 , Egypt
| | - Islam A Khalil
- Nanomedicine Lab, Center of Materials Sciences (CMS) , Zewail City of Science and Technology , 6th of October, Giza 12578 , Egypt
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy and Drug Manufacturing , Misr University of Science and Technology (MUST) , 6th of October, Giza 12566 , Egypt
| | - Ibrahim M El-Sherbiny
- Nanomedicine Lab, Center of Materials Sciences (CMS) , Zewail City of Science and Technology , 6th of October, Giza 12578 , Egypt
| |
Collapse
|
30
|
Richbourg NR, Peppas NA, Sikavitsas VI. Tuning the biomimetic behavior of scaffolds for regenerative medicine through surface modifications. J Tissue Eng Regen Med 2019; 13:1275-1293. [PMID: 30946537 PMCID: PMC6715496 DOI: 10.1002/term.2859] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 01/22/2019] [Accepted: 01/29/2019] [Indexed: 11/11/2022]
Abstract
Tissue engineering and regenerative medicine rely extensively on biomaterial scaffolds to support cell adhesion, proliferation, and differentiation physically and chemically in vitro and in vivo. Changes to the surface characteristics of the scaffolds have the greatest impact on cell response. Here, we discuss five dominant surface modification approaches used to biomimetically improve the most common scaffolds for tissue engineering, those based on aliphatic polyesters. Scaffolds of aliphatic polyesters such as poly(l-lactic acid), poly(l-lactic-co-glycolic acid), and poly(ε-caprolactone) are often used in tissue engineering because they provide desirable, tunable properties such as ease of manufacturing, good mechanical properties, and nontoxic degradation products. However, cell-surface interactions necessary for tissue engineering are limited on these materials by their smooth postfabrication surfaces, hydrophobicity, and lack of recognizable biochemical binding sites. The surface modification techniques that have been developed for synthetic polymer scaffolds reduce initial barriers to cell adhesion, proliferation, and differentiation. Topographical modification, protein adsorption, mineral coating, functional group incorporation, and biomacromolecule immobilization each contribute through varying mechanisms to improving cell interactions with aliphatic polyester scaffolds. Furthermore, rational combination of methods from these categories can provide nuanced, specific environments for targeted tissue development.
Collapse
Affiliation(s)
- Nathan R Richbourg
- School of Chemical, Biological, and Materials Engineering, The University of Oklahoma, Norman, OK, USA
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Nicholas A Peppas
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Vassilios I Sikavitsas
- School of Chemical, Biological, and Materials Engineering, The University of Oklahoma, Norman, OK, USA
| |
Collapse
|
31
|
Roi A, Ardelean LC, Roi CI, Boia ER, Boia S, Rusu LC. Oral Bone Tissue Engineering: Advanced Biomaterials for Cell Adhesion, Proliferation and Differentiation. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E2296. [PMID: 31323766 PMCID: PMC6679077 DOI: 10.3390/ma12142296] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/11/2019] [Accepted: 07/16/2019] [Indexed: 12/23/2022]
Abstract
The advancements made in biomaterials have an important impact on oral tissue engineering, especially on the bone regeneration process. Currently known as the gold standard in bone regeneration, grafting procedures can sometimes be successfully replaced by a biomaterial scaffold with proper characteristics. Whether natural or synthetic polymers, biomaterials can serve as potential scaffolds with major influences on cell adhesion, proliferation and differentiation. Continuous research has enabled the development of scaffolds that can be specifically designed to replace the targeted tissue through changes in their surface characteristics and the addition of growth factors and biomolecules. The progress in tissue engineering is incontestable and research shows promising contributions to the further development of this field. The present review aims to outline the progress in oral tissue engineering, the advantages of biomaterial scaffolds, their direct implication in the osteogenic process and future research directions.
Collapse
Affiliation(s)
- Alexandra Roi
- Department of Oral Pathology, "Victor Babes" University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Lavinia Cosmina Ardelean
- Department of Technology of Materials and Devices in Dental Medicine, "Victor Babes" University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu sq, 300041 Timisoara, Romania.
| | - Ciprian Ioan Roi
- Department of Anaesthesiology and Oral Surgery, "Victor Babes" University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Eugen-Radu Boia
- Department of Ear, Nose and Throat, "Victor Babes" University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Simina Boia
- Department of Periodontology, "Victor Babes" University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Laura-Cristina Rusu
- Department of Oral Pathology, "Victor Babes" University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
| |
Collapse
|
32
|
Electrospun polymer micro/nanofibers as pharmaceutical repositories for healthcare. J Control Release 2019; 302:19-41. [DOI: 10.1016/j.jconrel.2019.03.020] [Citation(s) in RCA: 180] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/22/2019] [Accepted: 03/23/2019] [Indexed: 12/19/2022]
|
33
|
Jing W, Huang Y, Wei P, Cai Q, Yang X, Zhong W. Roles of electrical stimulation in promoting osteogenic differentiation of BMSCs on conductive fibers. J Biomed Mater Res A 2019; 107:1443-1454. [PMID: 30786145 DOI: 10.1002/jbm.a.36659] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/30/2019] [Accepted: 02/15/2019] [Indexed: 01/22/2023]
Abstract
The strategy of using conductive materials in regenerating bone defects is attractive, benefiting from the bioelectricity feature of natural bone tissues. Thereby, POP conductive fibers were fabricated by coating polypyrrole (PPY) onto electrospun poly(l-lactide) (PLLA) fibers, and their potentials in promoting osteogenic differentiation of bone mesenchymal stromal cells (BMSCs) were investigated. Different from the smooth-surfaced PLLA fibers, POP fibers were rough-surfaced and favorable for protein adsorption and mineralization nucleation. When electrical stimulation (ES) was applied, the surface charges on the conductive POP fibers further promoted the protein adsorption and the mineral deposition, while the non-conductive PLLA fibers displayed no such promotion. When BMSCs were cultured on these fibers, strong cell viability was detected, indicating their good biocompatibility and cell affinity. In osteogenic differentiation studies, BMSCs demonstrated the strongest ability in differentiating toward osteoblasts when they were cultured on the POP fibers under ES, followed by the case without ES. In comparison with the conductive POP fibers, the non-conductive PLLA fibers displayed significantly weaker ability in inducing the osteogenic differentiation of BMSCs with ES being applied or not. Alongside the differentiation, both the calcium deposition on BMSC/material complexes and the intracellular Ca2+ concentration were identified the most abundant when BMSCs grew on the POP fibers under ES. These findings suggested that the surface charges of conductive fibers played roles in regulating protein adsorption, ion migration and nucleation, particularly under ES, which contributed much to the increased intracellular Ca2+ ions, and thus accelerated the osteogenic differentiation of the seeded cells. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A, 2019.
Collapse
Affiliation(s)
- Wei Jing
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Yiqian Huang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Pengfei Wei
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Qing Cai
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Xiaoping Yang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Weihong Zhong
- School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington, 99164
| |
Collapse
|
34
|
Karimi Z, Seyedjafari E, Mahdavi FS, Hashemi SM, Khojasteh A, Kazemi B, Mohammadi-Yeganeh S. Baghdadite nanoparticle-coated poly l-lactic acid (PLLA) ceramics scaffold improved osteogenic differentiation of adipose tissue-derived mesenchymal stem cells. J Biomed Mater Res A 2019; 107:1284-1293. [PMID: 30706628 DOI: 10.1002/jbm.a.36638] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 10/27/2018] [Accepted: 01/28/2019] [Indexed: 01/18/2023]
Abstract
Bone repair has been a new approach in regenerative medicine especially by application of stem cells. Discovering a suitable combination of scaffolds to stimulate osteogenesis is one of the major concerns in this issue. Porous polymeric scaffolds such as poly l-lactic acid (PLLA) have been attracted a lot of attention because of their biodegradability. In the present study, we have been coated Baghdadite on the plasma-treated surface of PLLA and evaluated osteogenic potential of mesenchymal stem cells (MSCs). Adipose tissue-derived mesenchymal stem cells (AD-MSCs) were cultured on PLLA and PLLA-Baghdadite scaffolds, and cell properties were characterized by MTT assay, scanning electron microscope, and FTIR analysis. Then, osteogenic differentiation potential of AD-MSCs has been investigated, such as alkaline phosphatase (ALP) activity, calcium mineral deposition, and the expression of bone-related genes (RUNX2, ALP, and OCN). The results have been indicated that calcium content and ALP activity of cells cultured on PLLA-Baghdadite nanofibers were higher than that of tissue culture polystyrenes (TCPs). Gene expression analysis showed that PLLA-Baghdadite had effectively induced osteogenesis-related genes. Taken together, these results suggest that porous nanofiber scaffolds which coated with Baghdadite can enhance osteogenic differentiation of AD-MSC, and PLLA-Baghdadite can be used as a new biodegradable scaffold for bone regeneration. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 1284-1293, 2019.
Collapse
Affiliation(s)
- Zohreh Karimi
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ehsan Seyedjafari
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Fatemeh Sadat Mahdavi
- Department of Animal and Poultry Science, College of Aburaihan, University of Tehran, Pakdasht, Tehran, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arash Khojasteh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahram Kazemi
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Mohammadi-Yeganeh
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
Xia Y, Chen H, Zhao Y, Zhang F, Li X, Wang L, Weir MD, Ma J, Reynolds MA, Gu N, Xu HHK. Novel magnetic calcium phosphate-stem cell construct with magnetic field enhances osteogenic differentiation and bone tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 98:30-41. [PMID: 30813031 DOI: 10.1016/j.msec.2018.12.120] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/20/2018] [Accepted: 12/27/2018] [Indexed: 01/09/2023]
Abstract
Superparamagnetic iron oxide nanoparticles (IONPs) are promising bioactive additives to fabricate magnetic scaffolds for bone tissue engineering. To date, there has been no report on osteoinductivity of IONP-incorporated calcium phosphate cement (IONP-CPC) scaffold on stem cells using an exterior static magnetic field (SMF). The objectives of this study were to: (1) develop a novel magnetic IONP-CPC construct for bone tissue engineering, and (2) investigate the effects of IONP-incorporation and SMF application on the proliferation, osteogenic differentiation and bone mineral synthesis of human dental pulp stem cells (hDPSCs) seeded on IONP-CPC scaffold for the first time. The novel magnetic IONP-CPC under SMF enhanced the cellular performance of hDPSCs, yielding greater alkaline phosphatase activities (about 3-fold), increased expressions of osteogenic marker genes, and more cell-synthesized bone minerals (about 2.5-fold), compared to CPC control and nonmagnetic IONP-CPC. In addition, IONP-CPC induced more active osteogenesis than CPC control in rat mandible defects. These results were consistent with the enhanced cellular performance by magnetic IONP in media under SMF. Moreover, nano-aggregates were detected inside the cells by transmission electron microscopy (TEM). Therefore, the enhanced cell performance was attributed to the physical forces generated by the magnetic field together with cell internalization of the released magnetic nanoparticles from IONP-CPC constructs.
Collapse
Affiliation(s)
- Yang Xia
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China; Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Huimin Chen
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yantao Zhao
- Beijing Engineering Research Center of Orthopedic Implants, First Affiliated Hospital of CPLA General Hospital, Beijing 100048, China
| | - Feimin Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou, Jiangsu 215123, China
| | - Xiaodong Li
- Department of Oral Medicine, School of Stomatology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lin Wang
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA; VIP Integrated Department, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Michael D Weir
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Junqing Ma
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Mark A Reynolds
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Ning Gu
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China; Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou, Jiangsu 215123, China.
| | - Hockin H K Xu
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA; Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; University of Maryland Marlene and Stewart Greene Baum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
36
|
Chen S, Zhao X, Du C. Macroporous poly (l-lactic acid)/chitosan nanofibrous scaffolds through cloud point thermally induced phase separation for enhanced bone regeneration. Eur Polym J 2018. [DOI: 10.1016/j.eurpolymj.2018.10.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
37
|
Liu Z, Chen X, Zhang Z, Zhang X, Saunders L, Zhou Y, Ma PX. Nanofibrous Spongy Microspheres To Distinctly Release miRNA and Growth Factors To Enrich Regulatory T Cells and Rescue Periodontal Bone Loss. ACS NANO 2018; 12:9785-9799. [PMID: 30141906 PMCID: PMC6205210 DOI: 10.1021/acsnano.7b08976] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
In addition to T cells' roles in immune response and autoimmune diseases, certain types of T cells, called regulatory T cells (Tregs), play important roles in microenvironment modulation for resolution and tissue regeneration. However, there are currently few options available other than introducing more Tregs or immunosuppressive drugs to locally enrich Tregs. Herein, poly(l-lactic acid) (PLLA) nanofibrous spongy microspheres (NF-SMS), PLLA/polyethylene glycol (PEG) co-functionalized mesoporous silica nanoparticles (MSN), and poly(lactic acid- co-glycolic acid) microspheres (PLGA MS) are integrated into one multibiologic delivery vehicle for in situ Treg manipulation, where the MSNs and PLGA MS were utilized to distinctly release IL-2/TGF-β and miR-10a to locally recruit T cells and stimulate their differentiation into Tregs, while PLLA NF-SMS serve as an injectable scaffold for the adhesion and proliferation of these Tregs. In a mouse model of periodontitis, the injectable and biomolecule-delivering PLLA NF-SMS lead to Treg enrichment, expansion, and Treg-mediated immune therapy against bone loss. This system can potentially be utilized in a wide variety of other immune and regenerative therapies.
Collapse
Affiliation(s)
- Zhongning Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology; National Clinical Research Center for Oral Diseases; National Engineering Laboratory for Digital and Material Technology of Stomatology; Beijing Key Laboratory of Digital Stomatology, Beijing 100081, PR China
- Department of Biologic and Materials Sciences, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Xin Chen
- Department of Biologic and Materials Sciences, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Zhanpeng Zhang
- Department of Biologic and Materials Sciences, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Xiaojin Zhang
- Department of Biologic and Materials Sciences, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Laura Saunders
- Macromolecular Science and Engineering Center, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology; National Clinical Research Center for Oral Diseases; National Engineering Laboratory for Digital and Material Technology of Stomatology; Beijing Key Laboratory of Digital Stomatology, Beijing 100081, PR China
- Corresponding Authors (Y. Zhou): , (P.X. Ma):
| | - Peter X. Ma
- Department of Biologic and Materials Sciences, The University of Michigan, Ann Arbor, MI 48109, USA
- Macromolecular Science and Engineering Center, The University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, Department of Materials Sciences and Engineering, The University of Michigan, Ann Arbor, MI 48109, USA
- Corresponding Authors (Y. Zhou): , (P.X. Ma):
| |
Collapse
|
38
|
Wang X, Wang G, Zingales S, Zhao B. Biomaterials Enabled Cell-Free Strategies for Endogenous Bone Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2018; 24:463-481. [PMID: 29897021 DOI: 10.1089/ten.teb.2018.0012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Repairing bone defects poses a major orthopedic challenge because current treatments are constrained by the limited regenerative capacity of human bone tissue. Novel therapeutic strategies, such as stem cell therapy and tissue engineering, have the potential to enhance bone healing and regeneration, and hence may improve quality of life for millions of people. However, the ex vivo expansion of stem cells and their in vivo delivery pose technical difficulties that hamper clinical translation and commercial development. A promising alternative to cell delivery-based strategies is to stimulate or augment the inherent self-repair mechanisms of the patient to promote endogenous restoration of the lost/damaged bone. There is growing evidence indicating that increasing the endogenous regenerative potency of bone tissues for therapeutics will require the design and development of new generations of biomedical devices that provide key signaling molecules to instruct cell recruitment and manipulate cell fate for in situ tissue regeneration. Currently, a broad range of biomaterial-based deployment technologies are becoming available, which allow for controlled spatial presentation of biological cues required for endogenous bone regeneration. This article aims to explore the proposed concepts and biomaterial-enabled strategies involved in the design of cell-free endogenous techniques in bone regenerative medicine.
Collapse
Affiliation(s)
- Xiaojing Wang
- 1 Dental Implant Center, Affiliated Hospital of Qingdao University , Qingdao, P.R. China .,2 School of Stomatology, Qingdao University , Qingdao, Shandong, P.R. China
| | - Guowei Wang
- 3 Department of Stomatology, Laoshan Branch of No. 401 Hospital of the Chinese Navy , Qingdao, Shandong, P.R. China
| | - Sarah Zingales
- 4 Department of Chemistry and Biochemistry, Georgia Southern University , Savannah, Georgia
| | - Baodong Zhao
- 1 Dental Implant Center, Affiliated Hospital of Qingdao University , Qingdao, P.R. China .,2 School of Stomatology, Qingdao University , Qingdao, Shandong, P.R. China
| |
Collapse
|
39
|
Turnbull G, Clarke J, Picard F, Riches P, Jia L, Han F, Li B, Shu W. 3D bioactive composite scaffolds for bone tissue engineering. Bioact Mater 2018; 3:278-314. [PMID: 29744467 PMCID: PMC5935790 DOI: 10.1016/j.bioactmat.2017.10.001] [Citation(s) in RCA: 584] [Impact Index Per Article: 97.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/31/2017] [Accepted: 10/31/2017] [Indexed: 12/13/2022] Open
Abstract
Bone is the second most commonly transplanted tissue worldwide, with over four million operations using bone grafts or bone substitute materials annually to treat bone defects. However, significant limitations affect current treatment options and clinical demand for bone grafts continues to rise due to conditions such as trauma, cancer, infection and arthritis. Developing bioactive three-dimensional (3D) scaffolds to support bone regeneration has therefore become a key area of focus within bone tissue engineering (BTE). A variety of materials and manufacturing methods including 3D printing have been used to create novel alternatives to traditional bone grafts. However, individual groups of materials including polymers, ceramics and hydrogels have been unable to fully replicate the properties of bone when used alone. Favourable material properties can be combined and bioactivity improved when groups of materials are used together in composite 3D scaffolds. This review will therefore consider the ideal properties of bioactive composite 3D scaffolds and examine recent use of polymers, hydrogels, metals, ceramics and bio-glasses in BTE. Scaffold fabrication methodology, mechanical performance, biocompatibility, bioactivity, and potential clinical translations will be discussed.
Collapse
Affiliation(s)
- Gareth Turnbull
- Department of Biomedical Engineering, Wolfson Building, University of Strathclyde, 106 Rottenrow, Glasgow, G4 0NW, United Kingdom
- Department of Orthopaedic Surgery, Golden Jubilee National Hospital, Agamemnon St, Clydebank, G81 4DY, United Kingdom
| | - Jon Clarke
- Department of Orthopaedic Surgery, Golden Jubilee National Hospital, Agamemnon St, Clydebank, G81 4DY, United Kingdom
| | - Frédéric Picard
- Department of Biomedical Engineering, Wolfson Building, University of Strathclyde, 106 Rottenrow, Glasgow, G4 0NW, United Kingdom
- Department of Orthopaedic Surgery, Golden Jubilee National Hospital, Agamemnon St, Clydebank, G81 4DY, United Kingdom
| | - Philip Riches
- Department of Biomedical Engineering, Wolfson Building, University of Strathclyde, 106 Rottenrow, Glasgow, G4 0NW, United Kingdom
| | - Luanluan Jia
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, PR China
| | - Fengxuan Han
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, PR China
| | - Bin Li
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, PR China
| | - Wenmiao Shu
- Department of Biomedical Engineering, Wolfson Building, University of Strathclyde, 106 Rottenrow, Glasgow, G4 0NW, United Kingdom
| |
Collapse
|
40
|
Jiang N, Guo Z, Sun D, Li Y, Yang Y, Chen C, Zhang L, Zhu S. Promoting Osseointegration of Ti Implants through Micro/Nanoscaled Hierarchical Ti Phosphate/Ti Oxide Hybrid Coating. ACS NANO 2018; 12:7883-7891. [PMID: 29979574 DOI: 10.1021/acsnano.8b02227] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In this study, micro/nanoscaled hierarchical hybrid coatings containing titanium (Ti) phosphate and Ti oxide have been fabricated with the aim of promoting osseointegration of Ti-based implants. Three representative surface coatings, namely, micro/nanograss Ti (P-G-Ti), micro/nanoclump Ti, (P-C-Ti), and micro/nanorod Ti (P-R-Ti), have been produced. In-depth investigations into the coating surface morphology, topography, chemical composition, and the surface/cell interaction have been carried out using scanning electron microscopy, transmission electron microscope, X-ray photoelectron spectroscopy, X-ray diffraction, contact-angle measurement, and protein adsorption assay. In addition, in vitro performance of the coating (cell proliferation, adhesion, and differentiation) has been evaluated using rat bone marrow stromal cells (BMSCs), and in vivo assessments have been carried out based on a rat tibia implantation model. All the hybrid coating modified implants demonstrated enhanced protein adsorption and BMSC viability, adhesion and differentiation, with P-G-Ti showing the best bioactivity among all samples. Subsequent i n vivo osseointegration tests confirmed that P-G-Ti has induced a much stronger interfacial bonding with the host tissue, indicated by the 2-fold increase in the ultimate shear strength and over 6-fold increase in the maximum push-out force compared to unmodified Ti implants. The state-of-the-art coating technology proposed for Ti-based implants in this study holds great potential in advancing medical devices for next-generation healthcare technology.
Collapse
Affiliation(s)
- Nan Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, and West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China
| | - Zhijun Guo
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610065, China
- School of Mechanical and Aerospace Engineering, Queens University Belfast, Belfast BT7 1NN, U.K
| | - Dan Sun
- School of Mechanical and Aerospace Engineering, Queens University Belfast, Belfast BT7 1NN, U.K
| | - Yubao Li
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610065, China
| | - Yutao Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, and West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China
| | - Chen Chen
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610065, China
| | - Li Zhang
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610065, China
| | - Songsong Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, and West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China
| |
Collapse
|
41
|
Rahman SU, Nagrath M, Ponnusamy S, Arany PR. Nanoscale and Macroscale Scaffolds with Controlled-Release Polymeric Systems for Dental Craniomaxillofacial Tissue Engineering. MATERIALS (BASEL, SWITZERLAND) 2018; 11:E1478. [PMID: 30127246 PMCID: PMC6120038 DOI: 10.3390/ma11081478] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/03/2018] [Accepted: 08/10/2018] [Indexed: 12/11/2022]
Abstract
Tremendous progress in stem cell biology has resulted in a major current focus on effective modalities to promote directed cellular behavior for clinical therapy. The fundamental principles of tissue engineering are aimed at providing soluble and insoluble biological cues to promote these directed biological responses. Better understanding of extracellular matrix functions is ensuring optimal adhesive substrates to promote cell mobility and a suitable physical niche to direct stem cell responses. Further, appreciation of the roles of matrix constituents as morphogen cues, termed matrikines or matricryptins, are also now being directly exploited in biomaterial design. These insoluble topological cues can be presented at both micro- and nanoscales with specific fabrication techniques. Progress in development and molecular biology has described key roles for a range of biological molecules, such as proteins, lipids, and nucleic acids, to serve as morphogens promoting directed behavior in stem cells. Controlled-release systems involving encapsulation of bioactive agents within polymeric carriers are enabling utilization of soluble cues. Using our efforts at dental craniofacial tissue engineering, this narrative review focuses on outlining specific biomaterial fabrication techniques, such as electrospinning, gas foaming, and 3D printing used in combination with polymeric nano- or microspheres. These avenues are providing unprecedented therapeutic opportunities for precision bioengineering for regenerative applications.
Collapse
Affiliation(s)
- Saeed Ur Rahman
- Departments of Oral Biology and Biomedical Engineering, School of Dentistry, University at Buffalo, Buffalo, NY 14214, USA.
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad, Lahore Campus, Lahore 54000, Pakistan.
| | - Malvika Nagrath
- Departments of Oral Biology and Biomedical Engineering, School of Dentistry, University at Buffalo, Buffalo, NY 14214, USA.
- Department of Biomedical Engineering, Ryerson University, Toronto, ON M5B 2K3, Canada.
| | - Sasikumar Ponnusamy
- Departments of Oral Biology and Biomedical Engineering, School of Dentistry, University at Buffalo, Buffalo, NY 14214, USA.
| | - Praveen R Arany
- Departments of Oral Biology and Biomedical Engineering, School of Dentistry, University at Buffalo, Buffalo, NY 14214, USA.
| |
Collapse
|
42
|
Li Q, Zhang B, Kasoju N, Ma J, Yang A, Cui Z, Wang H, Ye H. Differential and Interactive Effects of Substrate Topography and Chemistry on Human Mesenchymal Stem Cell Gene Expression. Int J Mol Sci 2018; 19:E2344. [PMID: 30096912 PMCID: PMC6121573 DOI: 10.3390/ijms19082344] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/05/2018] [Accepted: 08/07/2018] [Indexed: 12/22/2022] Open
Abstract
Variations in substrate chemistry and the micro-structure were shown to have a significant effect on the biology of human mesenchymal stromal cells (hMSCs). This occurs when differences in the surface properties indirectly modulate pathways within numerous signaling networks that control cell fate. To understand how the surface features affect hMSC gene expression, we performed RNA-sequencing analysis of bone marrow-derived hMSCs cultured on tissue culture-treated polystyrene (TCP) and poly(l-lactide) (PLLA) based substrates of differing topography (Fl: flat and Fs: fibrous) and chemistry (Pr: pristine and Am: aminated). Whilst 80% of gene expression remained similar for cells cultured on test substrates, the analysis of differentially expressed genes (DEGs) revealed that surface topography significantly altered gene expression more than surface chemistry. The Fl and Fs topologies introduced opposite directional alternations in gene expression when compared to TCP control. In addition, the effect of chemical treatment interacted with that of topography in a synergistic manner with the Pr samples promoting more DEGs than Am samples in all gene ontology function groups. These findings not only highlight the significance of the culture surface on regulating the overall gene expression profile but also provide novel insights into cell-material interactions that could help further design the next-generation biomaterials to facilitate hMSC applications. At the same time, further studies are required to investigate whether or not the observations noted correlate with subsequent protein expression and functionality of cells.
Collapse
Affiliation(s)
- Qiongfang Li
- China National GeneBank-Shenzhen, BGI-Shenzhen, 518083 Shenzhen, China.
| | - Bo Zhang
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ Oxford, UK.
- Department of Engineering Science, University of Oxford, OX1 3PJ Oxford, UK.
| | - Naresh Kasoju
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ Oxford, UK.
| | - Jinmin Ma
- China National GeneBank-Shenzhen, BGI-Shenzhen, 518083 Shenzhen, China.
| | - Aidong Yang
- Department of Engineering Science, University of Oxford, OX1 3PJ Oxford, UK.
| | - Zhanfeng Cui
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ Oxford, UK.
| | - Hui Wang
- China National GeneBank-Shenzhen, BGI-Shenzhen, 518083 Shenzhen, China.
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ Oxford, UK.
- Oxford Suzhou Centre for Advanced Research, Suzhou Industrial Park, 215123 Suzhou, China.
| | - Hua Ye
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ Oxford, UK.
| |
Collapse
|
43
|
Zhou X, Ma C, Hu B, Tao Y, Wang J, Huang X, Zhao T, Han B, Li H, Liang C, Chen Q, Li F. FoxA2 regulates the type II collagen-induced nucleus pulposus-like differentiation of adipose-derived stem cells by activation of the Shh signaling pathway. FASEB J 2018; 32:fj201800373R. [PMID: 29890089 DOI: 10.1096/fj.201800373r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Adipose tissue-derived stem cell (ADSC)-based therapy is promising for the treatment of intervertebral disc (IVD) degeneration, but the difficulty in inducing nucleus pulposus (NP)-like differentiation limits its clinical applications. Forkhead box (Fox)-A2 is an essential transcription factor for the formation of a normal NP. We demonstrated that type II collagen stimulates NP-like differentiation of ADSCs, partly by increasing the expression of FoxA2. We constructed FoxA2-overexpressing and -knockdown ADSCs by using lentiviral vectors. FoxA2 overexpression significantly enhanced NP-specific gene expression and the synthesis of glycosaminoglycan and collagen, whereas FoxA2 knockdown decreased NP-like differentiation and the expression of aggrecan and collagen II. The enhanced NP-like differentiation related to FoxA2 overexpression was partially rescued by an Shh signaling pathway inhibitor. In addition, FoxA2 inhibited the expression of Itg-α2 and further promoted NP-like differentiation induced by type II collagen. Furthermore, FoxA2-overexpressing ADSCs combined with type II collagen hydrogels promoted regeneration of degenerated NP in vivo. Our findings suggest that FoxA2 plays an essential role in the NP-like differentiation of ADSCs by activating the Shh signaling pathway.-Zhou, X., Ma, C., Hu, B., Tao, Y., Wang, J., Huang, X., Zhao, T., Han, B., Li, H., Liang, C., Chen, Q., Li, F. FoxA2 regulates the type II collagen-induced nucleus pulposus-like differentiation of adipose-derived stem cells by activation of the Shh signaling pathway.
Collapse
Affiliation(s)
- Xiaopeng Zhou
- Department of Orthopedics Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Orthopedics, Research Institute of Zhejiang University, Hangzhou, China
| | - Chiyuan Ma
- Department of Orthopedics Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Orthopedics, Research Institute of Zhejiang University, Hangzhou, China
| | - Bin Hu
- Department of Orthopedics Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Orthopedics, Research Institute of Zhejiang University, Hangzhou, China
| | - Yiqing Tao
- Department of Orthopedics Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Orthopedics, Research Institute of Zhejiang University, Hangzhou, China
| | - Jingkai Wang
- Department of Orthopedics Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Orthopedics, Research Institute of Zhejiang University, Hangzhou, China
| | - Xianpeng Huang
- Department of Orthopedics Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Orthopedics, Research Institute of Zhejiang University, Hangzhou, China
| | - Tengfei Zhao
- Department of Orthopedics Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Orthopedics, Research Institute of Zhejiang University, Hangzhou, China
| | - Bin Han
- Department of Orthopedics Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Orthopedics, Research Institute of Zhejiang University, Hangzhou, China
| | - Hao Li
- Department of Orthopedics Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Orthopedics, Research Institute of Zhejiang University, Hangzhou, China
| | - Chengzhen Liang
- Department of Orthopedics Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Orthopedics, Research Institute of Zhejiang University, Hangzhou, China
| | - Qixin Chen
- Department of Orthopedics Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Orthopedics, Research Institute of Zhejiang University, Hangzhou, China
| | - Fangcai Li
- Department of Orthopedics Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Orthopedics, Research Institute of Zhejiang University, Hangzhou, China
| |
Collapse
|
44
|
From nano to micro to macro: Electrospun hierarchically structured polymeric fibers for biomedical applications. Prog Polym Sci 2018. [DOI: 10.1016/j.progpolymsci.2017.12.003] [Citation(s) in RCA: 210] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
45
|
Onur T, Yuca E, Olmez TT, Seker UOS. Self-assembly of bacterial amyloid protein nanomaterials on solid surfaces. J Colloid Interface Sci 2018. [DOI: 10.1016/j.jcis.2018.03.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
46
|
Rahman SU, Oh JH, Cho YD, Chung SH, Lee G, Baek JH, Ryoo HM, Woo KM. Fibrous Topography-Potentiated Canonical Wnt Signaling Directs the Odontoblastic Differentiation of Dental Pulp-Derived Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2018; 10:17526-17541. [PMID: 29741358 DOI: 10.1021/acsami.7b19782] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Nanofibrous engineered matrices have significant potential in cellular differentiation and tissue regeneration. Stem cells require specific extracellular signals that lead to the induction of different lineages. However, the mechanisms by which the nanofibrous matrix promotes mesenchymal stem cell (MSC) differentiation are largely unknown. Here, we investigated the mechanisms that underlie nanofibrous matrix-induced odontoblastic differentiation of human dental pulp MSCs (DP-MSCs). An electrospun polystyrene nanofibrous (PSF) matrix was prepared, and the cell responses to the PSF matrix were assessed in comparison with those on conventional tissue culture dishes. The PSF matrix promoted the expression of Wnt3a, Wnt5a, Wnt10a, BMP2, BMP4, and BMP7 in the DP-MSCs, concomitant with the induction of odontoblast/osteoblast differentiation markers, dentin sialophosphoprotein (DSPP), osteocalcin, and bone sialoprotein, whose levels were further enhanced by treatment with recombinant Wnt3a. The DP-MSCs cultured on the PSF matrix also exhibited a high alkaline phosphatase activity and intense Alizarin Red staining, indicating that the PSF matrix promotes odontoblast differentiation. Besides inducing the expression of Wnt3a, the PSF matrix maintained high levels of β-catenin protein and enhanced its translocation to the nucleus, leading to its transcriptional activity. Forced expression of LEF1 or treatments with LiCl further enhanced the DSPP expression. Blocking the Wnt3a-initiated signaling abrogated the PSF-induced DSPP expression. Furthermore, the cells on the PSF matrix increased the DSPP promoter activity. The β-catenin complex was bound to the conserved motifs on the DSPP promoter dictating its transcription. Transplantations of the preodontoblast-seeded PSF matrix to the subcutaneous tissues of nude mice confirmed the association of the PSF matrix with the Wnt3a and DSPP expressions in vivo. Taken together, these results demonstrate the nanofibrous engineered matrix strongly supports odontoblastic differentiation of DP-MSCs by enhancing Wnt/β-catenin signaling.
Collapse
|
47
|
Manoukian OS, Aravamudhan A, Lee P, Arul MR, Yu X, Rudraiah S, Kumbar SG. Spiral Layer-by-Layer Micro-Nanostructured Scaffolds for Bone Tissue Engineering. ACS Biomater Sci Eng 2018; 4:2181-2192. [PMID: 30976659 DOI: 10.1021/acsbiomaterials.8b00393] [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] [Indexed: 12/31/2022]
Abstract
This Article reports the fabrication and characterization of composite micro-nanostructured spiral scaffolds functionalized with nanofibers and hydroxyapatite (HA) for bone regeneration. The spiral poly(lactic acid-co-glycolic acid) (PLGA) porous microstructure was coated with sparsely spaced PLGA nanofibers and HA to enhance surface area and bioactivity. Polyelectrolyte-based HA coating in a layer-by-layer (LBL) fashion allowed 10-70 μM Ca2+/mm2 incorporation. These scaffolds provided a controlled release of Ca2+ ions up to 60 days with varied release kinetics accounting up to 10-50 μg. Spiral scaffolds supported superior adhesion, proliferation, and osteogenic differentiation of rat bone marrow stromal cells (MSCs) as compared to controls microstructures. Spiral micro-nanostructures supported homogeneous tissue ingrowth and resulted in bone-island formation in the center of the scaffold as early as 3 weeks in a rabbit ulnar bone defect model. In contrast, control cylindrical scaffolds showed tissue ingrowth only at the surface because of limitations in scaffold transport features.
Collapse
Affiliation(s)
- Ohan S Manoukian
- Department of Orthopaedic Surgery, University of Connecticut Health, 263 Farmington Avenue, Farmington, Connecticut 06030, United States.,Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, Connecticut 06269, United States
| | - Aja Aravamudhan
- Department of Orthopaedic Surgery, University of Connecticut Health, 263 Farmington Avenue, Farmington, Connecticut 06030, United States
| | - Paul Lee
- Department of Chemistry, Chemical Biology, and Biomedical Engineering, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, New Jersey 07030, United States
| | - Michael R Arul
- Department of Orthopaedic Surgery, University of Connecticut Health, 263 Farmington Avenue, Farmington, Connecticut 06030, United States
| | - Xiaojun Yu
- Department of Chemistry, Chemical Biology, and Biomedical Engineering, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, New Jersey 07030, United States
| | - Swetha Rudraiah
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Saint Joseph, 229 Trumbull St., Hartford Connecticut 06103, United States
| | - Sangamesh G Kumbar
- Department of Orthopaedic Surgery, University of Connecticut Health, 263 Farmington Avenue, Farmington, Connecticut 06030, United States.,Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, Connecticut 06269, United States
| |
Collapse
|
48
|
Rainer A, Centola M, Spadaccio C, Gherardi G, Genovese JA, Licoccia S, Trombetta M. Comparative Study of Different Techniques for the Sterilization of Poly-L-lactide Electrospun Microfibers: Effectiveness vs. Material Degradation. Int J Artif Organs 2018. [DOI: 10.1177/039139881003300203] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Electrospinning of biopolymeric scaffolds is a new and effective approach for creating replacement tissues to repair defects and/or damaged tissues with direct clinical application. However, many hurdles and technical concerns regarding biological issues, such as cell retention and the ability to grow, still need to be overcome to gain full access to the clinical arena. Interaction with the host human tissues, immunogenicity, pathogen transmission as well as production costs, technical expertise, and good manufacturing and laboratory practice requirements call for careful consideration when aiming at the production of a material that is available off-the-shelf, to be used immediately in operative settings. The issue of sterilization is one of the most important steps for the clinical application of these scaffolds. Nevertheless, relatively few studies have been performed to systematically investigate how sterilization treatments may affect the properties of electrospun polymers for tissue engineering. This paper presents the results of a comparative study of different sterilization techniques applied to an electrospun poly-L-lactide scaffold: soaking in absolute ethanol, dry oven and autoclave treatments, UV irradiation, and hydrogen peroxide gas plasma treatment. Morphological and chemical characterization was coupled with microbiological sterility assay to validate the examined sterilization techniques in terms of effectiveness and modifications to the scaffold. The results of this study reveal that UV irradiation and hydrogen peroxide gas plasma are the most effective sterilization techniques, as they ensure sterility of the electrospun scaffolds without affecting their chemical and morphological features.
Collapse
Affiliation(s)
- Alberto Rainer
- Center of Integrated Research (CIR) – Laboratory of Chemistry & Biomaterials, University Campus Bio-Medico of Rome, Rome
| | - Matteo Centola
- Center of Integrated Research (CIR) – Laboratory of Chemistry & Biomaterials, University Campus Bio-Medico of Rome, Rome
| | - Cristiano Spadaccio
- CIR - Area of Cardiovascular Surgery, University Campus Bio-Medico of Rome, Rome
| | - Giovanni Gherardi
- CIR - Laboratory of Microbiology, University Campus Bio-Medico of Rome, Rome
| | - Jorge A. Genovese
- CIR - Area of Cardiovascular Surgery, University Campus Bio-Medico of Rome, Rome
| | - Silvia Licoccia
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Rome
- NAST Center for Nanoscience, Nanotechnology & Innovative Instrumentation, University of Rome Tor Vergata, Rome - Italy
| | - Marcella Trombetta
- Center of Integrated Research (CIR) – Laboratory of Chemistry & Biomaterials, University Campus Bio-Medico of Rome, Rome
| |
Collapse
|
49
|
Sang Y, Li M, Liu J, Yao Y, Ding Z, Wang L, Xiao L, Lu Q, Fu X, Kaplan DL. Biomimetic Silk Scaffolds with an Amorphous Structure for Soft Tissue Engineering. ACS APPLIED MATERIALS & INTERFACES 2018; 10:9290-9300. [PMID: 29485270 DOI: 10.1021/acsami.7b19204] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Fine tuning physical cues of silk fibroin (SF) biomaterials to match specific requirements for different soft tissues would be advantageous. Here, amorphous SF nanofibers were used to fabricate scaffolds with better hierarchical extracellular matrix (ECM) mimetic microstructures than previous silk scaffolds. Kinetic control was introduced into the scaffold forming process, resulting in the direct production of water-stable scaffolds with tunable secondary structures and thus mechanical properties. These biomaterials remained with amorphous structures, offering softer properties than prior scaffolds. The fine mechanical tunability of these systems provides a feasible way to optimize physical cues for improved cell proliferation and enhanced neovascularization in vivo. Multiple physical cues, such as partly ECM mimetic structures and optimized stiffness, provided suitable microenvironments for tissue ingrowth, suggesting the possibility of actively designing bioactive SF biomaterials. These systems suggest a promising strategy to develop novel SF biomaterials for soft tissue repair and regenerative medicine.
Collapse
Affiliation(s)
| | - Meirong Li
- Healing and Cell Biology Laboratory, Institute of Basic Medicine Science , Chinese PLA General Hospital , Beijing 100853 , People's Republic of China
| | - Jiejie Liu
- Healing and Cell Biology Laboratory, Institute of Basic Medicine Science , Chinese PLA General Hospital , Beijing 100853 , People's Republic of China
| | | | | | | | | | | | - Xiaobing Fu
- Healing and Cell Biology Laboratory, Institute of Basic Medicine Science , Chinese PLA General Hospital , Beijing 100853 , People's Republic of China
| | - David L Kaplan
- Department of Biomedical Engineering , Tufts University , Medford , Massachusetts 02155 , United States
| |
Collapse
|
50
|
Soares DG, Zhang Z, Mohamed F, Eyster TW, de Souza Costa CA, Ma PX. Simvastatin and nanofibrous poly(l-lactic acid) scaffolds to promote the odontogenic potential of dental pulp cells in an inflammatory environment. Acta Biomater 2018; 68:190-203. [PMID: 29294374 DOI: 10.1016/j.actbio.2017.12.037] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 12/15/2017] [Accepted: 12/22/2017] [Indexed: 12/17/2022]
Abstract
In this study, we investigated the anti-inflammatory, odontogenic and pro-angiogenic effects of integrating simvastatin and nanofibrous poly(l-lactic acid) (NF-PLLA) scaffolds on dental pulp cells (DPCs). Highly porous NF-PLLA scaffolds that mimic the nanofibrous architecture of extracellular matrix were first fabricated, then seeded with human DPCs and cultured with 0.1 μM simvastatin and/or 10 μg/mL pro-inflammatory stimulator lipopolysaccharide (LPS). The gene expression of pro-inflammatory mediators (TNF-α, IL-1β and MMP-9 mRNA) and odontoblastic markers (ALP activity, calcium content, DSPP, DMP-1 and BMP-2 mRNA) were quantified after long-term culture in vitro. In addition, we evaluated the scaffold's pro-angiogenic potential after 24 h of in vitro co-culture with endothelial cells. Finally, we assessed the combined effects of simvastatin and NF-PLLA scaffolds in vivo using a subcutaneous implantation mouse model. The in vitro studies demonstrated that, compared with the DPC/NF-PLLA scaffold constructs cultured only with pro-inflammatory stimulator LPS, adding simvastatin significantly repress the expression of pro-inflammatory mediators. Treating LPS+ DPC/NF-PLLA constructs with simvastatin also reverted the negative effects of LPS on expression of odontoblastic markers in vitro and in vivo. Western blot analysis demonstrated that these effects were related to a reduction in NFkBp65 phosphorylation and up-regulation of PPARγ expression, as well as to increased phosphorylation of pERK1/2 and pSmad1, mediated by simvastatin on LPS-stimulated DPCs. The DPC/NF-PLLA constructs treated with LPS/simvastatin also led to an increase in vessel-like structures, correlated with increased VEGF expression in both DPSCs and endothelial cells. Therefore, the combination of low dosage simvastatin and NF-PLLA scaffolds appears to be a promising strategy for dentin regeneration with inflamed dental pulp tissue, by minimizing the inflammatory reaction and increasing the regenerative potential of resident stem cells. STATEMENT OF SIGNIFICANCE The regeneration potential of stem cells is dependent on their microenvironment. In this study, we investigated the effect of the microenvironment of dental pulp stem cells (DPSCs), including 3D structure of a macroporous and nanofibrous scaffold, the inflammatory stimulus lipopolysaccharide (LPS) and a biological molecule simvastatin, on their regenerative potential of mineralized dentin tissue. The results demonstrated that LPS upregulated inflammatory mediators and suppressed the odontogenic potential of DPSCs. Known as a lipid-lowing agent, simvastatin was excitingly found to repress the expression of pro-inflammatory mediators, up-regulate odontoblastic markers, and exert a pro-angiogenic effect on endothelial cells, resulting in enhanced vascularization and mineralized dentin tissue regeneration in a biomimetic 3D tissue engineering scaffold. This novel finding is significant for the fields of stem cells, inflammation and dental tissue regeneration.
Collapse
|