1
|
Gavva C, Sharan K, Chilkunda N. Mechanistic studies of chondroitin sulfate/dermatan sulfate isolated from freshwater fish discards on osteogenesis in MC3T3-E1 cells. Glycoconj J 2025; 42:15-26. [PMID: 39883365 DOI: 10.1007/s10719-025-10178-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 01/31/2025]
Abstract
Glycosaminoglycans (GAGs) are essential bone extracellular matrix molecules that regulate osteoblast differentiation. Numerous studies have explored endogenous and exogenous GAG osteoanabolic activities using appropriate in vitro and in vivo models. However, GAGs' underlying the mechanism of action and structure-function relationships need to be elucidated in detail. Earlier, we showed that exogenous GAG can bring about osteogenesis in pre-osteoblast cells. In the present study, we have elucidated the mechanism of action of exogenous GAGs, especially of the chondroitin sulfate/dermatan sulfate (CS/DS) class on osteogenesis. GAGs were immobilized, and osteoblast differentiation was evaluated in MC3T3-E1 cells. Results indicated that GAGs supported osteoblast differentiation by promoting collagen production, extracellular matrix formation, and subsequent mineralization. We elucidated the mechanisms underlying these effects by assessing the key signaling molecules involved in osteogenesis in response to exogenous CS/DS with/without BMP2. CS/DS alone significantly increased pERK1/2 and ATF4 expression levels differentially in a time-dependent manner without significant effects on BMP2, RUNX2, and pSMAD5 protein expression. On the other hand, CS/DS, in the presence of BMP2, differentially increased BMP2, pSMAD5, pERK1/2, RUNX2, and ATF4 expression levels at various time points. Collectively, these results strongly suggest that CS/DS can promote osteogenesis, and in the presence of BMP2, it could promote SMAD-mediated ERK-dependent osteogenesis.
Collapse
Affiliation(s)
- Chandra Gavva
- Department of Molecular Nutrition, CSIR-CFTRI, Mysuru, 570020, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Kunal Sharan
- Department of Molecular Nutrition, CSIR-CFTRI, Mysuru, 570020, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Nandini Chilkunda
- Department of Molecular Nutrition, CSIR-CFTRI, Mysuru, 570020, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
2
|
Toba K, Yamada A, Sasa K, Shirota T, Kamijo R. Expression of Kielin/chordin-like protein is regulated by BMP-2 in osteoblasts. Bone Rep 2024; 22:101793. [PMID: 39139593 PMCID: PMC11321374 DOI: 10.1016/j.bonr.2024.101793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 08/15/2024] Open
Abstract
Bone morphogenetic protein (BMP), an osteoinductive factor, is a cytokine that induces osteoblast differentiation and mineralization, and expected to be applicable for hard tissue reconstruction. Kielin/chordin-like protein (Kcp), a member of the family of cysteine-rich proteins, enhances BMP signaling. The present study found that expression of Kcp in osteoblasts was induced by BMP-2 in a concentration- and time-dependent manner. Up-regulation of Kcp by BMP-2 was inhibited by Dorsomorphin, a SMAD signaling inhibitor. The involvement of up-regulation of Kcp by BMP-2 in induction of osteoblast differentiation by BMP-2 was also examined, which showed that suppression of Kcp expression by si Kcp partially inhibited induction of osteoblast differentiation and mineralization by BMP-2. Together, these results suggest that Kcp induced by BMP-2 functions to provide positive feedback for promotion of osteoblastic differentiation and mineralization by BMP-2 in osteoblasts.
Collapse
Affiliation(s)
- Kazuki Toba
- Department of Biochemistry, Graduate School of Dentistry, Showa University, Tokyo, Japan
- Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Showa University, Tokyo, Japan
| | - Atsushi Yamada
- Department of Biochemistry, Graduate School of Dentistry, Showa University, Tokyo, Japan
| | - Kiyohito Sasa
- Department of Biochemistry, Graduate School of Dentistry, Showa University, Tokyo, Japan
| | - Tatsuo Shirota
- Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Showa University, Tokyo, Japan
| | - Ryutaro Kamijo
- Department of Biochemistry, Graduate School of Dentistry, Showa University, Tokyo, Japan
| |
Collapse
|
3
|
Che Z, Sun Q, Zhao Z, Wu Y, Xing H, Song K, Chen A, Wang B, Cai M. Growth factor-functionalized titanium implants for enhanced bone regeneration: A review. Int J Biol Macromol 2024; 274:133153. [PMID: 38897500 DOI: 10.1016/j.ijbiomac.2024.133153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/02/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024]
Abstract
Titanium and titanium alloys are widely favored materials for orthopedic implants due to their exceptional mechanical properties and biological inertness. The additional benefit of sustained local release of bioactive substances further promotes bone tissue formation, thereby augmenting the osseointegration capacity of titanium implants and attracting increasing attention in bone tissue engineering. Among these bioactive substances, growth factors have shown remarkable osteogenic and angiogenic induction capabilities. Consequently, researchers have developed various physical, chemical, and biological loading techniques to incorporate growth factors into titanium implants, ensuring controlled release kinetics. In contrast to conventional treatment modalities, the localized release of growth factors from functionalized titanium implants not only enhances osseointegration but also reduces the risk of complications. This review provides a comprehensive examination of the types and mechanisms of growth factors, along with a detailed exploration of the methodologies used to load growth factors onto the surface of titanium implants. Moreover, it highlights recent advancements in the application of growth factors to the surface of titanium implants (Scheme 1). Finally, the review discusses current limitations and future prospects for growth factor-functionalized titanium implants. In summary, this paper presents cutting-edge design strategies aimed at enhancing the bone regenerative capacity of growth factor-functionalized titanium implants-a significant advancement in the field of enhanced bone regeneration.
Collapse
Affiliation(s)
- Zhenjia Che
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China.
| | - Qi Sun
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Zhenyu Zhao
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Yanglin Wu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Hu Xing
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Kaihang Song
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Aopan Chen
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Bo Wang
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China.
| | - Ming Cai
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China.
| |
Collapse
|
4
|
Wilkinson P, Jackson B, Fermor H, Davies R. A new mRNA structure prediction based approach to identifying improved signal peptides for bone morphogenetic protein 2. BMC Biotechnol 2024; 24:34. [PMID: 38783306 PMCID: PMC11112908 DOI: 10.1186/s12896-024-00858-1] [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: 01/26/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Signal peptide (SP) engineering has proven able to improve production of many proteins yet is a laborious process that still relies on trial and error. mRNA structure around the translational start site is important in translation initiation and has rarely been considered in this context, with recent improvements in in silico mRNA structure potentially rendering it a useful predictive tool for SP selection. Here we attempt to create a method to systematically screen candidate signal peptide sequences in silico based on both their nucleotide and amino acid sequences. Several recently released computational tools were used to predict signal peptide activity (SignalP), localization target (DeepLoc) and predicted mRNA structure (MXFold2). The method was tested with Bone Morphogenetic Protein 2 (BMP2), an osteogenic growth factor used clinically for bone regeneration. It was hoped more effective BMP2 SPs could improve BMP2-based gene therapies and reduce the cost of recombinant BMP2 production. RESULTS Amino acid sequence analysis indicated 2,611 SPs from the TGF-β superfamily were predicted to function when attached to BMP2. mRNA structure prediction indicated structures at the translational start site were likely highly variable. The five sequences with the most accessible translational start sites, a codon optimized BMP2 SP variant and the well-established hIL2 SP sequence were taken forward to in vitro testing. The top five candidates showed non-significant improvements in BMP2 secretion in HEK293T cells. All showed reductions in secretion versus the native sequence in C2C12 cells, with several showing large and significant decreases. None of the tested sequences were able to increase alkaline phosphatase activity above background in C2C12s. The codon optimized control sequence and hIL2 SP showed reasonable activity in HEK293T but very poor activity in C2C12. CONCLUSIONS These results support the use of peptide sequence based in silico tools for basic predictions around signal peptide activity in a synthetic biology context. However, mRNA structure prediction requires improvement before it can produce reliable predictions for this application. The poor activity of the codon optimized BMP2 SP variant in C2C12 emphasizes the importance of codon choice, mRNA structure, and cellular context for SP activity.
Collapse
Affiliation(s)
- Piers Wilkinson
- Department of Mechanical Engineering, Institute of Medical and Biological Engineering, University of Leeds, Leeds, UK.
| | - Brian Jackson
- Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Hazel Fermor
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Robert Davies
- Oral Biology, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| |
Collapse
|
5
|
Ganesan V, Ulgekar G, Ramalingam A, Sen Sharma S, Ganguli N, Majumdar SS. Goat mammary epithelial cells provide a better expression system for production of recombinant human bone morphogenetic protein 2 compared to Chinese hamster ovarian cells. Cell Biochem Funct 2024; 42:e3982. [PMID: 38488412 DOI: 10.1002/cbf.3982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/13/2024] [Accepted: 03/06/2024] [Indexed: 03/19/2024]
Abstract
Bone Morphogenetic Protein 2 (BMP2), a member of the Transforming Growth Factor-β (TGF-β) super family of proteins and is instrumental in the repair of fractures. The synthesis of BMP2 involves extensive post-translational processing and several studies have demonstrated the abysmally low production of rhBMP2 in eukaryotic systems, which may be due to the short half-life of the bioactive protein. Consequently, production costs of rhBMP2 are quite high, limiting its availability to the general populace. Therefore, there is an urgent need to identify better in-vitro systems for large scale production of rhBMP2. In the present study, we have carried out a comparative analysis of rhBMP2 production by the conventionally used Chinese Hamster ovarian cells (CHO) and goat mammary epithelial cells (GMEC), upon transfection with appropriate construct. Udder gland cells are highly secretory, and we reasoned that such cells may serve as a better in-vitro model for large scale production of rhBMP2. Our results indicated that the synthesis and secretion of bioactive rhBMP2 by goat mammary epithelial cells was significantly higher as compared to that by CHO-K1 cells. Our results provide strong evidence that GMECs may serve as a better alternative to other mammalian cells used for therapeutic protein production.
Collapse
Affiliation(s)
- Venkateswaran Ganesan
- National Institute of Animal Biotechnology, Hyderabad, Telangana, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Goutam Ulgekar
- National Institute of Animal Biotechnology, Hyderabad, Telangana, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | | | - Souvik Sen Sharma
- National Institute of Animal Biotechnology, Hyderabad, Telangana, India
| | - Nirmalya Ganguli
- National Institute of Animal Biotechnology, Hyderabad, Telangana, India
- Adjunct Faculty, Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Subeer S Majumdar
- National Institute of Animal Biotechnology, Hyderabad, Telangana, India
- Adjunct Faculty, Regional Centre for Biotechnology, Faridabad, Haryana, India
| |
Collapse
|
6
|
Phan A, MacKay JA. Steric stabilization of bioactive nanoparticles using elastin-like polypeptides. Adv Drug Deliv Rev 2024; 206:115189. [PMID: 38281625 PMCID: PMC11580827 DOI: 10.1016/j.addr.2024.115189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 01/30/2024]
Abstract
Elastin-like polypeptides (ELP) are versatile, thermo-responsive polymers that can be conjugated to virtually any therapeutic cargo. Derived from short amino-acid sequences and abundant in humans, certain ELPs display low immunogenicity. Substrates for endogenous proteases, ELPs are biodegradable and thus, are candidate biomaterials. Peptides and proteins can be directly coupled with ELPs through genetic engineering, while other polymers and small molecules can be appended through covalent bioconjugation or non-covalent complexation. ELPs that phase separate at physiological temperatures can form the core of nano assemblies; however, ELPs that remain soluble can sterically stabilize the corona of a variety of nanoparticles. Nanoparticles with ELPs at their corona promote colloids with favorable pharmacokinetic (PK) properties that enables therapeutic efficacy with intermittent administration. This review highlights a comprehensive spectrum of ELP fusions shown to stabilize the solubility, and sometimes bioactivity, of their cargo - with a focus on biophysical properties that underlie their therapeutic effects.
Collapse
Affiliation(s)
- Alvin Phan
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - J Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA; Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
7
|
Smith RAA, Luo X, Lu X, Tan TC, Le BQ, Zubkova OV, Tyler PC, Nurcombe V, Cool SM. Enhancing BMP-2-mediated osteogenesis with a synthetic heparan sulfate mimetic. BIOMATERIALS ADVANCES 2023; 155:213671. [PMID: 39492001 DOI: 10.1016/j.bioadv.2023.213671] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 10/06/2023] [Accepted: 10/22/2023] [Indexed: 11/05/2024]
Abstract
Bone morphogenetic protein 2 (BMP-2) is an osteoinductive protein and a potent inducers of bone formation, playing an essential role during bone fracture repair. Heparan sulfate (HS), a highly charged and linear polysaccharide, is known to interact with and enhance BMP-2 bioactivity. Despite showing potential as a potent adjuvant of the endogenous bone healing response, commercially available HS is derived from animal sources which are less desirable when considering translation into the clinic. In the present study, we screen twenty glycomimetics against BMP-2 to determine if fully synthetic analogues of HS can enhance the bioactivity of BMP-2 in vitro and bone healing in vivo. We found that a four-armed dendrimer harboring oversulfated maltose residues could bind BMP-2 with high affinity, enhance BMP-2 bioactivity in vitro and enhance bone regeneration in vivo. These data suggest fully synthetic glycomimetics are viable alternatives to naturally derived HS and offer an attractive alternative for clinical translation.
Collapse
Affiliation(s)
- Raymond A A Smith
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore; School of Chemical Engineering, The University of Queensland, Brisbane, Qld 4072, Australia.
| | - Xiaoman Luo
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Xiaohua Lu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Tuan Chun Tan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Bach Q Le
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Olga V Zubkova
- The Ferrier Research Institute, Victoria University of Wellington, Gracefield Research Centre, Lower Hutt, New Zealand
| | - Peter C Tyler
- The Ferrier Research Institute, Victoria University of Wellington, Gracefield Research Centre, Lower Hutt, New Zealand
| | - Victor Nurcombe
- Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - Simon M Cool
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore; Department of Orthopaedic Surgery, Yong Yoo Lin School of Medicine, National University of Singapore; School of Chemical Engineering, The University of Queensland, Brisbane, Qld 4072, Australia.
| |
Collapse
|
8
|
Dorogin J, Hochstatter HB, Shepherd SO, Svendsen JE, Benz MA, Powers AC, Fear KM, Townsend JM, Prell JS, Hosseinzadeh P, Hettiaratchi MH. Moderate-Affinity Affibodies Modulate the Delivery and Bioactivity of Bone Morphogenetic Protein-2. Adv Healthc Mater 2023; 12:e2300793. [PMID: 37379021 PMCID: PMC10592408 DOI: 10.1002/adhm.202300793] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/16/2023] [Indexed: 06/29/2023]
Abstract
Uncontrolled bone morphogenetic protein-2 (BMP-2) release can lead to off-target bone growth and other adverse events. To tackle this challenge, yeast surface display is used to identify unique BMP-2-specific protein binders known as affibodies that bind to BMP-2 with different affinities. Biolayer interferometry reveals an equilibrium dissociation constant of 10.7 nm for the interaction between BMP-2 and high-affinity affibody and 34.8 nm for the interaction between BMP-2 and the low-affinity affibody. The low-affinity affibody-BMP-2 interaction also exhibits an off-rate constant that is an order of magnitude higher. Computational modeling of affibody-BMP-2 binding predicts that the high- and low-affinity affibodies bind to two distinct sites on BMP-2 that function as different cell-receptor binding sites. BMP-2 binding to affibodies reduces expression of the osteogenic marker alkaline phosphatase (ALP) in C2C12 myoblasts. Affibody-conjugated polyethylene glycol-maleimide hydrogels increase uptake of BMP-2 compared to affibody-free hydrogels, and high-affinity hydrogels exhibit lower BMP-2 release into serum compared to low-affinity hydrogels and affibody-free hydrogels over four weeks. Loading BMP-2 into affibody-conjugated hydrogels prolongs ALP activity of C2C12 myoblasts compared to soluble BMP-2. This work demonstrates that affibodies with different affinities can modulate BMP-2 delivery and activity, creating a promising approach for controlling BMP-2 delivery in clinical applications.
Collapse
Affiliation(s)
- Jonathan Dorogin
- Department of BioengineeringKnight Campus for Accelerating Scientific ImpactUniversity of Oregon6231 University of OregonEugeneOR97403USA
| | - Henry B. Hochstatter
- Department of BioengineeringKnight Campus for Accelerating Scientific ImpactUniversity of Oregon6231 University of OregonEugeneOR97403USA
- Department of Human PhysiologyUniversity of Oregon1320 E 15th Ave.EugeneOR97403USA
| | - Samantha O. Shepherd
- Department of Chemistry and BiochemistryUniversity of Oregon1253 University of OregonEugeneOR97403USA
| | - Justin E. Svendsen
- Department of BioengineeringKnight Campus for Accelerating Scientific ImpactUniversity of Oregon6231 University of OregonEugeneOR97403USA
- Department of Chemistry and BiochemistryUniversity of Oregon1253 University of OregonEugeneOR97403USA
| | - Morrhyssey A. Benz
- Department of BioengineeringKnight Campus for Accelerating Scientific ImpactUniversity of Oregon6231 University of OregonEugeneOR97403USA
- Department of Chemistry and BiochemistryUniversity of Oregon1253 University of OregonEugeneOR97403USA
| | - Andrew C. Powers
- Department of BioengineeringKnight Campus for Accelerating Scientific ImpactUniversity of Oregon6231 University of OregonEugeneOR97403USA
| | - Karly M. Fear
- Department of BioengineeringKnight Campus for Accelerating Scientific ImpactUniversity of Oregon6231 University of OregonEugeneOR97403USA
| | - Jakob M. Townsend
- Department of BioengineeringKnight Campus for Accelerating Scientific ImpactUniversity of Oregon6231 University of OregonEugeneOR97403USA
| | - James S. Prell
- Department of Chemistry and BiochemistryUniversity of Oregon1253 University of OregonEugeneOR97403USA
| | - Parisa Hosseinzadeh
- Department of BioengineeringKnight Campus for Accelerating Scientific ImpactUniversity of Oregon6231 University of OregonEugeneOR97403USA
- Department of Chemistry and BiochemistryUniversity of Oregon1253 University of OregonEugeneOR97403USA
| | - Marian H. Hettiaratchi
- Department of BioengineeringKnight Campus for Accelerating Scientific ImpactUniversity of Oregon6231 University of OregonEugeneOR97403USA
- Department of Chemistry and BiochemistryUniversity of Oregon1253 University of OregonEugeneOR97403USA
| |
Collapse
|
9
|
Fang W, Yang M, Liu M, Jin Y, Wang Y, Yang R, Wang Y, Zhang K, Fu Q. Review on Additives in Hydrogels for 3D Bioprinting of Regenerative Medicine: From Mechanism to Methodology. Pharmaceutics 2023; 15:1700. [PMID: 37376148 PMCID: PMC10302687 DOI: 10.3390/pharmaceutics15061700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
The regeneration of biological tissues in medicine is challenging, and 3D bioprinting offers an innovative way to create functional multicellular tissues. One common way in bioprinting is bioink, which is one type of the cell-loaded hydrogel. For clinical application, however, the bioprinting still suffers from satisfactory performance, e.g., in vascularization, effective antibacterial, immunomodulation, and regulation of collagen deposition. Many studies incorporated different bioactive materials into the 3D-printed scaffolds to optimize the bioprinting. Here, we reviewed a variety of additives added to the 3D bioprinting hydrogel. The underlying mechanisms and methodology for biological regeneration are important and will provide a useful basis for future research.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kaile Zhang
- Department of Urology, Affiliated Sixth People’s Hospital, Shanghai Jiaotong University, No. 600 Yi-Shan Road, Shanghai 200233, China; (W.F.); (M.Y.)
| | - Qiang Fu
- Department of Urology, Affiliated Sixth People’s Hospital, Shanghai Jiaotong University, No. 600 Yi-Shan Road, Shanghai 200233, China; (W.F.); (M.Y.)
| |
Collapse
|
10
|
Wytrwal M, Sekuła-Stryjewska M, Pomorska A, Oclon E, Zuba-Surma E, Zapotoczny S, Szczubiałka K. Cellular Response to Bone Morphogenetic Proteins-2 and -7 Covalently Bound to Photocrosslinked Heparin-Diazoresin Multilayer. Biomolecules 2023; 13:biom13050842. [PMID: 37238712 DOI: 10.3390/biom13050842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/22/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Despite the plethora of research that exists on recombinant human bone morphogenetic protein-2 and -7 (rhBMP-2 and rhBMP-7) and has been clinically approved, there is still a need to gain information that would allow for their more rational use in bone implantology. The clinical application of supra-physiological dosages of these superactive molecules causes many serious adverse effects. At the cellular level, they play a role in osteogenesis and cellular adhesion, migration, and proliferation around the implant. Therefore, in this work, we investigated the role of the covalent binding of rhBMP-2 and rhBMP-7 separately and in combination with ultrathin multilayers composed of heparin and diazoresin in stem cells. In the first step, we optimized the protein deposition conditions via quartz crystal microbalance (QCM). Then, atomic force microscopy (AFM) and enzyme-linked immunosorbent assay (ELISA) were used to analyze protein-substrate interactions. The effect of the protein binding on the initial cell adhesion, migration, and short-term expression of osteogenesis markers was tested. In the presence of both proteins, cell flattening and adhesion became more prominent, resulting in limited motility. However, the early osteogenic marker expression significantly increased compared to the single protein systems. The presence of single proteins resulted in the elongation of cells, which promoted their migration activity.
Collapse
Affiliation(s)
- Magdalena Wytrwal
- Academic Centre for Materials and Nanotechnology, AGH University of Science and Technology, al. A. Mickiewicza 30, 30-059 Krakow, Poland
| | | | - Agata Pomorska
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Niezapominajek 8, 30-239 Krakow, Poland
| | - Ewa Oclon
- Laboratory of Recombinant Proteins Production, Centre for Experimental and Innovative Medicine, University of Agriculture in Krakow, 1C Redzina Street, 30-248 Krakow, Poland
| | - Ewa Zuba-Surma
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Szczepan Zapotoczny
- Academic Centre for Materials and Nanotechnology, AGH University of Science and Technology, al. A. Mickiewicza 30, 30-059 Krakow, Poland
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Krzysztof Szczubiałka
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| |
Collapse
|
11
|
Li C, Wang J, Niu Y, Zhang H, Ouyang H, Zhang G, Fu Y. Baicalin Nanocomplexes with an In Situ-Forming Biomimetic Gel Implant for Repair of Calvarial Bone Defects via Localized Sclerostin Inhibition. ACS APPLIED MATERIALS & INTERFACES 2023; 15:9044-9057. [PMID: 36753285 DOI: 10.1021/acsami.2c20946] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
In situ-forming hydrogels are highly effective in covering complex and irregular tissue defects. Herein, a biomimetic gel implant (CS-GEL) consisting of methacrylated chondroitin sulfate and gelatin is obtained via visible light irradiation, which displays rapid gelation (∼30 s), suitable mechanical properties, and biological features to support osteoblast attachment and proliferation. Sclerostin is proven to be a viable target to promote osteogenesis. Hence, baicalin, a natural flavonoid with a high affinity to sclerostin, is selected as the therapeutic compound to achieve localized neutralization of sclerostin. To overcome its poor solubility and permeability, a baicalin nanocomplex (BNP) is synthesized using Solutol HS15, which is then dispersed in the CS-GEL to afford a nanocomposite delivery system, i.e., BNP-loaded gel (BNP@CS-GEL). In vitro, BNP significantly downregulated the level of sclerostin in MLO-Y4 osteocytes. In vivo, either CS-GEL or BNP@CS-GEL is proven to effectively promote osteogenesis and angiogenesis in a calvarial critical-sized bone defect rat model, with BNP@CS-GEL showing the best pro-healing effect. Specifically, the BNP@CS-GEL-treated group significantly downregulated the sclerostin level as compared to the sham group (p < 0.05). RANKL expression was also significantly suppressed by BNP in MLO-Y4 cells and BNP@CS-GEL in vivo. Collectively, our study offers a facile and viable gel platform in combination with nanoparticulated baicalin for the localized neutralization of sclerostin to promote bone regeneration and repair.
Collapse
Affiliation(s)
- Chenrui Li
- Key Laboratory for Space Biosciences & Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, China
| | - Junru Wang
- Key Laboratory for Space Biosciences & Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, China
| | - Yining Niu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Haonan Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Hongling Ouyang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Guangwei Zhang
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Department of Public Health & College of Clinical Medicine, Xi'an Medical University, Xi'an 710021, Shaanxi, China
| | - Yao Fu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
12
|
Fok SW, Gresham RCH, Ryan W, Osipov B, Bahney C, Leach JK. Macromolecular crowding and decellularization method increase the growth factor binding potential of cell-secreted extracellular matrices. Front Bioeng Biotechnol 2023; 11:1091157. [PMID: 36756385 PMCID: PMC9899907 DOI: 10.3389/fbioe.2023.1091157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 01/13/2023] [Indexed: 01/25/2023] Open
Abstract
Recombinant growth factors are used in tissue engineering to stimulate cell proliferation, migration, and differentiation. Conventional methods of growth factor delivery for therapeutic applications employ large amounts of these bioactive cues. Effective, localized growth factor release is essential to reduce the required dose and potential deleterious effects. The endogenous extracellular matrix (ECM) sequesters native growth factors through its negatively charged sulfated glycosaminoglycans. Mesenchymal stromal cells secrete an instructive extracellular matrix that can be tuned by varying culture and decellularization methods. In this study, mesenchymal stromal cell-secreted extracellular matrix was modified using λ-carrageenan as a macromolecular crowding (MMC) agent and decellularized with DNase as an alternative to previous decellularized extracellular matrices (dECM) to improve growth factor retention. Macromolecular crowding decellularized extracellular matrix contained 7.7-fold more sulfated glycosaminoglycans and 11.7-fold more total protein than decellularized extracellular matrix, with no significant difference in residual DNA. Endogenous BMP-2 was retained in macromolecular crowding decellularized extracellular matrix, whereas BMP-2 was not detected in other extracellular matrices. When implanted in a murine muscle pouch, we observed increased mineralized tissue formation with BMP-2-adsorbed macromolecular crowding decellularized extracellular matrix in vivo compared to conventional decellularized extracellular matrix. This study demonstrates the importance of decellularization method to retain endogenous sulfated glycosaminoglycans in decellularized extracellular matrix and highlights the utility of macromolecular crowding to upregulate sulfated glycosaminoglycan content. This platform has the potential to aid in the delivery of lower doses of BMP-2 or other heparin-binding growth factors in a tunable manner.
Collapse
Affiliation(s)
- Shierly W. Fok
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, United States
| | - Robert C. H. Gresham
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, United States
| | - Weston Ryan
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, United States
| | - Benjamin Osipov
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, United States
| | - Chelsea Bahney
- Steadman Philippon Research Institute, Vail, CO, United States
| | - J. Kent Leach
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, United States,Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States,*Correspondence: J. Kent Leach,
| |
Collapse
|
13
|
Seyedi Z, Amiri MS, Mohammadzadeh V, Hashemzadeh A, Haddad-Mashadrizeh A, Mashreghi M, Qayoomian M, Hashemzadeh MR, Simal-Gandara J, Taghavizadeh Yazdi ME. Icariin: A Promising Natural Product in Biomedicine and Tissue Engineering. J Funct Biomater 2023; 14:44. [PMID: 36662090 PMCID: PMC9862744 DOI: 10.3390/jfb14010044] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 01/14/2023] Open
Abstract
Among scaffolds used in tissue engineering, natural biomaterials such as plant-based materials show a crucial role in cellular function due to their biocompatibility and chemical indicators. Because of environmentally friendly behavior and safety, green methods are so important in designing scaffolds. A key bioactive flavonoid of the Epimedium plant, Icariin (ICRN), has a broad range of applications in improving scaffolds as a constant and non-immunogenic material, and in stimulating the cell growth, differentiation of chondrocytes as well as differentiation of embryonic stem cells towards cardiomyocytes. Moreover, fusion of ICRN into the hydrogel scaffolds or chemical crosslinking can enhance the secretion of the collagen matrix and proteoglycan in bone and cartilage tissue engineering. To scrutinize, in various types of cancer cells, ICRN plays a decisive role through increasing cytochrome c secretion, Bax/Bcl2 ratio, poly (ADP-ribose) polymerase as well as caspase stimulations. Surprisingly, ICRN can induce apoptosis, reduce viability and inhibit proliferation of cancer cells, and repress tumorigenesis as well as metastasis. Moreover, cancer cells no longer grow by halting the cell cycle at two checkpoints, G0/G1 and G2/M, through the inhibition of NF-κB by ICRN. Besides, improving nephrotoxicity occurring due to cisplatin and inhibiting multidrug resistance are the other applications of this biomaterial.
Collapse
Affiliation(s)
- Zahra Seyedi
- Department of Stem Cells and Regenerative Medicine, Royesh Stem Cell Biotechnology Institute, Mashhad 9188758156, Iran
- Department of Cancer and Oncology, Royesh Stem Cell Biotechnology Institute, Mashhad 9188758156, Iran
| | | | - Vahideh Mohammadzadeh
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 91778, Iran
| | - Alireza Hashemzadeh
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 91778, Iran
| | - Aliakbar Haddad-Mashadrizeh
- Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad 9177948974, Iran
| | - Mohammad Mashreghi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 91778, Iran
| | - Mohsen Qayoomian
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad 91778, Iran
| | - Mohammad Reza Hashemzadeh
- Department of Stem Cells and Regenerative Medicine, Royesh Stem Cell Biotechnology Institute, Mashhad 9188758156, Iran
- Department of Cancer and Oncology, Royesh Stem Cell Biotechnology Institute, Mashhad 9188758156, Iran
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Department of Analytical and Food Chemistry, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E32004 Ourense, Spain
| | | |
Collapse
|
14
|
Ma L, Fu L, Gu C, Wang H, Yu Z, Gao X, Zhao D, Ge B, Zhang N. Delivery of bone morphogenetic protein-2 by crosslinking heparin to nile tilapia skin collagen for promotion of rat calvaria bone defect repair. Prog Biomater 2022; 12:61-73. [PMID: 36495399 PMCID: PMC9958213 DOI: 10.1007/s40204-022-00213-7] [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: 05/06/2022] [Accepted: 11/26/2022] [Indexed: 12/14/2022] Open
Abstract
Collagen has been widely used as a biomaterial for tissue regeneration. At the present, aqua-collagen derived from fish is poorly explored for biomedical material applications due to its insufficient thermal stability. To improve the bone repair ability and thermal stability of fish collagen, the tilapia skin collagen was crosslinked by EDC/NHS with heparin to bind specifically to BMP-2. The thermal stability of tilapia skin collagen crosslinked with heparin (HC-COL) was detected by differential scanning calorimetry (DSC). Cytotoxicity of HC-COL was assessed by detecting MC3T3-E1 cell proliferation using CCK-8 assay. The specific binding of BMP-2 to HC-COL was tested and the bioactivity of BMP-2-loaded HC-COL (HC-COL-BMP-2) was evaluated in vitro by inducing MC3T3-E1 cell differentiation. In vivo, the bone repair ability of HC-COL-2 was evaluated using micro-CT and histological observation. After crosslinking by EDC/NHS, the heparin-linked and the thermostability of the collagen of Nile Tilapia were improved simultaneously. HC-COL has no cytotoxicity. In addition, the binding of BMP-2 to HC-COL was significantly increased. Furthermore, the in vitro study revealed the effective bioactivity of BMP-2 binding on HC-COL by inducing MC3T3-E1 cells with higher ALP activity and the formation of mineralized nodules. In vivo studies showed that more mineralized and mature bone formation was achieved in HC-COL-BMP-2 group. The prepared HC-COL was an effective BMP-2 binding carrier with enough thermal stability and could be a useful biomaterial for bone repair.
Collapse
Affiliation(s)
- Lina Ma
- grid.440653.00000 0000 9588 091XDepartment of Diagnostics, The Second School of Medicine, Binzhou Medical University, Laishan, Yantai, 264003 Shandong China ,grid.440653.00000 0000 9588 091XRongxiang Xu Regenerative Medicine Research Center, Binzhou Medical University, Laishan, Yantai, 264003 Shandong China
| | - Li Fu
- grid.440653.00000 0000 9588 091XRongxiang Xu Regenerative Medicine Research Center, Binzhou Medical University, Laishan, Yantai, 264003 Shandong China ,grid.440653.00000 0000 9588 091XDepartment of Human Anatomy, School of Basic MedicalScience, Binzhou Medical University, Laishan, Yantai, 264003 Shandong China
| | - Chengxu Gu
- grid.440653.00000 0000 9588 091XDepartment of Human Anatomy, School of Basic MedicalScience, Binzhou Medical University, Laishan, Yantai, 264003 Shandong China
| | - Haonan Wang
- grid.497420.c0000 0004 1798 1132State Key Laboratory of Heavy Oil Processing and Center for Bioengineering and Biotechnology, China University of Petroleum (East China), Qingdao, 266580 People’s Republic of China
| | - Zhenghai Yu
- grid.440653.00000 0000 9588 091XDepartment of Human Anatomy, School of Basic MedicalScience, Binzhou Medical University, Laishan, Yantai, 264003 Shandong China
| | - Xiuwei Gao
- Shandong Junxiu Biotechnology Co. LTD, 32 Zhujiang Road, Economic and Technological Development Zone, Yantai, 264006 Shandong China
| | - Dongmei Zhao
- Department of Human Anatomy, School of Basic MedicalScience, Binzhou Medical University, Laishan, Yantai, 264003, Shandong, China.
| | - Baosheng Ge
- State Key Laboratory of Heavy Oil Processing and Center for Bioengineering and Biotechnology, China University of Petroleum (East China), Qingdao, 266580, People's Republic of China.
| | - Naili Zhang
- Rongxiang Xu Regenerative Medicine Research Center, Binzhou Medical University, Laishan, Yantai, 264003, Shandong, China. .,Department of Human Anatomy, School of Basic MedicalScience, Binzhou Medical University, Laishan, Yantai, 264003, Shandong, China.
| |
Collapse
|
15
|
Hu J, Wang Z, Miszuk JM, Zeng E, Sun H. High Molecular Weight Poly(glutamic acid) to Improve BMP2-Induced Osteogenic Differentiation. Mol Pharm 2022; 19:4565-4575. [PMID: 35675584 PMCID: PMC9729371 DOI: 10.1021/acs.molpharmaceut.2c00141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
FDA-approved bone morphogenetic protein 2 (BMP2) has serious side effects due to the super high dose requirement. Heparin is one of the most well-studied sulfated polymers to stabilize BMP2 and improve its functionality. However, the clinical use of heparin is questionable because of its undesired anticoagulant activity. Recent studies suggest that poly(glutamic acid) (pGlu) has the potential to improve BMP2 bioactivity with less safety concerns; however, the knowledge on pGlu's contribution remains largely unknown. Therefore, we aimed to study the role of pGlu in BMP2-induced osteogenesis and its potential application in bone tissue engineering. Our data, for the first time, indicated that both low (L-pGlu) and high molecular weight pGlu (H-pGlu) were able to significantly improve the BMP2-induced early osteoblastic differentiation marker (ALP) in MC3T3-E1 preosteoblasts. Importantly, the matrix mineralization was more rapidly enhanced by H-pGlu compared to L-pGlu. Additionally, our data indicated that only α-H-pGlu could significantly improve BMP2's activity, whereas γ-H-pGlu failed to do so. Moreover, both gene expression and mineralization data demonstrated that α-H-pGlu enabled a single dose of BMP2 to induce a high level of osteoblastic differentiation without multiple doses of BMP2. To study the potential application of pGlu in tissue engineering, we incorporated the H-pGlu+BMP2 nanocomplexes into the collagen hydrogel with significantly elevated osteoblastic differentiation. Furthermore, H-pGlu-coated 3D porous gelatin and chitosan scaffolds significantly enhanced osteogenic differentiation through enabling sustained release of BMP2. Thus, our findings suggest that H-pGlu is a promising new alternative with great potential for bone tissue engineering applications.
Collapse
Affiliation(s)
- Jue Hu
- Department of Oral and Maxillofacial Surgery, University of Iowa College of Dentistry, Iowa City, IA 52242, USA
- Iowa Institute for Oral Health Research, University of Iowa College of Dentistry, Iowa City, IA 52242, USA
| | - Zhuozhi Wang
- Department of Oral and Maxillofacial Surgery, University of Iowa College of Dentistry, Iowa City, IA 52242, USA
- Iowa Institute for Oral Health Research, University of Iowa College of Dentistry, Iowa City, IA 52242, USA
| | - Jacob M. Miszuk
- Department of Oral and Maxillofacial Surgery, University of Iowa College of Dentistry, Iowa City, IA 52242, USA
- Iowa Institute for Oral Health Research, University of Iowa College of Dentistry, Iowa City, IA 52242, USA
| | - Erliang Zeng
- Iowa Institute for Oral Health Research, University of Iowa College of Dentistry, Iowa City, IA 52242, USA
| | - Hongli Sun
- Department of Oral and Maxillofacial Surgery, University of Iowa College of Dentistry, Iowa City, IA 52242, USA
- Iowa Institute for Oral Health Research, University of Iowa College of Dentistry, Iowa City, IA 52242, USA
| |
Collapse
|
16
|
Rather HA, Varghese JF, Dhimmar B, Yadav UC, Vasita R. Polycaprolactone-collagen nanofibers loaded with dexamethasone and simvastatin as an osteoinductive and immunocompatible scaffold for bone regeneration applications. BIOMATERIALS AND BIOSYSTEMS 2022; 8:100064. [PMID: 36824372 PMCID: PMC9934467 DOI: 10.1016/j.bbiosy.2022.100064] [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: 04/16/2022] [Revised: 09/25/2022] [Accepted: 09/26/2022] [Indexed: 11/29/2022] Open
Abstract
Physiological inflammation has been shown to promote bone regeneration; however, prolonged inflammation impedes the osteogenesis and bone repair process. To overcome the latter we aimed to develop a dual drug delivering nanofibrous scaffold to promote osteogenic differentiation of mesenchymal stromal cells (MSCs) and modulate the pro-inflammatory response of macrophages. The polycaprolactone (PCL)-collagen nanofibrous delivery system incorporating dexamethasone and simvastatin was fabricated by electrospinning process. The morphological analysis and mRNA, as well as protein expression of proinflammatory and anti-inflammatory cytokines in human monocytes (U937 cells), demonstrated the immunocompatibility effect of dual drug-releasing nanofibrous scaffolds. Nitric oxide estimation also demonstrated the anti-inflammatory effect of dual drug releasing scaffolds. The scaffolds demonstrated the osteogenic differentiation of adipose-derived MSCs by enhancing the alkaline phosphatase (ALP) activity and mineral deposition after 17 days of cell culture. The increased expression of Runt-related transcription factor-2 (RUNX-2) and osteocalcin at mRNA and protein levels supported the osteogenic potential of dual drug-loaded fibrous scaffolds. Hence, the results indicate that our fabricated nanofibrous scaffolds exhibit immunomodulatory properties and could be employed for bone regeneration applications after further in-vivo validation.
Collapse
Affiliation(s)
- Hilal Ahmad Rather
- Biomaterials & Biomimetics Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, 382030, India
| | | | - Bindiya Dhimmar
- Biomaterials & Biomimetics Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, 382030, India
| | - Umesh C.S. Yadav
- Metabolic Disorders and Inflammatory pathologies Laboratory, Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Rajesh Vasita
- Biomaterials & Biomimetics Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, 382030, India,Corresponding author.
| |
Collapse
|
17
|
Gultian KA, Gandhi R, DeCesari K, Romiyo V, Kleinbart EP, Martin K, Gentile PM, Kim TWB, Vega SL. Injectable hydrogel with immobilized BMP-2 mimetic peptide for local bone regeneration. FRONTIERS IN BIOMATERIALS SCIENCE 2022; 1. [PMID: 37090104 PMCID: PMC10120851 DOI: 10.3389/fbiom.2022.948493] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Osteoporosis is a disease characterized by a decrease in bone mineral density, thereby increasing the risk of sustaining a fragility fracture. Most medical therapies are systemic and do not restore bone in areas of need, leading to undesirable side effects. Injectable hydrogels can locally deliver therapeutics with spatial precision, and this study reports the development of an injectable hydrogel containing a peptide mimic of bone morphogenetic protein-2 (BMP-2). To create injectable hydrogels, hyaluronic acid was modified with norbornene (HANor) or tetrazine (HATet) which upon mixing click into covalently crosslinked Nor-Tet hydrogels. By modifying HANor macromers with methacrylates (Me), thiolated BMP-2 mimetic peptides were immobilized to HANor via a Michael addition reaction, and coupling was confirmed with 1H NMR spectroscopy. BMP-2 peptides presented in soluble and immobilized form increased alkaline phosphatase (ALP) expression in MSCs cultured on 2D and encapsulated in 3D Nor-Tet hydrogels. Injection of bioactive Nor-Tet hydrogels into hollow intramedullary canals of Lewis rat femurs showed a local increase in trabecular bone density as determined by micro-CT imaging. The presented work shows that injectable hydrogels with immobilized BMP-2 peptides are a promising biomaterial for the local regeneration of bone tissue and for the potential local treatment of osteoporosis.
Collapse
Affiliation(s)
- Kirstene A. Gultian
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ, United States
| | - Roshni Gandhi
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ, United States
| | - Kayla DeCesari
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ, United States
| | - Vineeth Romiyo
- Department of Orthopaedic Surgery, Cooper University Health Care, Camden, NJ, United States
| | - Emily P. Kleinbart
- Department of Orthopaedic Surgery, Cooper University Health Care, Camden, NJ, United States
| | - Kelsey Martin
- Department of Orthopaedic Surgery, Cooper University Health Care, Camden, NJ, United States
| | - Pietro M. Gentile
- Department of Orthopaedic Surgery, Cooper University Health Care, Camden, NJ, United States
| | - Tae Won B. Kim
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ, United States
- Department of Orthopaedic Surgery, Cooper University Health Care, Camden, NJ, United States
| | - Sebastián L. Vega
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ, United States
- CORRESPONDENCE Sebastián L. Vega,
| |
Collapse
|
18
|
Zhang W, Wu Y, Chen Q, Zhang H, Zhou M, Chen K, Cao C, Guo H, Xu J, Liu H, Lin H, Liu C, Liu R. Statistic Copolymers Working as Growth Factor-Binding Mimics of Fibronectin. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200775. [PMID: 35570405 PMCID: PMC9313494 DOI: 10.1002/advs.202200775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/06/2022] [Indexed: 05/07/2023]
Abstract
Growth factors (GFs) play important roles in biological system and are widely used in tissue regeneration. However, their application is greatly hindered by short in vivo lifetime of GFs. GFs are bound to fibronectin dynamically in the extracellular matrix, which inspired the authors to mimic the GF binding domain of fibronectin and design GF-binding amphiphilic copolymers bearing positive charges. The optimal amino acid polymer can bind to a variety of representative GFs, such as bone morphogenetic protein-2 (BMP-2) and TGF-β1 from the transforming growth factor-β superfamily, PDGF-AA and PDGF-BB from the platelet-derived growth factor family, FGF-10 and FGF-21 from the fibroblast growth factor family, epidermal growth factor from the EGF family and hepatocyte growth factor from the plasminogen-related growth factor family, with binding affinities up to the nanomolar level. 3D scaffolds immobilized with the optimal copolymer enable sustained release of loaded BMP-2 without burst release and significantly enhances the in vivo function of BMP-2 for bone formation. This strategy opens new avenues in designing GF-binding copolymers as synthetic mimics of fibronectin for diverse applications.
Collapse
Affiliation(s)
- Wenjing Zhang
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Yueming Wu
- Key Laboratory for Ultrafine Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistryResearch Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Qi Chen
- Key Laboratory for Ultrafine Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistryResearch Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Haodong Zhang
- Key Laboratory for Ultrafine Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistryResearch Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Min Zhou
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Kang Chen
- Key Laboratory for Ultrafine Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistryResearch Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Chuntao Cao
- Key Laboratory for Ultrafine Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistryResearch Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Han Guo
- Shanghai Synchrotron Radiation Facility (SSRF)Shanghai Advanced Research InstituteChinese Academy of SciencesShanghai201204China
| | - Jianrong Xu
- Academy of Integrative MedicineShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Honglai Liu
- School of Chemistry and Molecular EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Haodong Lin
- Department of Orthopedic SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistryResearch Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Runhui Liu
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghai200237China
- Key Laboratory for Ultrafine Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistryResearch Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| |
Collapse
|
19
|
Jeon EY, Um SH, Park J, Jung Y, Cheon CH, Jeon H, Chung JJ. Precisely Localized Bone Regeneration Mediated by Marine-Derived Microdroplets with Superior BMP-2 Binding Affinity. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2200416. [PMID: 35543974 DOI: 10.1002/smll.202200416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/20/2022] [Indexed: 06/15/2023]
Abstract
Prompt and robust bone regeneration has been clinically achieved using supraphysiological doses of bone morphogenetic protein-2 (BMP-2) to overcome the short half-life and rapid clearance. However, uncontrolled burst release of exogenous BMP-2 causes severe complications such as heterotopic ossification and soft tissue inflammation. Therefore, numerous researches have focused on developing a new BMP-2 delivery system for a sustained release profile by immobilizing BMP-2 in various polymeric vehicles. Herein, to avoid denaturation of BMP-2 and enhance therapeutic action via localized delivery, a complex coacervate consisting of fucoidan, a marine-derived glycosaminoglycan, and poly-l-lysine (PLL) is fabricated. Superior BMP-2 binding ability and electrostatic interaction-driven engulfment enable facile and highly efficient microencapsulation of BMP-2. The microencapsulation ability of the coacervate significantly improves BMP-2 bioactivity and provides protection against antagonist and proteolysis, while allowing prolonged release. Moreover, BMP-2 containing coacervate is coated on conventional collagen sponges. The bioactivity and localized bone regenerating ability are confirmed through in vitro (human-derived stem cells), and in vivo (calvarial bone defect model) evaluations.
Collapse
Affiliation(s)
- Eun Young Jeon
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Seung-Hoon Um
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Jaeho Park
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Department of Materials Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Youngmee Jung
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Yonsei-KIST Convergence Research Institute, Seoul, 03722, Republic of Korea
| | - Cheol-Hong Cheon
- Department of Chemistry, Korea University, Seoul, 02841, Republic of Korea
| | - Hojeong Jeon
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Justin J Chung
- Transdisciplinary Department of Medicine and Advanced Technology, Seoul National University Hospital, Seoul, 03080, Republic of Korea
- Department of Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| |
Collapse
|
20
|
Sharma T, Kapoor A, Mandal CC. Duality of bone morphogenetic proteins in cancer: A comprehensive analysis. J Cell Physiol 2022; 237:3127-3163. [DOI: 10.1002/jcp.30785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/06/2022] [Accepted: 04/29/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Tanu Sharma
- Department of Biochemistry, School of Life Sciences Central University of Rajasthan Ajmer Rajasthan India
| | - Anmol Kapoor
- Department of Biochemistry, School of Life Sciences Central University of Rajasthan Ajmer Rajasthan India
| | - Chandi C. Mandal
- Department of Biochemistry, School of Life Sciences Central University of Rajasthan Ajmer Rajasthan India
| |
Collapse
|
21
|
Wang J, Xiao L, Wang W, Zhang D, Ma Y, Zhang Y, Wang X. The Auxiliary Role of Heparin in Bone Regeneration and its Application in Bone Substitute Materials. Front Bioeng Biotechnol 2022; 10:837172. [PMID: 35646879 PMCID: PMC9133562 DOI: 10.3389/fbioe.2022.837172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/13/2022] [Indexed: 11/18/2022] Open
Abstract
Bone regeneration in large segmental defects depends on the action of osteoblasts and the ingrowth of new blood vessels. Therefore, it is important to promote the release of osteogenic/angiogenic growth factors. Since the discovery of heparin, its anticoagulant, anti-inflammatory, and anticancer functions have been extensively studied for over a century. Although the application of heparin is widely used in the orthopedic field, its auxiliary effect on bone regeneration is yet to be unveiled. Specifically, approximately one-third of the transforming growth factor (TGF) superfamily is bound to heparin and heparan sulfate, among which TGF-β1, TGF-β2, and bone morphogenetic protein (BMP) are the most common growth factors used. In addition, heparin can also improve the delivery and retention of BMP-2 in vivo promoting the healing of large bone defects at hyper physiological doses. In blood vessel formation, heparin still plays an integral part of fracture healing by cooperating with the platelet-derived growth factor (PDGF). Importantly, since heparin binds to growth factors and release components in nanomaterials, it can significantly facilitate the controlled release and retention of growth factors [such as fibroblast growth factor (FGF), BMP, and PDGF] in vivo. Consequently, the knowledge of scaffolds or delivery systems composed of heparin and different biomaterials (including organic, inorganic, metal, and natural polymers) is vital for material-guided bone regeneration research. This study systematically reviews the structural properties and auxiliary functions of heparin, with an emphasis on bone regeneration and its application in biomaterials under physiological conditions.
Collapse
Affiliation(s)
- Jing Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Lan Xiao
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia
- Australia−China Centre for Tissue Engineering and Regenerative Medicine, Brisbane, Australia
| | - Weiqun Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Dingmei Zhang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yaping Ma
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yi Zhang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi, China
| | - Xin Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia
- Australia−China Centre for Tissue Engineering and Regenerative Medicine, Brisbane, Australia
| |
Collapse
|
22
|
Gélébart P, Cuenot S, Sinquin C, Halgand B, Sourice S, Le Visage C, Guicheux J, Colliec-Jouault S, Zykwinska A. Microgels based on Infernan, a glycosaminoglycan-mimetic bacterial exopolysaccharide, as BMP-2 delivery systems. Carbohydr Polym 2022; 284:119191. [DOI: 10.1016/j.carbpol.2022.119191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023]
|
23
|
Kim S, Fan J, Lee CS, Chen C, Lee M. Sulfonate Hydrogel-siRNA Conjugate Facilitates Osteogenic Differentiation of Mesenchymal Stem Cells by Controlled Gene Silencing and Activation of BMP Signaling. ACS APPLIED BIO MATERIALS 2021; 4:5189-5200. [PMID: 34661086 DOI: 10.1021/acsabm.1c00369] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hydrogels have been widely used in bone tissue engineering due to their tunable characteristics that allow facile modifications with various biochemical properties to support cell growth and guide proper cell functions. Herein, we report a design of hydrogel-siRNA conjugate that facilitates osteogenesis via gene silencing and activation of bone morphogenetic protein (BMP) signaling. A sulfonate hydrogel is prepared by modifying chitosan with sulfoacetic acid to mimic a natural sulfated polysaccharide and to provide a hydrogel surface that enables BMP binding. Then, siRNA targeting noggin, an endogenous extracellular antagonist of BMP signaling, is covalently conjugated to the sulfonate hydrogel by visible blue light crosslinking. The sulfonate hydrogel-siRNA conjugate is efficient to bind BMPs and also successfully prolongs the release of siRNA for sustained noggin suppression, thereby resulting in significantly increased osteogenic differentiation. Lastly, demineralized bone matrix (DBM) is incorporated into the sulfonate hydrogel-siRNA conjugate, wherein the DBM incorporation induces noggin expression via a negative feedback mechanism that regulates BMP signaling in DBM. However, simultaneous delivery of siRNA downregulates noggin thus facilitating endogenous BMP activity and enhancing the osteogenic efficacy of DBM. These findings support a promising hydrogel RNA silencing platform for bone tissue engineering applications.
Collapse
Affiliation(s)
- Soyon Kim
- Division of Advanced Prosthodontics, University of California, Los Angeles, USA
| | - Jiabing Fan
- Division of Advanced Prosthodontics, University of California, Los Angeles, USA
| | - Chung-Sung Lee
- Division of Advanced Prosthodontics, University of California, Los Angeles, USA
| | - Chen Chen
- Division of Advanced Prosthodontics, University of California, Los Angeles, USA
| | - Min Lee
- Division of Advanced Prosthodontics, University of California, Los Angeles, USA.,Department of Bioengineering, University of California, Los Angeles, USA
| |
Collapse
|
24
|
Kim J, Lee S, Choi Y, Choi J, Kang BJ. Sustained Release of Bone Morphogenetic Protein-2 through Alginate Microbeads Enhances Bone Regeneration in Rabbit Tibial Metaphyseal Defect Model. MATERIALS (BASEL, SWITZERLAND) 2021; 14:2600. [PMID: 34067593 PMCID: PMC8156783 DOI: 10.3390/ma14102600] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 12/03/2022]
Abstract
Bone morphogenetic protein-2 (BMP-2) is widely used to enhance bone regeneration. However, because of its short half-life and rapid disappearance, large amounts of BMP-2 are needed, leading to unintended side effects. In this study, BMP-2-encapsulated alginate microbeads (AM) were used to enhance bone regeneration. Enzyme-linked immunosorbent assay confirmed the sustained release of BMP-2 from AM. Vascular endothelial growth factor (VEGF)-adsorbing aptamer-conjugated hydroxyapatite (Apt-HA) was used for osteoconduction and dual delivery of VEGF and BMP-2. For in vivo bone regeneration evaluation, the grafts (1) Apt-HA + phosphate-buffered saline (PBS), (2) Apt-HA + AM without BMP-2, (3) Apt-HA + BMP-2, and (4) Apt-HA + AM encapsulated with BMP-2 were implanted into rabbit tibial metaphyseal defects. After four weeks, micro-computed tomography (CT), histological, and histomorphometric analyses were performed to evaluate bone regeneration. The Apt-HA + AM with BMP-2 group revealed a significantly higher new bone volume and bone volume/total volume (BV/TV) in both cortical and trabecular bone than the others. Furthermore, as evaluated by histomorphometric analysis, BMP-2 AM exhibited a significantly higher bone formation area than the others, indicating that AM could be used to efficiently deliver BMP-2 through sustained release. Moreover, the combined application of BMP-2-encapsulated Apt-HA + AM may effectively promote bone regeneration.
Collapse
Affiliation(s)
- Junhyung Kim
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea;
| | - Seoyun Lee
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea;
- BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, Seoul 08826, Korea
| | - Yonghyun Choi
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea; (Y.C.); (J.C.)
| | - Jonghoon Choi
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea; (Y.C.); (J.C.)
| | - Byung-Jae Kang
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea;
- BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
25
|
Albashari AA, He Y, Albaadani MA, Xiang Y, Ali J, Hu F, Zhang Y, Zhang K, Luo L, Wang J, Ye Q. Titanium Nanotube Modified With Silver Cross-Linked Basic Fibroblast Growth Factor Improves Osteoblastic Activities of Dental Pulp Stem Cells and Antibacterial Effect. Front Cell Dev Biol 2021; 9:654654. [PMID: 33869214 PMCID: PMC8047219 DOI: 10.3389/fcell.2021.654654] [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: 01/17/2021] [Accepted: 03/04/2021] [Indexed: 11/25/2022] Open
Abstract
Titanium modifications with different silver loading methods demonstrate excellent antibacterial properties. Yet pure silver nanoparticles with limited bioactive properties may delay regeneration of bone surrounding the dental implant. Therefore, loading silver with bioactive drugs on titanium surfaces seems to be a very promising strategy. Herein, we designed a silver (Ag) step-by-step cross-linking with the basic fibroblast growth factor (bFGF) by polydopamine (PDA) and heparin on titanium nanotube (TNT) as its cargo (TNT/PDA/Ag/bFGF) to improve the implant surface. Our results showed that TNT/PDA/Ag/bFGF significantly enhanced the osteogenic differentiation of dental pulp stem cells (DPSCs). It also showed an excellent effect in bacterial inhibition and a reduction of pro-inflammatory factors through inhibition of M1 macrophage activity. These results showed that bFGF cross-linked silver coating on TNTs presented good osteogenic differentiation and early anti-infiammatory and antibacterial properties. Together, this novel design on titanium provides a promising therapeutic for dental implants.
Collapse
Affiliation(s)
| | - Yan He
- Laboratory for Regenerative Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | | | - Yangfan Xiang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Jihea Ali
- College of Life and Environmental Science, Wenzhou University, Wenzhou, China
| | - Fengting Hu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yuan Zhang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Keke Zhang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Lihua Luo
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Jianming Wang
- Laboratory for Regenerative Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Qingsong Ye
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.,Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
26
|
Han S, Paeng KW, Park S, Jung UW, Cha JK, Hong J. Programmed BMP-2 release from biphasic calcium phosphates for optimal bone regeneration. Biomaterials 2021; 272:120785. [PMID: 33819813 DOI: 10.1016/j.biomaterials.2021.120785] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 03/20/2021] [Accepted: 03/25/2021] [Indexed: 01/09/2023]
Abstract
This study aimed to fabricate a multi-layered biphasic calcium phosphate (BCP) platform for programmed bone morphogenetic protein-2 (BMP-2) release, which means to block the initial burst release and promote releasing during the differentiation phase of osteogenic cells. And it is to confirm in vivo whether this platform has osteogenic inductivity even when extremely low doses of BMP-2 are loaded compared to the conventional soaking method. Our strategy consisted of preparing a multilayer coating on BCP to minimize the contact between BMP-2 and BCP and allow the loading of BMP-2. The multilayer, which is surface-modified on BCP, is composed of an organosilicate and a natural polymer-based layer-by-layer (LbL) film. We applied (3-Aminopropyl)triethoxysilane (APTES) as an organosilicate was used for amine-functionalized BCP and (collagen/heparin)5 film was used to delay and sustain BMP-2 release. The coated multilayer not only reduced the initial burst release by more than 50% but also loaded more BMP-2. For in vivo experiment, histomorphometric analysis, it was observed that the BCP platform loaded with extremely low concentration BMP-2 (0.01 mg/ml) induced a significantly larger amount of new bones at 8 weeks compared to the conventional soaking method in the rabbit calvarium onlay graft model.
Collapse
Affiliation(s)
- Seora Han
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Kyeong-Won Paeng
- Department of Periodontology, Research Institute for Periodontal Regeneration, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Sohyeon Park
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Ui-Won Jung
- Department of Periodontology, Research Institute for Periodontal Regeneration, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Jae-Kook Cha
- Department of Periodontology, Research Institute for Periodontal Regeneration, Yonsei University College of Dentistry, Seoul, Republic of Korea.
| | - Jinkee Hong
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
27
|
Wang Y, Sun J, Zhang Y, Liu W, Yang S, Yang J. Stichopus japonicus Polysaccharide Stimulates Osteoblast Differentiation through Activation of the Bone Morphogenetic Protein Pathway in MC3T3-E1 Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:2576-2584. [PMID: 33417444 DOI: 10.1021/acs.jafc.0c06466] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
This study aimed to examine the combined use of bone morphogenetic protein-2 (BMP-2) and polysaccharide isolated from Stichopus japonicus on osteogenic differentiation of MC3T3-E1 cells. Osteogenic differentiation was measured via histochemical staining of alkaline phosphatase (ALP) assay, alizarin red staining of mineralization assay, Western blotting, ELISA, and a qRT-PCR evaluation for the expression of BMP-2, runt-related transcription factor-2 (Runx-2), osteocalcin (OCN), osteopontin (OPN), and collagen type I (Col I) in MC3T3-E1 cells. Immunofluorescence assay was utilized to assess the BMP-2 localized on the cell surface. The results illustrated that SP-2 was able to increase ALP expression and accelerate the mineralization. Osteoblasts cultured on BMP-2/SP-2 substrate increased the expression levels of BMP-2, Runx-2, Col I, OCN, and OPN. SP-2 increased the binding efficiency involving a BMP-2 and its cell surface receptor. The dose of 5 μg/mL SP-2 used showed the best function of inducing osteoblast differentiation. These findings indicated that SP-2 is a more effective enhancer that cooperated with BMP-2 to induce osteoblastic differentiation by utilizing the BMP-2 signaling pathway.
Collapse
Affiliation(s)
- Yanjie Wang
- School of Food Science and Technology, Dalian Polytechnic University, National Engineering Research Center of Seafood, No.1 Qinggongyuan, Dalian 116034, P. R. China
| | - Jinghe Sun
- School of Food Science and Technology, Dalian Polytechnic University, National Engineering Research Center of Seafood, No.1 Qinggongyuan, Dalian 116034, P. R. China
| | - Yanqi Zhang
- Department of Statistics, Iowa State University, 2438 Osborn Drive, Ames, Iowa 50011, United States
| | - Wenzhuan Liu
- School of Food Science and Technology, Dalian Polytechnic University, National Engineering Research Center of Seafood, No.1 Qinggongyuan, Dalian 116034, P. R. China
| | - Sheng Yang
- Roy J. Carver Department of Biochemistry, Biophysics & Molecular Biology, Iowa State University, 3192 Molecular Biology Building, Ames, Iowa 50011, United States
| | - Jingfeng Yang
- School of Food Science and Technology, Dalian Polytechnic University, National Engineering Research Center of Seafood, No.1 Qinggongyuan, Dalian 116034, P. R. China
| |
Collapse
|
28
|
Selective endocytosis of recombinant human BMPs through cell surface heparan sulfate proteoglycans in CHO cells: BMP-2 and BMP-7. Sci Rep 2021; 11:3378. [PMID: 33564092 PMCID: PMC7873082 DOI: 10.1038/s41598-021-82955-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 01/25/2021] [Indexed: 12/15/2022] Open
Abstract
Cell surface heparan sulfate proteoglycan (HSPG)-mediated endocytosis results in poor yields of recombinant human bone morphogenetic proteins (rhBMPs) from CHO cell cultures. Upon incubation of rhBMP-2 and rhBMP-7 with CHO cells at 37 °C, both rhBMP-2 and rhBMP-7 bound to the cell surface HSPGs in CHO cells, but only rhBMP-2 was actively internalized into CHO cells. Cell surface HSPGs were found to serve as the main receptor for rhBMP-2 internalization. It was also found that the cell surface HSPG-mediated endocytosis of rhBMP-2 occurred through both the clathrin- and caveolin-dependent pathways. Blockage of rhBMP-2 internalization by the addition of structural analogs of HSPGs such as dextran sulfate (DS) and heparin dramatically increased rhBMP-2 production in recombinant CHO (rCHO) cell cultures. Compared to the control cultures, addition of DS (1.0 g/L) and heparin (0.2 g/L) resulted in a 22.0- and 19.0-fold increase in the maximum rhBMP-2 concentration, respectively. In contrast, the production of rhBMP-7, which was not internalized into the rCHO cells, did not dramatically increase upon addition of DS and heparin. Taken together, rhBMPs have a different fate in terms of HSPG-mediated internalization in CHO cells. HSPG-mediated endocytosis of each rhBMP should be understood individually to increase the rhBMP yield in rCHO cell cultures.
Collapse
|
29
|
Terauchi M, Tamura A, Arisaka Y, Masuda H, Yoda T, Yui N. Cyclodextrin-Based Supramolecular Complexes of Osteoinductive Agents for Dental Tissue Regeneration. Pharmaceutics 2021; 13:136. [PMID: 33494320 PMCID: PMC7911178 DOI: 10.3390/pharmaceutics13020136] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/13/2021] [Accepted: 01/18/2021] [Indexed: 12/16/2022] Open
Abstract
Oral tissue regeneration has received growing attention for improving the quality of life of patients. Regeneration of oral tissues such as alveolar bone and widely defected bone has been extensively investigated, including regenerative treatment of oral tissues using therapeutic cells and growth factors. Additionally, small-molecule drugs that promote bone formation have been identified and tested as new regenerative treatment. However, treatments need to progress to realize successful regeneration of oral functions. In this review, we describe recent progress in development of regenerative treatment of oral tissues. In particular, we focus on cyclodextrin (CD)-based pharmaceutics and polyelectrolyte complexation of growth factors to enhance their solubility, stability, and bioactivity. CDs can encapsulate hydrophobic small-molecule drugs into their cavities, resulting in inclusion complexes. The inclusion complexation of osteoinductive small-molecule drugs improves solubility of the drugs in aqueous solutions and increases in vitro osteogenic differentiation efficiency. Additionally, various anionic polymers such as heparin and its mimetic polymers have been developed to improve stability and bioactivity of growth factors. These polymers protect growth factors from deactivation and degradation by complex formation through electrostatic interaction, leading to potentiation of bone formation ability. These approaches using an inclusion complex and polyelectrolyte complexes have great potential in the regeneration of oral tissues.
Collapse
Affiliation(s)
- Masahiko Terauchi
- Department of Maxillofacial Surgery, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo, Tokyo 113-8549, Japan; (M.T.); (H.M.); (T.Y.)
| | - Atsushi Tamura
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan; (Y.A.); (N.Y.)
| | - Yoshinori Arisaka
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan; (Y.A.); (N.Y.)
| | - Hiroki Masuda
- Department of Maxillofacial Surgery, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo, Tokyo 113-8549, Japan; (M.T.); (H.M.); (T.Y.)
| | - Tetsuya Yoda
- Department of Maxillofacial Surgery, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo, Tokyo 113-8549, Japan; (M.T.); (H.M.); (T.Y.)
| | - Nobuhiko Yui
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan; (Y.A.); (N.Y.)
| |
Collapse
|
30
|
Deng Z, Ng C, Inoue K, Chen Z, Xia Y, Hu X, Greenblatt M, Pernis A, Zhao B. Def6 regulates endogenous type-I interferon responses in osteoblasts and suppresses osteogenesis. eLife 2020; 9:e59659. [PMID: 33373293 PMCID: PMC7771961 DOI: 10.7554/elife.59659] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 12/15/2020] [Indexed: 12/28/2022] Open
Abstract
Bone remodeling involves a balance between bone resorption and formation. The mechanisms underlying bone remodeling are not well understood. DEF6 is recently identified as a novel loci associated with bone mineral density. However, it is unclear how Def6 impacts bone remodeling. We identify Def6 as a novel osteoblastic regulator that suppresses osteoblastogenesis and bone formation. Def6 deficiency enhances both bone resorption and osteogenesis. The enhanced bone resorption in Def6-/- mice dominates, leading to osteoporosis. Mechanistically, Def6 inhibits the differentiation of both osteoclasts and osteoblasts via a common mechanism through endogenous type-I IFN-mediated feedback inhibition. RNAseq analysis shows expression of a group of IFN stimulated genes (ISGs) during osteoblastogenesis. Furthermore, we found that Def6 is a key upstream regulator of IFNβ and ISG expression in osteoblasts. Collectively, our results identify a novel immunoregulatory function of Def6 in bone remodeling, and shed insights into the interaction between immune system and bone.
Collapse
Affiliation(s)
- Zhonghao Deng
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special SurgeryNew YorkUnited States
| | - Courtney Ng
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special SurgeryNew YorkUnited States
| | - Kazuki Inoue
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special SurgeryNew YorkUnited States
- Department of Medicine, Weill Cornell Medical CollegeNew YorkUnited States
| | - Ziyu Chen
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special SurgeryNew YorkUnited States
| | - Yuhan Xia
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special SurgeryNew YorkUnited States
| | - Xiaoyu Hu
- Institute for Immunology and School of Medicine, Tsinghua UniversityBeijingChina
| | - Matthew Greenblatt
- Pathology and Laboratory Medicine, Weill Cornell Medical CollegeNew YorkUnited States
- Research Division, Hospital for Special SurgeryNew YorkUnited States
| | - Alessandra Pernis
- Autoimmunity and Inflammation Program, Hospital for Special SurgeryNew YorkUnited States
- Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special SurgeryNew YorkUnited States
- Department of Medicine, Weill Cornell Medical CollegeNew YorkUnited States
- Graduate Program in Cell and Development Biology, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| |
Collapse
|
31
|
Xu H, Sun M, Wang C, Xia K, Xiao S, Wang Y, Ying L, Yu C, Yang Q, He Y, Liu A, Chen L. Growth differentiation factor-5-gelatin methacryloyl injectable microspheres laden with adipose-derived stem cells for repair of disc degeneration. Biofabrication 2020; 13:015010. [PMID: 33361566 DOI: 10.1088/1758-5090/abc4d3] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nucleus pulposus (NP) degeneration is the major cause of degenerative disc disease (DDD). This condition cannot be treated or attenuated by traditional open or minimally invasive surgical options. However, a combination of stem cells, growth factors (GFs) and biomaterials present a viable option for regeneration. Injectable biomaterials act as carriers for controlled release of GFs and deliver stem cells to target tissues through a minimally invasive approach. In this study, injectable gelatin methacryloyl microspheres (GMs) with controllable, uniform particle sizes were rapidly biosynthesized through a low-cost electrospraying method. The GMs were used as delivery vehicles for cells and GFs, and they exhibited good mechanical properties and biocompatibility and enhanced the in vitro differentiation of laden cells into NP-like phenotypes. Furthermore, this integrated system attenuated the in vivo degeneration of rat intervertebral discs, maintained NP tissue integrity and accelerated the synthesis of extracellular matrix. Therefore, this novel therapeutic system is a promising option for the treatment of DDD.
Collapse
Affiliation(s)
- Haibin Xu
- Department of Orthopedic Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, Zhejiang, People's Republic of China. Department of Orthopedic Research, Institute of Zhejiang University, Hangzhou 310009, Zhejiang, People's Republic of China. These two authors contributed equally to this work
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Chen CH, Hsu EL, Stupp SI. Supramolecular self-assembling peptides to deliver bone morphogenetic proteins for skeletal regeneration. Bone 2020; 141:115565. [PMID: 32745692 PMCID: PMC7680412 DOI: 10.1016/j.bone.2020.115565] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 07/27/2020] [Accepted: 07/29/2020] [Indexed: 12/15/2022]
Abstract
Recombinant human bone morphogenetic proteins (BMPs) have shown clinical success in promoting bone healing, but they are also associated with unwanted side effects. The development of improved BMP carriers that can retain BMP at the defect site and maximize its efficacy would decrease the therapeutic BMP dose and thus improve its safety profile. In this review, we discuss the advantages of using self-assembling peptides, a class of synthetic supramolecular biomaterials, to deliver recombinant BMPs. Peptide amphiphiles (PAs) are a broad class of self-assembling peptides, and the use of PAs for BMP delivery and bone regeneration has been explored extensively over the past decade. Like many self-assembling peptide systems, PAs can be designed to form nanofibrous supramolecular biomaterials in which molecules are held together by non-covalent bonds. Chemical and biological functionality can be added to PA nanofibers, through conjugation of chemical moieties or biological epitopes to PA molecules. For example, PA nanofibers have been designed to bind heparan sulfate, a natural polysaccharide that is known to bind BMPs and potentiate their signal. Alternatively, PA nanofibers have been designed to synthetically mimic the structure and function of heparan sulfate, or to directly bind BMP specifically. In small animal models, these bio-inspired PA materials have shown the capacity to promote bone regeneration using BMP at doses 10-100 times lower than established therapeutic doses. These promising results have motivated further evaluation of PAs in large animal models, where their safety and efficacy must be established before clinical translation. We conclude with a discussion on the possiblity of combining PAs with other materials used in orthopaedic surgery to maximize their utility for clinical translation.
Collapse
Affiliation(s)
- Charlotte H Chen
- Simpson Querrey Institute, Northwestern University, 303 East Superior Street, Chicago, IL 60611, USA; Department of Materials Science and Engineering, Northwestern University, 2220 Campus Drive, Evanston, IL 60208, USA
| | - Erin L Hsu
- Simpson Querrey Institute, Northwestern University, 303 East Superior Street, Chicago, IL 60611, USA; Department of Orthopaedic Surgery, Northwestern University, 676 North St. Clair Street, Chicago, IL 60611, USA
| | - Samuel I Stupp
- Simpson Querrey Institute, Northwestern University, 303 East Superior Street, Chicago, IL 60611, USA; Department of Materials Science and Engineering, Northwestern University, 2220 Campus Drive, Evanston, IL 60208, USA; Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA; Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA; Department of Medicine, Northwestern University, 676 North St. Clair Street, Chicago, IL 60611, USA.
| |
Collapse
|
33
|
Kim S, Lee M. Rational design of hydrogels to enhance osteogenic potential. CHEMISTRY OF MATERIALS : A PUBLICATION OF THE AMERICAN CHEMICAL SOCIETY 2020; 32:9508-9530. [PMID: 33551566 PMCID: PMC7857485 DOI: 10.1021/acs.chemmater.0c03018] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Bone tissue engineering (BTE) encompasses the field of biomaterials, cells, and bioactive molecules to successfully guide the growth and repair of bone tissue. Current BTE strategies rely on delivering osteogenic molecules or cells via scaffolding materials. However, growth factor- and stem cell-based treatments have several limitations, such as source restriction, low stability, difficulties in predicting long-term efficacy, and high costs, among others. These issues have promoted the development of material-based therapy with properties of accessibility, high stability, tunable efficacy, and low-cost production. Hydrogels are widely used in BTE applications because of their unique hydrophilic nature and tunable physicochemical properties to mimic the native bone environment. However, current hydrogel materials are not ideal candidates due to minimal osteogenic capability on their own. Therefore, recent studies of BTE hydrogels attempt to counterbalance these issues by modifying their biophysical properties. In this article, we review recent progress in the design of hydrogels to instruct osteogenic potential, and present strategies developed to precisely control its bone healing properties.
Collapse
Affiliation(s)
- Soyon Kim
- Division of Advanced Prosthodontics, University of California, Los Angeles, USA
| | - Min Lee
- Division of Advanced Prosthodontics, University of California, Los Angeles, USA
- Department of Bioengineering, University of California, Los Angeles, USA
| |
Collapse
|
34
|
Wigmosta TB, Popat KC, Kipper MJ. Bone morphogenetic protein-2 delivery from polyelectrolyte multilayers enhances osteogenic activity on nanostructured titania. J Biomed Mater Res A 2020; 109:1173-1182. [PMID: 32985077 DOI: 10.1002/jbm.a.37109] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/22/2020] [Accepted: 09/26/2020] [Indexed: 01/09/2023]
Abstract
Incomplete osseointegration is primary cause of failure for orthopedic implants. New biomaterials that present stable signals promoting osteogenesis could reduce failure rates of orthopedic implants. In this study bone morphogenetic protein-2 (BMP-2) was delivered from titania nanotubes (Nt) modified with chitosan/heparin polyelectrolyte multilayers (PEMs). The surfaces were characterized by scanning electron microscopy and X-ray photoelectron spectroscopy. BMP-2 release from the surfaces was measured in vitro for up to 28 days. After an initial burst release of BMP-2 during the first 2 days, most of the BMP-2 remained on the surface. To determine the osteogenic properties of these surfaces, they were seeded with rat bone marrow cells; alkaline phosphatase (ALP) activity, total protein, calcium deposition, and osteocalcin were measured up to 4 weeks in vitro. When compared to Nt surfaces, the surfaces with BMP-2 induce greater osteocalcin and calcium deposition. PEMs provide sustained presentation of BMP-2, from a biomimetic surface. This enhances the osteogenic properties of the surface without requiring supraphysiologic growth factor dose. This growth factor delivery strategy could be used to improve bone healing outcomes and reduce complications for recipients of orthopedic implants.
Collapse
Affiliation(s)
- Tara B Wigmosta
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, USA
| | - Ketul C Popat
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, USA.,School of Advanced Materials Discovery, Colorado State University, Fort Collins, Colorado, USA.,Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado, USA
| | - Matt J Kipper
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, USA.,School of Advanced Materials Discovery, Colorado State University, Fort Collins, Colorado, USA.,Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
35
|
Biniazan F, Manzari-Tavakoli A, Safaeinejad F, Moghimi A, Rajaei F, Niknejad H. The differentiation effect of bone morphogenetic protein (BMP) on human amniotic epithelial stem cells to express ectodermal lineage markers. Cell Tissue Res 2020; 383:751-763. [PMID: 32960356 DOI: 10.1007/s00441-020-03280-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 08/12/2020] [Indexed: 11/28/2022]
Abstract
Stem cells are a promising tool for treatment of a variety of degenerative diseases. Human amniotic epithelial stem cells (hAECs) have desirable and unique characteristics that make them a proper candidate for cell therapy. In this study, we have investigated the effects of BMP-4 (bone morphogenetic protein-4) and its inhibition on differentiation of AECs into ectodermal lineages. Analysis of AEC-derived ectodermal lineages (neurons and keratinocytes) was performed by using flow cytometry technique for Map2 and β-tubulin (as neuron markers), Olig2 and MBP (as oligodendrocyte markers), and K14 and K10 (as keratinocyte markers). The results of this study illustrated that noggin (as BMP antagonist), BMP4, and both BMP4 and heparin (together or separately) increased neural and keratinocyte marker expression, respectively. The expression of markers MAP2, olig2, and K14 in hAECs has been significantly decreased 21 days after exposure to differentiation medium (without growth factors) compared with isolation day, which supports the hypothesis that AECs can be dedifferentiated into pluripotent cells. Moreover, activation and inhibition of BMP signaling have no effects on viability of hAECs. The results of this study showed that BMP signaling and its inhibition are the key factors for ectodermal lineage differentiation of amnion-derived stem cells.
Collapse
Affiliation(s)
- Felor Biniazan
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Research Center, Qazvin University of Medical Science, Qazvin, Iran
| | - Asma Manzari-Tavakoli
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Fahimeh Safaeinejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Moghimi
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Farzad Rajaei
- Cellular and Molecular Research Center, Qazvin University of Medical Science, Qazvin, Iran.
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
36
|
Halloran D, Durbano HW, Nohe A. Bone Morphogenetic Protein-2 in Development and Bone Homeostasis. J Dev Biol 2020; 8:E19. [PMID: 32933207 PMCID: PMC7557435 DOI: 10.3390/jdb8030019] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/01/2020] [Accepted: 09/11/2020] [Indexed: 12/11/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are multi-functional growth factors belonging to the Transforming Growth Factor-Beta (TGF-β) superfamily. These proteins are essential to many developmental processes, including cardiogenesis, neurogenesis, and osteogenesis. Specifically, within the BMP family, Bone Morphogenetic Protein-2 (BMP-2) was the first BMP to be characterized and has been well-studied. BMP-2 has important roles during embryonic development, as well as bone remodeling and homeostasis in adulthood. Some of its specific functions include digit formation and activating osteogenic genes, such as Runt-Related Transcription Factor 2 (RUNX2). Because of its diverse functions and osteogenic potential, the Food and Drug Administration (FDA) approved usage of recombinant human BMP-2 (rhBMP-2) during spinal fusion surgery, tibial shaft repair, and maxillary sinus reconstructive surgery. However, shortly after initial injections of rhBMP-2, several adverse complications were reported, and alternative therapeutics have been developed to limit these side-effects. As the clinical application of BMP-2 is largely implicated in bone, we focus primarily on its role in bone. However, we also describe briefly the role of BMP-2 in development. We then focus on the structure of BMP-2, its activation and regulation signaling pathways, BMP-2 clinical applications, and limitations of using BMP-2 as a therapeutic. Further, this review explores other potential treatments that may be useful in treating bone disorders.
Collapse
Affiliation(s)
| | | | - Anja Nohe
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA; (D.H.); (H.W.D.)
| |
Collapse
|
37
|
Hurley-Novatny A, Arumugasaamy N, Kimicata M, Baker H, Mikos AG, Fisher JP. Concurrent multi-lineage differentiation of mesenchymal stem cells through spatial presentation of growth factors. Biomed Mater 2020; 15:055035. [PMID: 32526725 PMCID: PMC7648258 DOI: 10.1088/1748-605x/ab9bb0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Severe tendon and ligament injuries are estimated to affect between 300 000 and 400 000 people annually. Surgical repairs of these injuries often have poor long-term clinical outcomes because of resection of the interfacial tissue-the enthesis-and subsequent stress concentration at the attachment site. A healthy enthesis consists of distinct regions of bone, fibrocartilage, and tendon, each with distinct cell types, extracellular matrix components, and architecture, which are important for tissue function. Tissue engineering, which has been proposed as a potential strategy for replacing this tissue, is currently limited by its inability to differentiate multiple lineages of cells from a single stem cell population within a single engineered construct. In this study, we develop a multi-phasic gelatin methacrylate hydrogel construct system for spatial presentation of proteins, which is then validated for multi-lineage differentiation towards the cell types of the bone-tendon enthesis. This study determines growth factor concentrations for differentiation of mesenchymal stem cells towards osteoblasts, chondrocytes/fibrochondrocytes, and tenocytes, which maintain similar differentiation profiles in 3D hydrogel culture as assessed by qPCR and immunofluorescence staining. Finally, it is shown that this method is able to guide heterogeneous and spatially confined changes in mesenchymal stem cell genes and protein expressions with the tendency to result in osteoblast-, fibrochondrocyte-, and tenocyte-like expression profiles. Overall, we demonstrate the utility of the culture technique for engineering other musculoskeletal tissue interfaces and provide a biochemical approach for recapitulating the bone-tendon enthesis in vitro.
Collapse
Affiliation(s)
- Amelia Hurley-Novatny
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, United States of America. Center for Engineering Complex Tissues, University of Maryland and Rice University, College Park, MD 20742, United States of America
| | | | | | | | | | | |
Collapse
|
38
|
Ao Q, Wang S, He Q, Ten H, Oyama K, Ito A, He J, Javed R, Wang A, Matsuno A. Fibrin Glue/Fibronectin/Heparin-Based Delivery System of BMP2 Induces Osteogenesis in MC3T3-E1 Cells and Bone Formation in Rat Calvarial Critical-Sized Defects. ACS APPLIED MATERIALS & INTERFACES 2020; 12:13400-13410. [PMID: 32091872 DOI: 10.1021/acsami.0c01371] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Bone morphogenetic proteins (BMPs) have been used to promote bone formation in many clinical scenarios. However, the BMPs are inherently unstable in vivo and therefore need to be combined with carriers for controlled delivery. In this study, an innovative and efficient fibrin glue/fibronectin/heparin (FG/Fn/Hep)-based delivery system was developed for controlled release of BMP2. The incorporation of heparin can significantly slow the release of BMP2 without substantially affecting the structure and stiffness of the FG/Fn. The BMP2 release from the FG/Fn/Hep-BMP2 hydrogel is largely dominated by hydrogel degradation rather than simple diffusion. In vitro release experiments and MC3T3-E1 cell induction experiments showed that BMP2 can be released steadily and can induce MC3T3-E1 cells to differentiate into osteoblasts efficiently. This process is characterized by the significantly increased expression of calcium deposits, alkaline phosphatase, runt-related transcription factor-2, osteopontin, osteocalcin, and collagen I in comparison with the negative control. In vivo assessments revealed that the FG/Fn/Hep-BMP2 hydrogel significantly promotes bone regeneration in a rat calvarial critical-sized defect model. Our investigation indicates that FG/Fn/Hep-BMP2 hydrogel holds promise to be used as an alternative biomaterial for the repair of bone defects.
Collapse
Affiliation(s)
- Qiang Ao
- Department of Tissue Engineering, China Medical University, Shenyang 110122, China
- Institute of Regulatory Science for Medical Device, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Shilin Wang
- Department of Tissue Engineering, China Medical University, Shenyang 110122, China
| | - Qing He
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Hirotomo Ten
- Department of Neurosurgery, Teikyo University School of Medicine, Tokyo 163-8001, Japan
| | - Kenichi Oyama
- Department of Neurosurgery, Teikyo University School of Medicine, Tokyo 163-8001, Japan
| | - Akihiro Ito
- Department of Neurosurgery, Teikyo University School of Medicine, Tokyo 163-8001, Japan
| | - Jing He
- Department of Tissue Engineering, China Medical University, Shenyang 110122, China
| | - Rabia Javed
- Department of Tissue Engineering, China Medical University, Shenyang 110122, China
| | - Aijun Wang
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California, Davis, Sacramento, California 95817, United States
| | - Akira Matsuno
- Department of Neurosurgery, Teikyo University School of Medicine, Tokyo 163-8001, Japan
| |
Collapse
|
39
|
Kim S, Fan J, Lee CS, Chen C, Bubukina K, Lee M. Heparinized chitosan stabilizes the bioactivity of BMP-2 and potentiates the osteogenic efficacy of demineralized bone matrix. J Biol Eng 2020; 14:6. [PMID: 32165922 PMCID: PMC7059291 DOI: 10.1186/s13036-020-0231-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/25/2020] [Indexed: 12/23/2022] Open
Abstract
Background Demineralized bone matrix (DBM), an allograft bone processed to better expose osteoinductive factors such as bone morphogenetic proteins (BMPs), is increasingly used for clinical bone repair. However, more extensive use of DBM is limited by its unpredictable osteoinductivity and low bone formation capacity. Commercial DBM products often employ polymeric carriers to enhance handling properties but such carriers generally do not possess bioactive functions. Heparin is a highly sulfated polysaccharide and is shown to form a stable complex with growth factors to enhance their bioactivities. In this study, a new heparinized synthetic carrier for DBM is developed based on photocrosslinking of methacrylated glycol chitosan and heparin conjugation. Results Heparinized chitosan exerts protective effects on BMP bioactivity against physiological stressors related to bone fracture healing. It also enhances the potency of BMPs by inhibiting the activity of BMP antagonist, noggin. Moreover, heparinized chitosan is effective to deliver bone marrow stromal cells and DBM for enhanced osteogenesis by sequestering and localizing the cell-produced or DBM-released BMPs. Conclusions This research suggests an essential approach of developing a new hydrogel carrier to stabilize the bioactivity of BMPs and improve the clinical efficacy of current bone graft therapeutics for accelerated bone repair.
Collapse
Affiliation(s)
- Soyon Kim
- 1Division of Advanced Prosthodontics, University of California, Los Angeles, USA
| | - Jiabing Fan
- 1Division of Advanced Prosthodontics, University of California, Los Angeles, USA
| | - Chung-Sung Lee
- 1Division of Advanced Prosthodontics, University of California, Los Angeles, USA
| | - Chen Chen
- 1Division of Advanced Prosthodontics, University of California, Los Angeles, USA
| | - Ksenia Bubukina
- 1Division of Advanced Prosthodontics, University of California, Los Angeles, USA
| | - Min Lee
- 1Division of Advanced Prosthodontics, University of California, Los Angeles, USA.,2Department of Bioengineering, University of California, Los Angeles, USA
| |
Collapse
|
40
|
Gong Y, Zhang Y, Cao Z, Ye F, Lin Z, Li Y. Development of CaCO 3 microsphere-based composite hydrogel for dual delivery of growth factor and Ca to enhance bone regeneration. Biomater Sci 2020; 7:3614-3626. [PMID: 31210206 DOI: 10.1039/c9bm00463g] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Injectable scaffolds have attracted much attention because of their minimum surgical invasiveness. However, limited osteogenic induction property and low mechanical properties hampered their application in bone tissue engineering. CaCO3 microspheres, which possess osteoinductivity, rough surfaces and specific binding sites for BMP-2, were first fabricated; after BMP-2 uploading, microspheres were further entrapped in fibrin-glue hydrogel. CaCO3 microspheres were co-functionalized with casein and heparin. To obtain a high encapsulation of heparin and thus BMP-2 uploading, along with controlled release and simultaneous maintenance of the presence of vaterite which had osteogenic induction property, fabrication parameters were optimized and microspheres were characterized using XRD, FITR and SEM. The formed CaCO3 had a microsphere morphology of ∼1 μm. Both vaterite and calcite phases were present and the relative amount of calcite phase increased with the amount of heparin. Sample 25 mM_4-1Hep with the highest loading amount of heparin was selected as carrier for BMP-2 and BMP-2 loaded CaCO3 microspheres were further entrapped in fibrin-glue hydrogel (FC-B). For the as-prepared composite hydrogel, mechanical properties were characterized and the presence of CaCO3 significantly elevated the tensile strength; controlled release of BMP-2 was sustained until day 21. Based on ALP activity, alizarin red staining and RT-PCR, in vitro osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) was found to be significantly enhanced under induction of FC-B. Rabbit tibia bone defect model was applied to evaluate its in vivo performance. After implantation for 4 weeks, presence of composite hydrogel was observed in defects. After 8 weeks, bone defects of FC-B group were nearly completely healed. Using the fact that autologous scaffolds can be derived based on fibrin-glue hydrogel, the well-designed BMP-2 loaded fibrin-glue composite hydrogel demonstrated good potential in bone tissue engineering.
Collapse
Affiliation(s)
- Yihong Gong
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.
| | | | | | | | | | | |
Collapse
|
41
|
Liu L, Lam WMR, Yang Z, Wang M, Ren X, Hu T, Li J, Goh JCH, Wong HK. Improving the handling properties and long-term stability of polyelectrolyte complex by freeze-drying technique for low-dose bone morphogenetic protein 2 delivery. J Biomed Mater Res B Appl Biomater 2020; 108:2450-2460. [PMID: 32017424 DOI: 10.1002/jbm.b.34577] [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: 08/26/2019] [Revised: 01/06/2020] [Accepted: 01/20/2020] [Indexed: 11/09/2022]
Abstract
A variety of controlled release carriers for bone morphogenetic protein 2 (BMP-2) delivery have been developed and tested in animal models. An alginate-based polyelectrolyte complex (PEC) for controlled release of low-dose BMP-2 has shown promising results in preclinical research. However, the poor handling properties and long-term stability of PEC need to be improved for translational applications. This study aimed to address these limitations of alginate-based PEC by employing a freeze-drying technique. The size and structure of freeze-dried PEC (FD-PEC) were maintained with the addition of a cryoprotectant, trehalose. The release profile of BMP-2 from FD-PEC was similar to that of freshly prepared PEC. In vitro bioactivity analysis of the released BMP-2 showed that the carrier performance of PEC was not compromised by freeze-drying up to three-month storage at room temperature. BMP-2-bound FD-PEC induced comparable bone formation to that using freshly prepared regular PEC in a rat posterolateral spinal fusion model. These results suggest that FD-PEC is capable of delivering low-dose BMP-2 and could be developed as an off-the-shelf product for translational applications. The simplicity of this preservation method provides promise for the translational application of PEC.
Collapse
Affiliation(s)
- Ling Liu
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Wing M R Lam
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Zheng Yang
- NUS Tissue Engineering Program (NUSTEP), Life Sciences Institute, National University of Singapore, Singapore
| | - Ming Wang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Xiafei Ren
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Tao Hu
- Department of Spine Surgery, Shanghai East Hospital, Shanghai, China
| | - Jun Li
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore
| | - James Cho-Hong Goh
- NUS Tissue Engineering Program (NUSTEP), Life Sciences Institute, National University of Singapore, Singapore.,Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore
| | - Hee-Kit Wong
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,NUS Tissue Engineering Program (NUSTEP), Life Sciences Institute, National University of Singapore, Singapore
| |
Collapse
|
42
|
Hettiaratchi MH, Krishnan L, Rouse T, Chou C, McDevitt TC, Guldberg RE. Heparin-mediated delivery of bone morphogenetic protein-2 improves spatial localization of bone regeneration. SCIENCE ADVANCES 2020; 6:eaay1240. [PMID: 31922007 PMCID: PMC6941907 DOI: 10.1126/sciadv.aay1240] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 11/07/2019] [Indexed: 05/25/2023]
Abstract
Supraphysiologic doses of bone morphogenetic protein-2 (BMP-2) are used clinically to promote bone formation in fracture nonunions, large bone defects, and spinal fusion. However, abnormal bone formation (i.e., heterotopic ossification) caused by rapid BMP-2 release from conventional collagen sponge scaffolds is a serious complication. We leveraged the strong affinity interactions between heparin microparticles (HMPs) and BMP-2 to improve protein delivery to bone defects. We first developed a computational model to investigate BMP-2-HMP interactions and demonstrated improved in vivo BMP-2 retention using HMPs. We then evaluated BMP-2-loaded HMPs as a treatment strategy for healing critically sized femoral defects in a rat model that displays heterotopic ossification with clinical BMP-2 doses (0.12 mg/kg body weight). HMPs increased BMP-2 retention in vivo, improving spatial localization of bone formation in large bone defects and reducing heterotopic ossification. Thus, HMPs provide a promising opportunity to improve the safety profile of scaffold-based BMP-2 delivery.
Collapse
Affiliation(s)
- Marian H. Hettiaratchi
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30322, USA
- The Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR 97403, USA
| | - Laxminarayanan Krishnan
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Tel Rouse
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Catherine Chou
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Todd C. McDevitt
- The Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158, USA
| | - Robert E. Guldberg
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30322, USA
- The Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR 97403, USA
| |
Collapse
|
43
|
Denton NF, Eghleilib M, Al-Sharifi S, Todorčević M, Neville MJ, Loh N, Drakesmith A, Karpe F, Pinnick KE. Bone morphogenetic protein 2 is a depot-specific regulator of human adipogenesis. Int J Obes (Lond) 2019; 43:2458-2468. [PMID: 31324879 PMCID: PMC6892741 DOI: 10.1038/s41366-019-0421-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 05/13/2019] [Accepted: 05/17/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Bone morphogenetic proteins (BMPs) regulate adipogenesis but it is not clear whether they influence regional adipose tissue (AT) development in humans. OBJECTIVE To characterise BMP2 expression, BMP2-SMAD1/5/8 signalling, and BMP2's potential effect on proliferation and adipogenesis in human subcutaneous abdominal and gluteal AT and its constituent preadipocytes. METHODS BMP2 expression was measured in whole AT and immortalised preadipocytes via qPCR and Western blot; secreted/circulating BMP2 was measured by ELISA. The effect of BMP2 on preadipocyte proliferation was evaluated using a fluorescent assay. BMP2's effect on adipogenesis in immortalised preadipocytes was determined via qPCR of adipogenic markers and cellular triacylglycerol (TAG) accumulation. BMP2-SMAD1/5/8 signalling was assessed in immortalised preadipocytes via Western blot and qPCR of ID1 expression. RESULTS BMP2 was expressed and released by abdominal and gluteal AT and preadipocytes. Exogenous BMP2 dose dependently promoted adipogenesis in abdominal preadipocytes only; 50 ng/ml BMP2 increased PPARG2 expression (10-fold compared to vehicle, p < 0.001) and TAG accumulation (3-fold compared to vehicle; p < 0.001). BMP2 stimulated SMAD1/5/8 phosphorylation and ID1 expression in abdominal and gluteal preadipocytes but this was blocked by 500 nM K02288, a type 1 BMP receptor inhibitor (p < 0.001). Co-administration of 500 nM K02288 also inhibited the pro-adipogenic effect of 50 ng/ml BMP2 in abdominal cells; >90% inhibition of TAG accumulation (p < 0.001) and ~50% inhibition of PPARG2 expression (p < 0.001). The endogenous iron regulator erythroferrone reduced BMP2-SMAD1/5/8 signalling by ~30% specifically in subcutaneous abdominal preadipocytes (p < 0.01), suggesting it plays a role in restricting the expansion of the body's largest AT depot during energy deficiency. Additionally, a waist-hip ratio-increasing common polymorphism near BMP2 is an eQTL associated with ~15% lower BMP2 expression in abdominal and gluteal AT (p < 0.05) as well as altered adipocyte size in male abdominal AT (p < 0.05). CONCLUSIONS These data implicate BMP2-SMAD1/5/8 signalling in depot-specific preadipocyte development and abdominal AT expansion in humans.
Collapse
Affiliation(s)
- Nathan F Denton
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Mohamed Eghleilib
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Sama Al-Sharifi
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Marijana Todorčević
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Matt J Neville
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospital NHS Trust, Oxford, UK
| | - Nellie Loh
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Alexander Drakesmith
- The MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
- NIHR Oxford Biomedical Research Centre, Oxford University Hospital NHS Trust, Oxford, UK.
| | - Katherine E Pinnick
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
44
|
Terauchi M, Tamura A, Tonegawa A, Yamaguchi S, Yoda T, Yui N. Polyelectrolyte Complexes between Polycarboxylates and BMP-2 for Enhancing Osteogenic Differentiation: Effect of Chemical Structure of Polycarboxylates. Polymers (Basel) 2019; 11:polym11081327. [PMID: 31405005 PMCID: PMC6723113 DOI: 10.3390/polym11081327] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/07/2019] [Accepted: 08/07/2019] [Indexed: 01/15/2023] Open
Abstract
Bone morphogenetic protein 2 (BMP-2) has received considerable attention because of its osteoinductivity, but its use is limited owing to its instability and adverse effects. To reduce the dose of BMP-2, complexation with heparin is a promising approach, because heparin enhances the osteoinductivity of BMP-2. However, the clinical use of heparin is restricted because of its anticoagulant activity. Herein, to explore alternative polymers that show heparin-like activity, four polycarboxylates, poly(acrylic acid) (PAA), poly(methacrylic acid) (PMAA), poly(aspartic acid) (PAsp), and poly(glutamic acid) (PGlu), were selected and their capability to modulate the osteoinductivity of BMP-2 was evaluated. Dynamic light scattering indicated that these polycarboxylates formed polyelectrolyte complexes with BMP-2. The osteogenic differentiation efficiency of MC3T3-E1 cells treated with the polycarboxylate/BMP-2 complexes was investigated in comparison to that of the heparin/BMP-2 complex. As a result, PGlu/BMP-2 complex showed the highest activity of alkaline phosphatase, which is an early-stage marker of osteogenic differentiation, and rapid mineralization. Based on these observations, PGlu could serve as an alternative to heparin in the regenerative therapy of bone using BMP-2.
Collapse
Affiliation(s)
- Masahiko Terauchi
- Department of Maxillofacial Surgery, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo, Tokyo 113-8549, Japan
| | - Atsushi Tamura
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan.
| | - Asato Tonegawa
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan
| | - Satoshi Yamaguchi
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan
| | - Tetsuya Yoda
- Department of Maxillofacial Surgery, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo, Tokyo 113-8549, Japan
| | - Nobuhiko Yui
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan
| |
Collapse
|
45
|
Whitely M, Rodriguez-Rivera G, Waldron C, Mohiuddin S, Cereceres S, Sears N, Ray N, Cosgriff-Hernandez E. Porous PolyHIPE microspheres for protein delivery from an injectable bone graft. Acta Biomater 2019; 93:169-179. [PMID: 30685476 PMCID: PMC6615946 DOI: 10.1016/j.actbio.2019.01.044] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 01/03/2019] [Accepted: 01/23/2019] [Indexed: 12/30/2022]
Abstract
Delivery of osteoinductive factors such as bone morphogenetic protein 2 (BMP-2) has emerged as a prominent strategy to improve regeneration in bone grafting procedures. However, it remains challenging to identify a carrier that provides the requisite loading efficiency and release kinetics without compromising the mechanical properties of the bone graft. Previously, we reported on porous, polymerized high internal phase emulsion (polyHIPE) microspheres fabricated using controlled fluidics. Uniquely, this solvent-free method provides advantages over current microsphere fabrication strategies including in-line loading of growth factors to improve loading efficiency. In the current study, we utilized this platform to fabricate protein-loaded microspheres and investigated the effect of particle size (∼400 vs ∼800 μm) and pore size (∼15 vs 30 μm) on release profiles. Although there was no significant effect of these variables on the substantial burst release profile of the microspheres, the incorporation of the protein-loaded microspheres within the injectable polyHIPE resulted in a sustained release of protein from the bulk scaffold over a two-week period with minimal burst release. Bioactivity retention of encapsulated BMP-2 was confirmed first using a genetically-modified osteoblast reporter cell line. A functional assay with human mesenchymal stem cells established that the BMP-2 release from microspheres induced osteogenic differentiation. Finally, microsphere incorporation had minimal effect on the cure and compressive properties of an injectable polyHIPE bone graft. Overall, this work demonstrates that these microsphere-polyHIPE composites have strong potential to enhance bone regeneration through controlled release of BMP-2 and other growth factors. STATEMENT OF SIGNIFICANCE: This manuscript describes a method for solvent-free fabrication of porous microspheres from high internal phase emulsions using a controlled fluids setup. The principles of emulsion templating and fluid dynamics provide exceptional control of particle size and pore architecture. In addition to the advantage of solvent-free fabrication, this method provides in-line loading of protein directly into the pores of the microspheres with high loading efficiencies. The incorporation of the protein-loaded microspheres within an injectable polyHIPE scaffold resulted in a sustained release of protein over a two-week period with minimal burst release. Retention of BMP-2 bioactivity and incorporation of microspheres with minimal effect on scaffold compressive properties highlights the potential of these new bone grafts.
Collapse
Affiliation(s)
- Michael Whitely
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, 77843-3120, U.S.A
| | - Gabriel Rodriguez-Rivera
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, 78712, U.S.A
| | - Christina Waldron
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, 78712, U.S.A
| | - Sahar Mohiuddin
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, 77843-3120, U.S.A
| | - Stacy Cereceres
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, 77843-3120, U.S.A
| | - Nicholas Sears
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, 77843-3120, U.S.A
| | - Nicholas Ray
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, 78712, U.S.A
| | | |
Collapse
|
46
|
Fahimipour F, Dashtimoghadam E, Mahdi Hasani-Sadrabadi M, Vargas J, Vashaee D, Lobner DC, Jafarzadeh Kashi TS, Ghasemzadeh B, Tayebi L. Enhancing cell seeding and osteogenesis of MSCs on 3D printed scaffolds through injectable BMP2 immobilized ECM-Mimetic gel. Dent Mater 2019; 35:990-1006. [PMID: 31027908 PMCID: PMC7193177 DOI: 10.1016/j.dental.2019.04.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 04/01/2019] [Accepted: 04/09/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Design of bioactive scaffolds with osteogenic capacity is a central challenge in cell-based patient-specific bone tissue engineering. Efficient and spatially uniform seeding of (stem) cells onto such constructs is vital to attain functional tissues. Herein we developed heparin functionalized collagen gels supported by 3D printed bioceramic scaffolds, as bone extracellular matrix (ECM)-mimetic matrices. These matrices were designed to enhance cell seeding efficiency of mesenchymal stem cells (MSCs) as well as improve their osteogenic differentiation through immobilized bone morphogenic protein 2 (BMP2) to be used for personalized bone regeneration. METHODS A 3D gel based on heparin-conjugated collagen matrix capable of immobilizing recombinant human bone morphogenic protein 2 (BMP2) was synthesized. Isolated dental pulp Mesenchymal stem cells (MSCs) were then encapsulated into the bone ECM microenvironment to efficiently and uniformly seed a bioactive ceramic-based scaffold fabricated using additive manufacturing technique. The designed 3D cell-laden constructs were comprehensively investigated trough in vitro assays and in vivo study. RESULTS In-depth rheological characterizations of heparin-conjugated collagen gel revealed that elasticity of the matrix is significantly improved compared with freely incorporated heparin. Investigation of the MSCs laden collagen-heparin hydrogels revealed their capability to provide spatiotemporal bioavailability of BMP2 while suppressing the matrix contraction over time. The in vivo histology and real-time polymerase chain reaction (qPCR) analysis showed that the designed construct supported the osteogenic differentiation of MSCs and induced the ectopic bone formation in rat model. SIGNIFICANCE The presented hybrid constructs combine bone ECM chemical cues with mechanical function providing an ideal 3D microenvironment for patient-specific bone tissue engineering and cell therapy applications. The implemented methodology in design of ECM-mimetic 3D matrix capable of immobilizing BMP2 to improve seeding efficiency of customized scaffolds can be exploited for other bioactive molecules.
Collapse
Affiliation(s)
- Farahnaz Fahimipour
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA; Dental Biomaterials Department, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Mahdi Hasani-Sadrabadi
- Parker H. Petit Institute for Bioengineering and Bioscience, G.W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prothodontics, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Jessica Vargas
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| | - Daryoosh Vashaee
- Electrical and Computer Engineering Department, North Carolina State University, Raleigh, NC 27606, USA
| | - Douglas C Lobner
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI 53233, USA
| | - Tahereh S Jafarzadeh Kashi
- Dental Biomaterials Department, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Behnam Ghasemzadeh
- Department of Biomedical Sciences, Integrative Neuroscience Research Center, Marquette University, Milwaukee, WI 53201, USA
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA.
| |
Collapse
|
47
|
Dinoro J, Maher M, Talebian S, Jafarkhani M, Mehrali M, Orive G, Foroughi J, Lord MS, Dolatshahi-Pirouz A. Sulfated polysaccharide-based scaffolds for orthopaedic tissue engineering. Biomaterials 2019; 214:119214. [PMID: 31163358 DOI: 10.1016/j.biomaterials.2019.05.025] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 12/11/2022]
Abstract
Given their native-like biological properties, high growth factor retention capacity and porous nature, sulfated-polysaccharide-based scaffolds hold great promise for a number of tissue engineering applications. Specifically, as they mimic important properties of tissues such as bone and cartilage they are ideal for orthopaedic tissue engineering. Their biomimicry properties encompass important cell-binding motifs, native-like mechanical properties, designated sites for bone mineralisation and strong growth factor binding and signaling capacity. Even so, scientists in the field have just recently begun to utilise them as building blocks for tissue engineering scaffolds. Most of these efforts have so far been directed towards in vitro studies, and for these reasons the clinical gap is still substantial. With this review paper, we have tried to highlight some of the important chemical, physical and biological features of sulfated-polysaccharides in relation to their chondrogenic and osteogenic inducing capacity. Additionally, their usage in various in vivo model systems is discussed. The clinical studies reviewed herein paint a promising picture heralding a brave new world for orthopaedic tissue engineering.
Collapse
Affiliation(s)
- Jeremy Dinoro
- Intelligent Polymer Research Institute ARC Centre of Excellence for Electromaterials Science AIIM Facility University of Wollongong, Australia
| | - Malachy Maher
- Intelligent Polymer Research Institute ARC Centre of Excellence for Electromaterials Science AIIM Facility University of Wollongong, Australia
| | - Sepehr Talebian
- Intelligent Polymer Research Institute ARC Centre of Excellence for Electromaterials Science AIIM Facility University of Wollongong, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Mahboubeh Jafarkhani
- Technical University of Denmark, DTU Nanotech, Center for Intestinal Absorption and Transport of Biopharmaceuticals, 2800 Kgs, Denmark
| | - Mehdi Mehrali
- Technical University of Denmark, DTU Nanotech, Center for Intestinal Absorption and Transport of Biopharmaceuticals, 2800 Kgs, Denmark
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, Vitoria-Gasteiz, 01006, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain; Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore
| | - Javad Foroughi
- Intelligent Polymer Research Institute ARC Centre of Excellence for Electromaterials Science AIIM Facility University of Wollongong, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Megan S Lord
- Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Alireza Dolatshahi-Pirouz
- Technical University of Denmark, DTU Nanotech, Center for Intestinal Absorption and Transport of Biopharmaceuticals, 2800 Kgs, Denmark; Department of Regenerative Biomaterials, Radboud University Medical Center, Philips van Leydenlaan 25, Nijmegen, 6525 EX, the Netherlands.
| |
Collapse
|
48
|
Oki Y, Kirita K, Ohta S, Ohba S, Horiguchi I, Sakai Y, Ito T. Switching of Cell Proliferation/Differentiation in Thiol–Maleimide Clickable Microcapsules Triggered by in Situ Conjugation of Biomimetic Peptides. Biomacromolecules 2019; 20:2350-2359. [DOI: 10.1021/acs.biomac.9b00333] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Yuichiro Oki
- Department of Chemical System Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Katsuhisa Kirita
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Seiichi Ohta
- Center for
Disease
Biology and Integrative Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Shinsuke Ohba
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Center for
Disease
Biology and Integrative Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Ikki Horiguchi
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Taichi Ito
- Department of Chemical System Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Center for
Disease
Biology and Integrative Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
49
|
Li C, Ouyang L, Pence IJ, Moore AC, Lin Y, Winter CW, Armstrong JPK, Stevens MM. Buoyancy-Driven Gradients for Biomaterial Fabrication and Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1900291. [PMID: 30844123 PMCID: PMC6606439 DOI: 10.1002/adma.201900291] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/18/2019] [Indexed: 05/25/2023]
Abstract
The controlled fabrication of gradient materials is becoming increasingly important as the next generation of tissue engineering seeks to produce inhomogeneous constructs with physiological complexity. Current strategies for fabricating gradient materials can require highly specialized materials or equipment and cannot be generally applied to the wide range of systems used for tissue engineering. Here, the fundamental physical principle of buoyancy is exploited as a generalized approach for generating materials bearing well-defined compositional, mechanical, or biochemical gradients. Gradient formation is demonstrated across a range of different materials (e.g., polymers and hydrogels) and cargos (e.g., liposomes, nanoparticles, extracellular vesicles, macromolecules, and small molecules). As well as providing versatility, this buoyancy-driven gradient approach also offers speed (<1 min) and simplicity (a single injection) using standard laboratory apparatus. Moreover, this technique is readily applied to a major target in complex tissue engineering: the osteochondral interface. A bone morphogenetic protein 2 gradient, presented across a gelatin methacryloyl hydrogel laden with human mesenchymal stem cells, is used to locally stimulate osteogenesis and mineralization in order to produce integrated osteochondral tissue constructs. The versatility and accessibility of this fabrication platform should ensure widespread applicability and provide opportunities to generate other gradient materials or interfacial tissues.
Collapse
Affiliation(s)
- Chunching Li
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, Prince Consort Road, London, SW7 2AZ, United Kingdom
| | - Liliang Ouyang
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, Prince Consort Road, London, SW7 2AZ, United Kingdom
| | - Isaac J. Pence
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, Prince Consort Road, London, SW7 2AZ, United Kingdom
| | - Axel C. Moore
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, Prince Consort Road, London, SW7 2AZ, United Kingdom
| | - Yiyang Lin
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, Prince Consort Road, London, SW7 2AZ, United Kingdom
| | - Charles W. Winter
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, Prince Consort Road, London, SW7 2AZ, United Kingdom
| | - James P. K. Armstrong
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, Prince Consort Road, London, SW7 2AZ, United Kingdom
| | - Molly M. Stevens
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, Prince Consort Road, London, SW7 2AZ, United Kingdom
| |
Collapse
|
50
|
Zhu J, Xia K, Yu W, Wang Y, Hua J, Liu B, Gong Z, Wang J, Xu A, You Z, Chen Q, Li F, Tao H, Liang C. Sustained release of GDF5 from a designed coacervate attenuates disc degeneration in a rat model. Acta Biomater 2019; 86:300-311. [PMID: 30660009 DOI: 10.1016/j.actbio.2019.01.028] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 12/23/2018] [Accepted: 01/14/2019] [Indexed: 12/17/2022]
Abstract
Low back pain is often caused by intervertebral disc degeneration, which is characterized by nucleus pulposus (NP) and extracellular matrix (ECM) degeneration. Human adipose-derived stem cells (hADSCs) induced by growth and differentiation factor-5 (GDF5) can differentiate into an NP-like phenotype. Although stem cell-based therapy with prolonged exposure to growth factors is regarded as a promising treatment, the efficacy of this approach in attenuating the disc degeneration process is limited by the short lifespan of growth factors. In our study, a unique growth factor delivery vehicle composed of heparin and the synthetic polycation poly(ethylene argininylaspartate diglyceride) (PEAD) was used to sustain GDF5 release. The results showed that sustained release of GDF5 by the PEAD:heparin delivery system promoted hADSC differentiation to an NP-like phenotype in vitro. After injection of the PEAD:heparin:GDF5 delivery platform and hADSCs into intervertebral spaces of coccygeal (Co) vertebrae Co7/Co8 and Co8/Co9 of the rat, the disc height, water content, and structure of the NPs decreased more slowly than other treatment groups. This new strategy may be used as an alternative treatment for attenuating intervertebral disc degeneration with hADSCs without the need for gene therapy. STATEMENT OF SIGNIFICANCE: Low back pain is often caused by intervertebral disc degeneration, which is characterized by nucleus pulposus (NP) and extracellular matrix (ECM) degeneration. Human adipose-derived stem cells (hADSCs) induced by growth and differentiation factor-5 (GDF-5) can differentiate into an NP-like phenotype. Although stem cell-based therapy with prolonged exposure to growth factor is regarded as a promising treatment, the efficacy of this approach in the disc regeneration process is limited by the short life of growth factors. In our study, a unique growth factor delivery vehicle comprised of heparin and the synthetic polycation poly(ethylene argininylaspartate diglyceride) (PEAD) was used to sustain the release of GDF-5. Numerous groups have explored IDD regeneration methods in vitro and in vivo. Our study differs in that GDF5 was incorporated into a vehicle through charge attraction and exhibited a sustained release profile. Moreover, GDF-5 seeded coacervate combined with hADSC injection could be a minimally invasive approach for tissue engineering that is suitable for clinical application. We investigated the stimulatory effects of our GDF-5 seeded coacervate on the differentiation of ADSCs in vitro and the reparative effect of the delivery system on degenerated NP in vivo.
Collapse
|