1
|
Khandan-Nasab N, Torkamanzadeh B, Abbasi B, Mohajeri T, Oskuee RK, Sahebkar A. Application of Platelet-Rich Plasma-Based Scaffolds in Soft and Hard Tissue Regeneration. TISSUE ENGINEERING. PART B, REVIEWS 2025. [PMID: 40296834 DOI: 10.1089/ten.teb.2024.0285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Platelet-rich plasma (PRP) is a blood product with higher platelet concentrations than whole blood, offering controlled delivery of growth factors (GFs) for regenerative medicine. PRP plays pivotal roles in tissue restoration mechanisms, including angiogenesis, fibroblast proliferation, and extracellular matrix development, making it applicable across various regenerative medicine treatments. Despite promising results in different tissue injuries, challenges such as short half-life and rapid deactivation by proteases persist. To address these challenges, biomaterial-based delivery scaffolds, such as sponges or hydrogels, have been investigated. Current studies exhibit that PRP-loaded scaffolds fix these issues due to the sustained release of GFs. In this regard, given the widespread application of PRP in clinical studies, the use of PRP-loaded scaffolds has drawn significant consideration in tissue engineering (TE). Therefore, this review briefly introduces PRP as a rich origin of GFs, its classification, and preparation methods and discusses PRP applications in regenerative medicine. This study also emphasizes and reviews the latest research on the using scaffolds for PRP delivery in diverse fields of TE, including skin, bone, and cartilage repair.
Collapse
Affiliation(s)
- Niloofar Khandan-Nasab
- Targeted Drug Delivery Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Behdad Torkamanzadeh
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Behnam Abbasi
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Taraneh Mohajeri
- Department of Obstetrics & Gynecology, Mashhad Medical Sciences Branch, Islamic Azad University, Mashhad, Iran
| | - Reza Kazemi Oskuee
- Targeted Drug Delivery Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Centre for Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
2
|
Rahman MH, Mondal MIH. Stability, challenges, and prospects of chitosan for the delivery of anticancer drugs and tissue regenerative growth factors. Heliyon 2024; 10:e39879. [PMID: 39583848 PMCID: PMC11582409 DOI: 10.1016/j.heliyon.2024.e39879] [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: 04/02/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/26/2024] Open
Abstract
Chitosan, a biopolymer derived from chitin, offers significant potential for regulated anticancer drug administration and tissue regeneration growth factors, owing to its biocompatibility, low toxicity, biodegradability, and little immunogenicity. Moreover, its structure can be extensively modified, for example, to create scaffolds, hydrogels, nanoparticles, and membranes, allowing it to be engineered precisely to achieve specific outcomes However, the therapeutic utilisation of chitosan is impeded by significant challenges, such as its inadequate hemocompatibility, durability, and uniformity in commercial manufacturing. Additionally, there is insufficient research offering a thorough examination of the capabilities, limitations, and challenges related to chitosan as carriers for anticancer drugs and growth factors. This article examines the stability, challenges, and advanced application of chitosan as a drug carrier in anti-cancer therapy and growth factor delivery. The problems of unregulated chitosan degradation arising from unsuitable storage conditions are considered and potential solutions, and areas for future research, are proposed to deal with such problems. Consequently, this review is expected to be highly valuable for aspiring scientists studying chitosan-related systems for delivery of anti-cancer drugs and growth factors.
Collapse
Affiliation(s)
- Md Hasinur Rahman
- Polymer and Textile Research Lab, Department of Applied Chemistry and Chemical Engineering, Rajshahi University, Rajshahi, 6205, Bangladesh
| | - Md Ibrahim H. Mondal
- Polymer and Textile Research Lab, Department of Applied Chemistry and Chemical Engineering, Rajshahi University, Rajshahi, 6205, Bangladesh
| |
Collapse
|
3
|
Gaihre B, Camilleri E, Tilton M, Astudillo Potes MD, Liu X, Lucien F, Lu L. LAPONITE® nano-silicates potentiate the angiogenic effects of FG-4592 and osteogenic effects of BMP-2. Biomater Sci 2024; 12:5610-5619. [PMID: 39359127 PMCID: PMC11822916 DOI: 10.1039/d4bm00636d] [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] [Indexed: 10/04/2024]
Abstract
LAPONITE®-based drug delivery systems offer many advantages due to the unique ionic and physical properties of LAPONITE®. The high ionicity and large surface area of LAPONITE® nanoparticles enable the intercalation and dissolution of biomolecules. In this study, we explored the potential of LAPONITE® as a carrier for FG-4592 to support angiogenesis and as a carrier for bone morphogenic protein-2 (BMP-2) to support osteogenesis. Interestingly, we found that LAPONITE® promoted the FG-4592 induced upregulation of vascular endothelial growth factor (VEGF) gene expression of human umbilical cord endothelial cells (HUVECs). Additionally, we observed that LAPONITE® could provide a sustained release of BMP-2 and significantly potentiate the osteogenic effects of BMP-2 on adipose derived mesenchymal stem cells (AMSCs). Overall, current findings on the LAPONITE®-drug/protein model system provide a unique way to potentiate the angiogenic activities of FG-4592 on HUVECs and osteogenic effects of BMP-2 on AMSCs for tissue engineering application. Future studies will be directed towards gaining a deeper understanding of these effects on a co-culture system of HUVECs and AMSCs.
Collapse
Affiliation(s)
- Bipin Gaihre
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Emily Camilleri
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Maryam Tilton
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Maria D Astudillo Potes
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Xifeng Liu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Fabrice Lucien
- Department of Urology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Lichun Lu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| |
Collapse
|
4
|
Krutko M, Poling HM, Bryan AE, Sharma M, Singh A, Reza HA, Wikenheiser-Brokamp KA, Takebe T, Helmrath MA, Harris GM, Esfandiari L. Enhanced Piezoelectric Performance of PVDF-TrFE Nanofibers through Annealing for Tissue Engineering Applications. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.16.608345. [PMID: 39229142 PMCID: PMC11370437 DOI: 10.1101/2024.08.16.608345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
This study investigates bioelectric stimulation's role in tissue regeneration by enhancing the piezoelectric properties of tissue-engineered grafts using annealed poly(vinylidene fluoride-trifluoroethylene) (PVDF-TrFE) scaffolds. Annealing at temperatures of 80°C, 100°C, 120°C, and 140°C was assessed for its impact on material properties and physiological utility. Analytical techniques such as Differential Scanning Calorimetry (DSC), Fourier-Transform Infrared Spectroscopy (FTIR), and X-ray Diffraction (XRD) revealed increased crystallinity with higher annealing temperatures, peaking in β-phase content and crystallinity at 140°C. Scanning Electron Microscopy (SEM) showed that 140°C annealed scaffolds had enhanced lamellar structures, increased porosity, and maximum piezoelectric response. Mechanical tests indicated that 140°C annealing improved elastic modulus, tensile strength, and substrate stiffness, aligning these properties with physiological soft tissues. In vitro assessments in Schwann cells demonstrated favorable responses, with increased cell proliferation, contraction, and extracellular matrix attachment. Additionally, genes linked to extracellular matrix production, vascularization, and calcium signaling were upregulated. The foreign body response in C57BL/6 mice, evaluated through Hematoxylin and Eosin (H&E) and Picrosirius Red staining, showed no differences between scaffold groups, supporting the potential for future functional evaluation of the annealed group in tissue repair.
Collapse
|
5
|
Lee S, Lee SM, Lee SH, Choi WK, Park SJ, Kim DY, Oh SW, Oh J, Cho JY, Lee J, Chien PN, Nam SY, Heo CY, Lee YS, Kwak EA, Chung WJ. In situ photo-crosslinkable hyaluronic acid-based hydrogel embedded with GHK peptide nanofibers for bioactive wound healing. Acta Biomater 2023; 172:159-174. [PMID: 37832839 DOI: 10.1016/j.actbio.2023.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/18/2023] [Accepted: 10/06/2023] [Indexed: 10/15/2023]
Abstract
A versatile hydrogel was developed for enhancing bioactive wound healing by introducing the amphiphilic GHK peptide (GHK-C16) into a photo-crosslinkable tyramine-modified hyaluronic acid (HA-Ty). GHK-C16 self-assembled into GHK nanofibers (GHK NF) in HA-Ty solution, which underwent in situ gelation after the wound area was filled with precursor solution. Blue light irradiation (460-490 nm), with riboflavin phosphate as a photoinitiator, was used to trigger crosslinking, which enhanced the stability of the highly degradable hyaluronic acid and enabled sustained release of the nanostructured GHK derivatives. The hydrogels provided a microenvironment that promoted the proliferation of dermal fibroblasts and the activation of cytokines, leading to reduced inflammation and increased collagen expression during wound healing. The complexation of Cu2+ into GHK nanofibers resulted in superior wound healing capabilities compared with non-lipidated GHK peptide with a comparable level of growth factor (EGF). Additionally, nanostructured Cu-GHK improved angiogenesis through vascular endothelial growth factor (VEGF) activation, which exerted a synergistic therapeutic effect. Furthermore, in vivo wound healing experiments revealed that the Cu-GHK NF/HA-Ty hydrogel accelerated wound healing through densely packed remodeled collagen in the dermis and promoting the growth of denser fibroblasts. HA-Ty hydrogels incorporating GHK NF also possessed improved mechanical properties and a faster wound healing rate, making them suitable for advanced bioactive wound healing applications. STATEMENT OF SIGNIFICANCE: By combining photo-crosslinkable tyramine-modified hyaluronic acid with self-assembled Cu-GHK-C16 peptide nanofibers (Cu-GHK NF), the Cu-GHK NF/HA-Ty hydrogel offers remarkable advantages over conventional non-structured Cu-GHK for wound healing. It enhances cell proliferation, migration, and collagen remodeling-critical factors in tissue regeneration. The incorporation of GHK nanofibers complexed with copper ions imparts potent anti-inflammatory effects, promoting cytokine activation and angiogenesis during wound healing. The Cu-GHK NF/hydrogel's unique properties, including in situ photo-crosslinking, ensure high customization and potency in tissue regeneration, providing a cost-effective alternative to growth factors. In vivo experiments further validate its efficacy, demonstrating significant wound closure, collagen remodeling, and increased fibroblast density. Overall, the Cu-GHK NF/HA-Ty hydrogel represents an advanced therapeutic option for wound healing applications.
Collapse
Affiliation(s)
- Seohui Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Sang Min Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Sang Hyun Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Woong-Ku Choi
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Sung-Jun Park
- School of Chemical and Biological Engineering, Seoul National University, 151-744, Seoul, Republic of Korea
| | - Do Yeon Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Sae Woong Oh
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Jieun Oh
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Jongsung Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Pham Ngoc Chien
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Sun Young Nam
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Chan Yeong Heo
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Department of Medical Device Development, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Yoon-Sik Lee
- School of Chemical and Biological Engineering, Seoul National University, 151-744, Seoul, Republic of Korea
| | - Eun-A Kwak
- Research Institute of Biomolecule Control, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea.
| | - Woo-Jae Chung
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea; Research Institute of Biomolecule Control, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea; Center for Biologics, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
6
|
Chen Y, Xiang Y, Zhang H, Zhu T, Chen S, Li J, Du J, Yan X. A multifunctional chitosan composite aerogel based on high density amidation for chronic wound healing. Carbohydr Polym 2023; 321:121248. [PMID: 37739489 DOI: 10.1016/j.carbpol.2023.121248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/29/2023] [Accepted: 07/31/2023] [Indexed: 09/24/2023]
Abstract
The management of chronic wounds remains a challenging clinical problem worldwide, mainly because of secondary infections, excessive oxidative stress, and blocked angiogenesis. Aerogel is a novel material with high porosity and specific surface area that allows gas exchange and rapid absorption of a large amount of exudate as well as loading bioactive molecules. Therefore, functional aerogel can be an ideal material for chronic wound treatment. The multifunctional aerogel (CG-DA-VEGF) was prepared by a simple and eco-friendly freeze-drying process combined with harmless EDC/NHS as crosslinking agents using chitosan and dopamine-grafted gelatin as raw materials. The physicochemical characterization revealed that the CG-DA-VEGF aerogel had excellent water absorption, water retention, and mechanical properties, and could release VEGF continuously and stably. In vitro experiments demonstrated that the CG-DA-VEGF aerogel exhibited effective antioxidant and antibacterial properties, as well as superb cytocompatibility. In vivo experiments further confirmed that the CG-DA-VEGF aerogel could significantly improve angiogenesis and re-epithelialization, and promote collagen deposition, thus accelerating wound healing with excellent biosafety. These results suggest that the as-prepared CG-DA-VEGF aerogel may be adopted as a promising multifunctional graft for the treatment of chronic wounds.
Collapse
Affiliation(s)
- Yao Chen
- School of Chemistry and Chemical Engineering, Shanghai Engineering Research Center of Pharmaceutical Intelligent Equipment, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai 201620, PR China
| | - Yu Xiang
- Department of Sports Medicine, Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Rd., Shanghai 200233, PR China
| | - Hongmei Zhang
- School of Chemistry and Chemical Engineering, Shanghai Engineering Research Center of Pharmaceutical Intelligent Equipment, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai 201620, PR China.
| | - Tonghe Zhu
- School of Chemistry and Chemical Engineering, Shanghai Engineering Research Center of Pharmaceutical Intelligent Equipment, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai 201620, PR China; Department of Sports Medicine, Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Rd., Shanghai 200233, PR China
| | - Sihao Chen
- School of Chemistry and Chemical Engineering, Shanghai Engineering Research Center of Pharmaceutical Intelligent Equipment, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai 201620, PR China
| | - Jun Li
- Department of Orthopedics, Tongji Hospital, School of medicine, Tongji University, 389 Xincun Rd., Shanghai 200065, PR China
| | - Juan Du
- School of Chemistry and Chemical Engineering, Shanghai Engineering Research Center of Pharmaceutical Intelligent Equipment, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai 201620, PR China.
| | - Xiaoyu Yan
- Department of Sports Medicine, Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Rd., Shanghai 200233, PR China.
| |
Collapse
|
7
|
Beheshtizadeh N, Gharibshahian M, Bayati M, Maleki R, Strachan H, Doughty S, Tayebi L. Vascular endothelial growth factor (VEGF) delivery approaches in regenerative medicine. Biomed Pharmacother 2023; 166:115301. [PMID: 37562236 DOI: 10.1016/j.biopha.2023.115301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/28/2023] [Accepted: 08/05/2023] [Indexed: 08/12/2023] Open
Abstract
The utilization of growth factors in the process of tissue regeneration has garnered significant interest and has been the subject of extensive research. However, despite the fervent efforts invested in recent clinical trials, a considerable number of these studies have produced outcomes that are deemed unsatisfactory. It is noteworthy that the trials that have yielded the most satisfactory outcomes have exhibited a shared characteristic, namely, the existence of a mechanism for the regulated administration of growth factors. Despite the extensive exploration of drug delivery vehicles and their efficacy in delivering certain growth factors, the development of a reliable predictive approach for the delivery of delicate growth factors like Vascular Endothelial Growth Factor (VEGF) remains elusive. VEGF plays a crucial role in promoting angiogenesis; however, the administration of VEGF demands a meticulous approach as it necessitates precise localization and transportation to a specific target tissue. This process requires prolonged and sustained exposure to a low concentration of VEGF. Inaccurate administration of drugs, either through off-target effects or inadequate delivery, may heighten the risk of adverse reactions and potentially result in tumorigenesis. At present, there is a scarcity of technologies available for the accurate encapsulation of VEGF and its subsequent sustained and controlled release. The objective of this review is to present and assess diverse categories of VEGF administration mechanisms. This paper examines various systems, including polymeric, liposomal, hydrogel, inorganic, polyplexes, and microfluidic, and evaluates the appropriate dosage of VEGF for multiple applications.
Collapse
Affiliation(s)
- Nima Beheshtizadeh
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran; Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Maliheh Gharibshahian
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran; Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammad Bayati
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran
| | - Reza Maleki
- Department of Chemical Technologies, Iranian Research Organization for Science and Technology (IROST), P.O. Box 33535111, Tehran, Iran.
| | - Hannah Strachan
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| | - Sarah Doughty
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| |
Collapse
|
8
|
Chen QQ, Liu QY, Wang P, Qian TM, Wang XH, Yi S, Li SY. Potential application of let-7a antagomir in injured peripheral nerve regeneration. Neural Regen Res 2023; 18:1584-1590. [PMID: 36571366 PMCID: PMC10075095 DOI: 10.4103/1673-5374.357914] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Neurotrophic factors, particularly nerve growth factor, enhance neuronal regeneration. However, the in vivo applications of nerve growth factor are largely limited by its intrinsic disadvantages, such as its short biological half-life, its contribution to pain response, and its inability to cross the blood-brain barrier. Considering that let-7 (human miRNA) targets and regulates nerve growth factor, and that let-7 is a core regulator in peripheral nerve regeneration, we evaluated the possibilities of let-7 application in nerve repair. In this study, anti-let-7a was identified as the most suitable let-7 family molecule by analyses of endogenous expression and regulatory relationship, and functional screening. Let-7a antagomir demonstrated biosafety based on the results of in vivo safety assessments and it entered into the main cell types of the sciatic nerve, including Schwann cells, fibroblasts and macrophages. Use of hydrogel effectively achieved controlled, localized, and sustained delivery of let-7a antagomir. Finally, let-7a antagomir was integrated into chitosan conduit to construct a chitosan-hydrogel scaffold tissue-engineered nerve graft, which promoted nerve regeneration and functional recovery in a rat model of sciatic nerve transection. Our study provides an experimental basis for potential in vivo application of let-7a.
Collapse
Affiliation(s)
- Qian-Qian Chen
- State Key Laboratory of Pharmaceutical Biotechnology and Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School, Nanjing University, Nanjing; NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Qian-Yan Liu
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Pan Wang
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Tian-Mei Qian
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Xing-Hui Wang
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Sheng Yi
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Shi-Ying Li
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
9
|
Chiu A, Sharma D, Zhao F. Tissue Engineering-Based Strategies for Diabetic Foot Ulcer Management. Adv Wound Care (New Rochelle) 2023; 12:145-167. [PMID: 34939837 PMCID: PMC9810358 DOI: 10.1089/wound.2021.0081] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 10/26/2021] [Indexed: 01/13/2023] Open
Abstract
Significance: Diabetic foot ulcers (DFU) are a mounting problem with the increasingly frail population. Injuries that would otherwise heal are kept open by risk factors such as diabetes, obesity, and age-related conditions, which interferes with the natural wound healing processes. Recent Advances: This review summarizes recent advancements in the field of tissue engineering for the treatment of DFUs. FDA-approved approaches, including signaling-based therapies, stem cell therapies, and skin substitutes are summarized and cutting-edge experimental technologies that have the potential to manage chronic wounds, such as skin printing, skin organogenesis, skin self-assembly, and prevascularization, are discussed. Critical Issues: The standard of care for chronic wounds involves wound debridement, wound dressings, and resolving the underlying cause such as lowering the glycemic index and reducing wound pressure. Current DFU treatments are limited by low wound closure rates and poor regrown skin quality. New adjuvant therapies that facilitate wound closure in place of or in conjunction with standard care are critically needed. Future Directions: Tissue engineering strategies are limited by the plasticity of adult human cells. In addition to traditional techniques, genetic modification, although currently an emerging technology, has the potential to unlock human regeneration and can be incorporated in future therapeutics.
Collapse
Affiliation(s)
- Alvis Chiu
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Dhavan Sharma
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Feng Zhao
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
10
|
Tawade P, Tondapurkar N, Jangale A. Biodegradable and biocompatible synthetic polymers for applications in bone and muscle tissue engineering. JOURNAL OF MEDICAL SCIENCE 2022. [DOI: 10.20883/medical.e712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In medicine, tissue engineering has made significant advances. Using tissue engineering techniques, transplant treatments result in less donor site morbidity and need fewer surgeries overall. It is now possible to create cell-supporting scaffolds that degrade as new tissue grows on them, replacing them until complete body function is restored. Synthetic polymers have been a significant area of study for biodegradable scaffolds due to their ability to provide customizable biodegradable and mechanical features as well as a low immunogenic effect due to biocompatibility. The food and drug administration has given the biodegradable polymers widespread approval after they showed their reliability. In the context of tissue engineering, this paper aims to deliver an overview of the area of biodegradable and biocompatible synthetic polymers. Frequently used synthetic biodegradable polymers utilized in tissue scaffolding, scaffold specifications, polymer synthesis, degradation factors, as well as fabrication methods are discussed. In order to emphasize the many desired properties and corresponding needs for skeletal muscle and bone, particular examples of synthetic polymer scaffolds are investigated. Increased biocompatibility, functionality and clinical applications will be made possible by further studies into novel polymer and scaffold fabrication approaches.
Collapse
|
11
|
V. K. AD, Ray S, Arora U, Mitra S, Sionkowska A, Jaiswal AK. Dual drug delivery platforms for bone tissue engineering. Front Bioeng Biotechnol 2022; 10:969843. [PMID: 36172012 PMCID: PMC9511792 DOI: 10.3389/fbioe.2022.969843] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/23/2022] [Indexed: 11/22/2022] Open
Abstract
The dual delivery platforms used in bone tissue engineering provide supplementary bioactive compounds that include distinct medicines and growth factors thereby aiding enhanced bone regeneration. The delivery of these compounds can be adjusted for a short or prolonged time based on the requirement by altering various parameters of the carrier platform. The platforms thus used are fabricated to mimic the niche of the bone microenvironment, either in the form of porous 3D structures, microspheres, or films. Thus, this review article focuses on the concept of dual drug delivery platform and its importance, classification of various platforms for dual drug delivery specific to bone tissue engineering, and finally highlights the foresight into the future direction of these techniques for better clinical applications.
Collapse
Affiliation(s)
- Anupama Devi V. K.
- Tissue Engineering Group, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore, India
- School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, India
| | - Sarbajit Ray
- School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, India
| | - Udita Arora
- School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, India
| | - Sunrito Mitra
- School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, India
| | | | - Amit Kumar Jaiswal
- Tissue Engineering Group, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore, India
- *Correspondence: Amit Kumar Jaiswal,
| |
Collapse
|
12
|
Paiva JCC, Oliveira L, Vaz MF, Costa-de-Oliveira S. Biodegradable Bone Implants as a New Hope to Reduce Device-Associated Infections-A Systematic Review. Bioengineering (Basel) 2022; 9:409. [PMID: 36004934 PMCID: PMC9405200 DOI: 10.3390/bioengineering9080409] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/28/2022] [Accepted: 08/10/2022] [Indexed: 11/24/2022] Open
Abstract
Bone fractures often require fixation devices that frequently need to be surgically removed. These temporary implants and procedures leave the patient more prone to developing medical device-associated infections, and osteomyelitis associated with trauma is a challenging complication for orthopedists. In recent years, biodegradable materials have gained great importance as temporary medical implant devices, avoiding removal surgery. The purpose of this systematic review was to revise the literature regarding the use of biodegradable bone implants in fracture healing and its impact on the reduction of implant-associated infections. The systematic review followed the PRISMA guidelines and was conducted by searching published studies regarding the in vivo use of biodegradable bone fixation implants and its antibacterial activity. From a total of 667 references, 23 studies were included based on inclusion and exclusion criteria. Biodegradable orthopedic implants of Mg-Cu, Mg-Zn, and Zn-Ag have shown antibacterial activity, especially in reducing infection burden by MRSA strains in vivo osteomyelitis models. Their ability to prevent and tackle implant-associated infections and to gradually degrade inside the body reduces the need for a second surgery for implant removal, with expectable gains regarding patients' comfort. Further in vivo studies are mandatory to evaluate the efficiency of these antibacterial biodegradable materials.
Collapse
Affiliation(s)
- José C. C. Paiva
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Luís Oliveira
- DPS—Product Systems Development, INEGI—Institute of Science and Innovation in Mechanical and Industrial Engineering, 4200-465 Porto, Portugal
| | - Maria Fátima Vaz
- IDMEC—Instituto Superior Técnico, Universidade de Lisboa, 1499-002 Lisboa, Portugal
- Departamento de Engenharia Mecânica, Instituto Superior Técnico, Universidade de Lisboa, 1499-002 Lisboa, Portugal
| | - Sofia Costa-de-Oliveira
- Division of Microbiology, Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Center for Health Technology and Services Research—CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|
13
|
Cao Y, Shi X, Zhao X, Chen B, Li X, Li Y, Chen Y, Chen C, Lu H, Liu J. Acellular dermal matrix decorated with collagen-affinity peptide accelerate diabetic wound healing through sustained releasing Histatin-1 mediated promotion of angiogenesis. Int J Pharm 2022; 624:122017. [PMID: 35839983 DOI: 10.1016/j.ijpharm.2022.122017] [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: 01/17/2022] [Revised: 06/19/2022] [Accepted: 07/09/2022] [Indexed: 11/26/2022]
Abstract
Treating diabetic ulcers is a major challenge in clinical practice, persecuting millions of patients with diabetes and increasing the medical burden. Recombinant growth factor application can accelerate diabetic wound healing via angiogenesis. The local administration of recombinant growth factors has no robust clinical efficiency because of the degradation of append short duration of the molecules in the hostile inflammatoryenvironment.The present study focused on the pathophysiology of impaired neovascularization and growth factor short duration in the diabetic wound. We prepared a collagen-binding domain (CBD)-fused recombinant peptide (C-Histatin-1) that had both pro-angiogenesis capacity and collagen-affinity properties. Next, we created a biocompatible acellular dermal matrix (ADM) as a drug delivery carrier that featured collagen-richness, high porosity, and non-cytotoxicity. C-Histatin-1 was then tethered on ADM to obtain a sustained-release effect. Finally, a functional scaffold (C-Hst1/ADM) was developed. C-Hst1/ADM can sustain-release Histatin-1 to promote the adhesion, migration, and angiogenesisof vascular endothelial cells in vitro. Using a diabetic wound model, we showed that C-Hst1/ADM could significantly promote angiogenesis, reduce scar widths, and improve extracellular collagen accumulation. Therefore, the results of this study provide a foundation for the clinical application of C-Hst1/ADM covering scaffold in the treatment of diabetic wounds.
Collapse
Affiliation(s)
- Yanpeng Cao
- Department of Limbs (Foot and Hand) Microsurgery, Chenzhou No.1 people's hospital, Chenzhou, China
| | - Xin Shi
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China; Hunan Engineering Research Center of Sports and Health, Changsha, China
| | - Xin Zhao
- Department of Limbs (Foot and Hand) Microsurgery, Chenzhou No.1 people's hospital, Chenzhou, China
| | - Bei Chen
- Department of Limbs (Foot and Hand) Microsurgery, Chenzhou No.1 people's hospital, Chenzhou, China
| | - Xiying Li
- Department of Limbs (Foot and Hand) Microsurgery, Chenzhou No.1 people's hospital, Chenzhou, China
| | - Yabei Li
- Department of Limbs (Foot and Hand) Microsurgery, Chenzhou No.1 people's hospital, Chenzhou, China
| | - Yaowu Chen
- Department of Limbs (Foot and Hand) Microsurgery, Chenzhou No.1 people's hospital, Chenzhou, China
| | - Can Chen
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China; Hunan Engineering Research Center of Sports and Health, Changsha, China; Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Hongbin Lu
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China; Hunan Engineering Research Center of Sports and Health, Changsha, China; Mobile Health Ministry of Education - China Mobile Joint Laboratory, Changsha, China; Xiangya Hospital-International Chinese Musculeskeletal Research Society Sports Medicine Research Centre, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Jun Liu
- Department of Limbs (Foot and Hand) Microsurgery, Chenzhou No.1 people's hospital, Chenzhou, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; The First School of Clinical Medicine, Xiangnan University, Chenzhou, China.
| |
Collapse
|
14
|
Cherng JH, Lin CAJ, Liu CC, Yeh JZ, Fan GY, Tsai HD, Chung CF, Hsu SD. Hemostasis and Anti-Inflammatory Abilities of AuNPs-Coated Chitosan Dressing for Burn Wounds. J Pers Med 2022; 12:jpm12071089. [PMID: 35887586 PMCID: PMC9321560 DOI: 10.3390/jpm12071089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/25/2022] [Accepted: 06/30/2022] [Indexed: 01/03/2023] Open
Abstract
Burn injuries are a common hazard in the military, as fire is likely to be weaponized. Thus, it is important to find an effective substance to accelerate burn wound healing. This study used chitosan and gold nanoparticles (AuNPs) as wound dressings and investigated their effectiveness in femoral artery hemorrhage swine and rat burn models. Chitosan dressing has significant hemostatic properties compared with gauze. Histological results showed that burn wounds treated with chitosan or AuNP-coated chitosan dressings exhibited more cells and a continuous structure of the epidermis and dermis than those of the control and untreated lesion groups. Furthermore, both chitosan dressings have been shown to positively regulate the expression of genes- and cytokines/chemokines-related to the wound healing process; AuNP-coated chitosan significantly lessened severe sepsis and inflammation, balanced the activities of pro-fibrotic and anti-fibrotic ligands for tissue homeostasis, regulated angiogenesis, and inhibited apoptosis activity, thereby being beneficial for the burn microenvironment. Hence, chitosan alone or in combination with AuNPs represents a prospective therapeutic substance as a burn dressing which might be helpful for burn wound care. This study provides a novel hemostasis dressing for modern warfare that is simple to use by most medical and paramedical personnel handling for burn treatment.
Collapse
Affiliation(s)
- Juin-Hong Cherng
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan;
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei 114, Taiwan;
| | - Cheng-An J. Lin
- Department of Biomedical Engineering and Center for Biomedical Engineering in Cancer, Chung Yuan Christian University, Taoyuan 320, Taiwan;
| | - Cheng-Che Liu
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei 114, Taiwan;
| | - Jue-Zong Yeh
- Department of Pharmacy, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan;
| | - Gang-Yi Fan
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei 114, Taiwan;
- Laboratory of Adult Stem Cell and Tissue Regeneration, National Defense Medical Center, Taipei 114, Taiwan; (H.-D.T.); (C.-F.C.)
| | - Hsin-Da Tsai
- Laboratory of Adult Stem Cell and Tissue Regeneration, National Defense Medical Center, Taipei 114, Taiwan; (H.-D.T.); (C.-F.C.)
| | - Chun-Fang Chung
- Laboratory of Adult Stem Cell and Tissue Regeneration, National Defense Medical Center, Taipei 114, Taiwan; (H.-D.T.); (C.-F.C.)
| | - Sheng-Der Hsu
- Division of Traumatology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
- Correspondence:
| |
Collapse
|
15
|
Ivanova TA, Golubeva EN. Aliphatic Polyesters for Biomedical Purposes: Design and Kinetic Regularities of Degradation in vitro. RUSSIAN JOURNAL OF PHYSICAL CHEMISTRY B 2022. [DOI: 10.1134/s1990793122030162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
16
|
Kim K, Siddiqui Z, Acevedo-Jake AM, Roy A, Choudhury M, Grasman J, Kumar V. Angiogenic Hydrogels to Accelerate Early Wound Healing. Macromol Biosci 2022; 22:e2200067. [PMID: 35579914 DOI: 10.1002/mabi.202200067] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/19/2022] [Indexed: 11/09/2022]
Abstract
The metabolic disorder diabetes mellitus affects an increasing proportion of the population, a number projected to double by 2060. Non-life-threatening comorbidities contribute to an interrupted healing process which is first delayed, then prolonged, and associated with increased susceptibility to infection and sustained and unresolved inflammation. This leads to chronic non-healing wounds and eventually potential amputation of extremities. Here we examine the use of a bioactive angiogenic peptide-based hydrogel, SLan, to improve early wound healing in diabetic rats, and compare its performance to clinically utilized biosynthetic peptide-based materials such as Puramatrix. Streptozotocin-treated diabetic rats underwent 8 mm biopsy wounding in their dorsum to remove the epithelium, adipose tissues and muscle layer of the skin, and served as a model for diabetic wound healing. Wounds were treated with either Low (1w%) SLan, High (4w%) SLan, PBS, Puramatrix or K2 (an unfunctionalized non-bioactive control sequentially similar to SLan), covered with Tegaderm and monitored on days 0, 3, 7, 10, 14, 17, 21, 28; animals were sacrificed for histomorphic analyses and immunostaining. An LC/MS method developed to detect SLan in plasma allows pharmacokinetic analysis showing no trafficking of peptides from the wound site into the circulation. Low and High SLan groups show similar final outcomes of wound contraction as control groups (Puramatrix, PBS and K2). SLan-treated rats, however, show marked improvement in healing in earlier time points, including increased deposition of new mature blood vessels. Additionally, rats in the Low SLan treatment groups showed significantly improved wound contraction over other groups and significantly improved healing in early time points. Altogether our results suggest this material can be used to "jumpstart" the diabetic wound healing process. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- KaKyung Kim
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Zain Siddiqui
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Amanda M Acevedo-Jake
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Abhishek Roy
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Marwa Choudhury
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Jonathan Grasman
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Vivek Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA.,Department of Biology, New Jersey Institute of Technology, Newark, NJ, 07102, USA.,Department of Chemical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA.,Department of Restorative Dentistry, Rutgers School of Dental Medicine, Newark, NJ, 07102, USA
| |
Collapse
|
17
|
Kaur G, Narayanan G, Garg D, Sachdev A, Matai I. Biomaterials-Based Regenerative Strategies for Skin Tissue Wound Healing. ACS APPLIED BIO MATERIALS 2022; 5:2069-2106. [PMID: 35451829 DOI: 10.1021/acsabm.2c00035] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Skin tissue wound healing proceeds through four major stages, including hematoma formation, inflammation, and neo-tissue formation, and culminates with tissue remodeling. These four steps significantly overlap with each other and are aided by various factors such as cells, cytokines (both anti- and pro-inflammatory), and growth factors that aid in the neo-tissue formation. In all these stages, advanced biomaterials provide several functional advantages, such as removing wound exudates, providing cover, transporting oxygen to the wound site, and preventing infection from microbes. In addition, advanced biomaterials serve as vehicles to carry proteins/drug molecules/growth factors and/or antimicrobial agents to the target wound site. In this review, we report recent advancements in biomaterials-based regenerative strategies that augment the skin tissue wound healing process. In conjunction with other medical sciences, designing nanoengineered biomaterials is gaining significant attention for providing numerous functionalities to trigger wound repair. In this regard, we highlight the advent of nanomaterial-based constructs for wound healing, especially those that are being evaluated in clinical settings. Herein, we also emphasize the competence and versatility of the three-dimensional (3D) bioprinting technique for advanced wound management. Finally, we discuss the challenges and clinical perspective of various biomaterial-based wound dressings, along with prospective future directions. With regenerative strategies that utilize a cocktail of cell sources, antimicrobial agents, drugs, and/or growth factors, it is expected that significant patient-specific strategies will be developed in the near future, resulting in complete wound healing with no scar tissue formation.
Collapse
Affiliation(s)
- Gurvinder Kaur
- Materials Science and Sensor Applications, Central Scientific Instruments Organization, Chandigarh 160030, India
| | - Ganesh Narayanan
- Fiber and Polymer Science Program, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Deepa Garg
- Materials Science and Sensor Applications, Central Scientific Instruments Organization, Chandigarh 160030, India
| | - Abhay Sachdev
- Materials Science and Sensor Applications, Central Scientific Instruments Organization, Chandigarh 160030, India
| | - Ishita Matai
- Department of Biotechnology, School of Biological Sciences, Amity University Punjab, Mohali 140306, India
| |
Collapse
|
18
|
Characterization of a Human Platelet Lysate-Loaded Keratin Hydrogel for Wound Healing Applications In Vitro. Int J Mol Sci 2022; 23:ijms23084100. [PMID: 35456921 PMCID: PMC9031577 DOI: 10.3390/ijms23084100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 12/22/2022] Open
Abstract
One of the promising approaches to facilitate healing and regenerative capacity includes the application of growth-factor-loaded biomaterials. Human platelet lysate (hPL) derived from platelet-rich plasma through a freeze-thaw process has been used as a growth factor rich therapeutic in many regenerative applications. To provide sustained local delivery of the hPL-derived growth factors such as epidermal growth factor (EGF), the hPL can be loaded into biomaterials that do not degrade rapidly in vivo. Keratin (KSO), a strong filamentous protein found in human hair, when formulated as a hydrogel, is shown to sustain the release of drugs and promote wound healing. In the current study, we created a KSO biomaterial that spontaneously forms a hydrogel when rehydrated with hPL that is capable of controlled and sustained release of pro-regenerative molecules. Our study demonstrates that the release of hPL is controlled by changing the KSO hydrogel and hPL-loading concentrations, with hPL loading concentrations having a greater effect in changing release profiles. In addition, the 15% KSO concentration proved to form a stable hydrogel, and supported cell proliferation over 3 days without cytotoxic effects in vitro. The hPL-loaded keratin hydrogels show promise in potential applications for wound healing with the sustained release of pro-regenerative growth factors with easy tailoring of hydrogel properties.
Collapse
|
19
|
Nanocarrier-Mediated Topical Insulin Delivery for Wound Healing. MATERIALS 2021; 14:ma14154257. [PMID: 34361451 PMCID: PMC8348788 DOI: 10.3390/ma14154257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 07/23/2021] [Accepted: 07/25/2021] [Indexed: 12/12/2022]
Abstract
Wound care has been clinically demanding due to inefficacious treatment that represents an economic burden for healthcare systems. In Europe, approximately 7 million people are diagnosed with untreated wounds, leading to a cost between 6.000€ and 10.000€ per patient/year. In the United States of America, 1.5 million people over 65 years old suffer from chronic wounds. A promising therapeutic strategy is the use of exogenous growth factors because they are decreased at the wound site, limiting the recovery of the skin. Insulin is one of the cheapest growth factors in the market able to accelerate the re-epithelialization and stimulate angiogenesis and cell migration. However, the effectiveness of topical insulin in wound healing is hampered by the proteases in the wound bed. The encapsulation into nanoparticles improves its stability in the wound, providing adhesion to the mucosal surface and allowing its sustained release. The aim of this review is to perform a standing point about a promising strategy to treat different types of wounds by the topical delivery of insulin-loaded nanocarriers.
Collapse
|
20
|
Deng Y, Shavandi A, Okoro OV, Nie L. Alginate modification via click chemistry for biomedical applications. Carbohydr Polym 2021; 270:118360. [PMID: 34364605 DOI: 10.1016/j.carbpol.2021.118360] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/07/2021] [Accepted: 06/15/2021] [Indexed: 12/28/2022]
Abstract
Alginate biopolymers are characterized by favorable properties, of biocompatibility, degradability, and non-toxicity. However, the poor stability properties of alginate have limited its suitability for diverse applications. Recently, click chemistry has generated significant research interest due to its high reaction efficiency, high selectivity for a single product, harmless byproducts, and processing simplicity. Alginate modified using click chemistry enables the production of alginate derivatives with enhanced physical and chemical properties. Herein, we review the employment of click chemistry in the development of alginate-based materials or systems. Various click chemistries were highlighted, including azide and alkyne cycloaddition (e.g. Copper-(I)-catalyzed azide-alkyne cycloaddition (CuAAC), Strain-promoted alkyne-azide cycloaddition (SPAAC)), Diels-Alder reaction (Inverse electron demand Diels-Alder (IEDDA) cycloaddition, Tetrazine-norbornene Diels-Alder reactions), Thiol-ene/yne addition (Free-radical thiol-ene addition click reactions, Thiol-Michael addition click reactions, Thiol-yne addition click reaction), Oxime based click reactions, and other click reactions. Alginate functionalized with click chemistry and its properties were also discussed. The present study shows that click chemistry may be employed in modifying the mechanical strength, biochemical/biological properties of alginate-based materials. Finally, the applications of alginate-based materials in wound dressing, drug delivery, protein delivery, tissue regeneration, and 3D bioprinting were described and the future perspectives of alginates modified with click chemistry, are subsequently presented. This review provides new insights for readers to design structures and expand applications of alginate using click chemistry reactions in a detailed and more rational manner.
Collapse
Affiliation(s)
- Yaling Deng
- College of Intelligent Science and Control Engineering, Jinling Institute of Technology, Nanjing 211169, China
| | - Amin Shavandi
- BioMatter unit - 3BIO - École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50 - CP 165/61, 1050 Brussels, Belgium.
| | - Oseweuba Valentine Okoro
- BioMatter unit - 3BIO - École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50 - CP 165/61, 1050 Brussels, Belgium
| | - Lei Nie
- College of Life Sciences, Xinyang Normal University, Xinyang 464000, China.
| |
Collapse
|
21
|
Carleton MM, Sefton MV. Promoting endogenous repair of skeletal muscle using regenerative biomaterials. J Biomed Mater Res A 2021; 109:2720-2739. [PMID: 34041836 DOI: 10.1002/jbm.a.37239] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/14/2021] [Accepted: 05/18/2021] [Indexed: 02/06/2023]
Abstract
Skeletal muscles normally have a remarkable ability to repair themselves; however, large muscle injuries and several myopathies diminish this ability leading to permanent loss of function. No clinical therapy yet exists that reliably restores muscle integrity and function following severe injury. Consequently, numerous tissue engineering techniques, both acellular and with cells, are being investigated to enhance muscle regeneration. Biomaterials are an essential part of these techniques as they can present physical and biochemical signals that augment the repair process. Successful tissue engineering strategies require regenerative biomaterials that either actively promote endogenous muscle repair or create an environment supportive of regeneration. This review will discuss several acellular biomaterial strategies for skeletal muscle regeneration with a focus on those under investigation in vivo. This includes materials that release bioactive molecules, biomimetic materials and immunomodulatory materials.
Collapse
Affiliation(s)
- Miranda M Carleton
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Michael V Sefton
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.,Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Patel M, Jha A, Patel R. Potential application of PLGA microsphere for tissue engineering. JOURNAL OF POLYMER RESEARCH 2021. [DOI: 10.1007/s10965-021-02562-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
23
|
Biomimetic nanoengineered scaffold for enhanced full-thickness cutaneous wound healing. Acta Biomater 2021; 124:191-204. [PMID: 33508511 DOI: 10.1016/j.actbio.2021.01.029] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 01/16/2021] [Accepted: 01/19/2021] [Indexed: 12/16/2022]
Abstract
Wound healing is a complex process based on the coordinated signaling molecules and dynamic interactions between the engineered scaffold and newly formed tissue. So far, most of the engineered scaffolds used for the healing of full-thickness skin wounds do not mimic the natural extracellular matrix (ECM) complexity and therefore are not able to provide an appropriate niche for endogenous tissue regeneration [1]. To address this gap and to accelerate the wound healing process, we present biomimetic bilayer scaffolds compositing of gelatin nanofibers (GFS) and photocrosslinkable composite hydrogels loaded with epidermal growth factors (EGF). The nanofibers operate as the dermis layer, and EGF-loaded composite hydrogels acted as the epidermis matrix for the full-thickness wound healing application. The hydrogels are composed of gelatin metacryloyl (GelMA) modified with silicate nanoplatelets (Laponite). To overcome the challenges of transdermal delivery of EGF, including short half-life and lack of efficient formulation precise, controlled delivery was attained by immobilization of EGF on Laponite. It is shown that the addition of 1wt% silicate nanoplatelet increases the compressive modulus of the hydrogels by 170%. In vitro wound closure analysis also demonstrated improved adhesion of the scaffolds to the native tissue by 3.5 folds. Moreover, the tunable hemostatic ability of the scaffolds due to the negatively charged nanoplatelets is shown. In an established excisional full-thickness wound model, an enhanced wound closure (up to 93.1 ± 1.5%) after 14 days relative to controls (GFS and saline-treated groups) is demonstrated. The engineered adhesive and hemostatic scaffolds with sustained release of the growth factors have the potential to stimulate complete skin regeneration for full-thickness wound healing.
Collapse
|
24
|
Wei S, Wang W, Li L, Meng HY, Feng CZ, Dong YY, Fang XC, Dong QQ, Jiang W, Xin HL, Li ZZ, Wang X. Recombinant human epidermal growth factor combined with vacuum sealing drainage for wound healing in Bama pigs. Mil Med Res 2021; 8:18. [PMID: 33685528 PMCID: PMC7941968 DOI: 10.1186/s40779-021-00308-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 02/08/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Vacuum sealing drainage (VSD) and epidermal growth factor (EGF) both play an important role in the treatment of wounds. This study aims to explore the effects of the combination of VSD and EGF on wound healing and the optimal concentration and time of EGF. METHODS We tested the proliferation and migration capacity of HaCaT and L929 cells at different EGF concentrations (0, 1, 5, 10, and 100 ng/ml) and different EGF action times (2, 10, and 30 min). A full-thickness skin defect model was established using male, 30-week-old Bama pigs. The experiment included groups as follows: routine dressing change after covering with sterile auxiliary material (Control), continuous negative pressure drainage of the wound (VSD), continuous negative pressure drainage of the wound and injection of EGF 10 min followed by removal by continuous lavage (V + E 10 min), and continuous negative pressure drainage of the wound and injection of EGF 30 min followed by removal by continuous lavage (V + E 30 min). The wound healing rate, histological repair effect and collagen deposition were compared among the four groups. RESULTS An EGF concentration of 10 ng/ml and an action time of 10 min had optimal effects on the proliferation and migration capacities of HaCaT and L929 cells. The drug dispersion effect was better than drug infusion after bolus injection effect, and the contact surface was wider. Compared with other groups, the V + E 10 min group promoted wound healing to the greatest extent and obtained the best histological score. CONCLUSIONS A recombinant human epidermal growth factor (rhEGF) concentration of 10 ng/ml can promote the proliferation and migration of epithelial cells and fibroblasts to the greatest extent in vitro. VSD combined with rhEGF kept in place for 10 min and then washed, can promote wound healing better than the other treatments in vivo.
Collapse
Affiliation(s)
- Shuai Wei
- Institute of Orthopaedics, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Chinese PLA General Hospital, Beijing, 100583, China.,Zhoushan Dinghai Guanghua Hospital, Zhoushan, 316000, China.,Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Wei Wang
- Geriatric Neurological Department of the Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China
| | - Li Li
- Department of Orthopedics, Traditional Chinese Medical Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
| | - Hao-Ye Meng
- Institute of Orthopaedics, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Chinese PLA General Hospital, Beijing, 100583, China
| | - Chun-Zhen Feng
- Department of Stomatology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yu-Ying Dong
- Department of Plastic Surgery, General Hospital of Taiyuan Iron and Steel Limited Company, Taiyuan, 030009, China
| | - Xi-Chi Fang
- Hand Microsurgery Department, Shenzhen People's Hospital, Shenzhen, 518020, China
| | - Qi-Qiang Dong
- Third Surgery Department, Zhengzhou Renji Hospital, Zhengzhou, 450000, China
| | - Wen Jiang
- Department of Orthopedics, the First Affiliated Hospital of Medical College, Shihezi University, Shihezi, Xinjiang, 832000, Uygur Autonomous Region, China
| | - Hai-Li Xin
- Pharmacy Department, Chinese PLA General Hospital, Beijing, 100853, China
| | - Zhan-Zhen Li
- Zhoushan Dinghai Guanghua Hospital, Zhoushan, 316000, China.
| | - Xin Wang
- Institute of Orthopaedics, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Chinese PLA General Hospital, Beijing, 100583, China. .,Zhoushan Dinghai Guanghua Hospital, Zhoushan, 316000, China.
| |
Collapse
|
25
|
Cosmetic, Biomedical and Pharmaceutical Applications of Fish Gelatin/Hydrolysates. Mar Drugs 2021; 19:md19030145. [PMID: 33800149 PMCID: PMC8000627 DOI: 10.3390/md19030145] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/27/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
There are several reviews that separately cover different aspects of fish gelatin including its preparation, characteristics, modifications, and applications. Its packaging application in food industry is extensively covered but other applications are not covered or covered alongside with those of collagen. This review is comprehensive, specific to fish gelatin/hydrolysate and cites recent research. It covers cosmetic applications, intrinsic activities, and biomedical applications in wound dressing and wound healing, gene therapy, tissue engineering, implants, and bone substitutes. It also covers its pharmaceutical applications including manufacturing of capsules, coating of microparticles/oils, coating of tablets, stabilization of emulsions and drug delivery (microspheres, nanospheres, scaffolds, microneedles, and hydrogels). The main outcomes are that fish gelatin is immunologically safe, protects from the possibility of transmission of bovine spongiform encephalopathy and foot and mouth diseases, has an economic and environmental benefits, and may be suitable for those that practice religious-based food restrictions, i.e., people of Muslim, Jewish and Hindu faiths. It has unique rheological properties, making it more suitable for certain applications than mammalian gelatins. It can be easily modified to enhance its mechanical properties. However, extensive research is still needed to characterize gelatin hydrolysates, elucidate the Structure Activity Relationship (SAR), and formulate them into dosage forms. Additionally, expansion into cosmetic applications and drug delivery is needed.
Collapse
|
26
|
Gilbert-Honick J, Iyer SR, Somers SM, Takasuka H, Lovering RM, Wagner KR, Mao HQ, Grayson WL. Engineering 3D skeletal muscle primed for neuromuscular regeneration following volumetric muscle loss. Biomaterials 2020; 255:120154. [PMID: 32562942 PMCID: PMC11192434 DOI: 10.1016/j.biomaterials.2020.120154] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/02/2020] [Accepted: 05/27/2020] [Indexed: 12/14/2022]
Abstract
Volumetric muscle loss (VML) overwhelms the native regenerative capabilities of skeletal muscle and has few effective treatments to regain lost muscle mass and function. Tissue engineered muscle constructs designed to promote neuromuscular regeneration are a promising therapeutic avenue. To date, there has been no engineered muscle construct for VML treatment that has incorporated a pharmacologic agent to promote neuromuscular regeneration. Here, we have modified electrospun fibrin microfiber bundles, which have demonstrated muscle regenerative potential, with the heparan sulfate proteoglycan, agrin, to stimulate innervation post-VML. Myoblasts cultured on microfiber bundles with either soluble or chemically tethered agrin demonstrated statistically significant increased clustering of acetylcholine receptors (AChRs) with soluble agrin displaying AChR clusters throughout the myofiber bundles, and tethered agrin displaying AChR clusters only at 10 μm from the substrate surface. Following implantation into murine VML defects for 4 weeks, constructs pre-treated with soluble or tethered agrin resulted in statistically significant increased neuromuscular junctions, regenerating myofibers, vascular infiltration, neural infiltration, and nuclear yes-associated protein (YAP) expression within the defect site compared to the control without agrin. The agrin-tethered microfiber bundles provided sustained agrin signaling within the regenerating site during the 4-week post-implantation periods and further augmented the density of regenerating myofibers in regenerated tissue with statistical significance compared to constructs with soluble agrin. These data demonstrate the neuromuscular regenerative potential of engineered muscle constructs pre-treated to induce AChR clustering with locally delivered agrin at the site of VML regeneration.
Collapse
Affiliation(s)
- Jordana Gilbert-Honick
- Translational Tissue Engineering Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA; Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Shama R Iyer
- Department of Orthopedic Surgery, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Sarah M Somers
- Translational Tissue Engineering Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA; Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Hannah Takasuka
- Translational Tissue Engineering Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA; Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Richard M Lovering
- Department of Orthopedic Surgery, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Kathryn R Wagner
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD 21205, USA; Graduate Program in Cellular and Molecular Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; Departments of Neurology and Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Hai-Quan Mao
- Translational Tissue Engineering Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA; Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Materials Science & Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Institute for NanoBioTechnology (INBT), Johns Hopkins University School of Engineering, Baltimore, MD 21218, USA
| | - Warren L Grayson
- Translational Tissue Engineering Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA; Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Materials Science & Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Institute for NanoBioTechnology (INBT), Johns Hopkins University School of Engineering, Baltimore, MD 21218, USA.
| |
Collapse
|
27
|
Piard C, Luthcke R, Kamalitdinov T, Fisher J. Sustained delivery of vascular endothelial growth factor from mesoporous calcium-deficient hydroxyapatite microparticles promotes in vitro angiogenesis and osteogenesis. J Biomed Mater Res A 2020; 109:1080-1087. [PMID: 32918524 DOI: 10.1002/jbm.a.37100] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 05/13/2020] [Accepted: 05/21/2020] [Indexed: 01/05/2023]
Abstract
Promoting the growth of blood vessels within engineered tissues remains one of the main challenge in bone tissue engineering. One way to improve angiogenesis is the use of vascular endothelial growth factor (VEGF) as it holds the ability to increase the formation of a vascular network. In the present study, collagen scaffolds with VEGF-releasing hydroxyapatite particles were fabricated, in order to engineer a material both capable of presenting an osteoconductive surface and delivering an angiogenic growth factor in a localized and sustained manner, in order to enhance osteogenesis as well as angiogenesis. To this end, we developed microparticles and characterize their size, chemical properties and Ca/P ratio to validate the formation of hydroxyapatite. We then evaluated the osteogenic potential of HAp when cultured with mesenchymal stem cells and compare it to commercially available hydroxyapatite (SBp). Finally, we characterized the encapsulation and release of VEGF in the HAp and assess the angiogenic potential of the VEGF-HAp when cultured with endothelial cells. We demonstrated the successful fabrication of calcium deficient hydroxyapatite microparticles (CDHAp), with biological properties closer to the bone than stoichiometric, commercially available hydroxyapatite. This CDHAp exhibited a well-defined 3D network of crystalline nanoplates forming mesoporous and hollow structures. The high specific area created by those structures enabled the loading of VEGF with high efficiency when compared to the loading efficiency of SBp. Furthermore, their biological performances were evaluated in vitro. Our results indicate that VEGF-CDHAp can be used to improve both osteogenesis and angiogenesis in vitro.
Collapse
Affiliation(s)
- Charlotte Piard
- Fischell Department of Bioengineering, University of Maryland, Maryland, USA
| | - Rachel Luthcke
- Fischell Department of Bioengineering, University of Maryland, Maryland, USA
| | - Timur Kamalitdinov
- Fischell Department of Bioengineering, University of Maryland, Maryland, USA
| | - John Fisher
- Fischell Department of Bioengineering, University of Maryland, Maryland, USA.,Center for Engineering Complex Tissues, University of Maryland, Maryland, USA
| |
Collapse
|
28
|
Khalil AS, Xie AW, Johnson HJ, Murphy WL. Sustained release and protein stabilization reduce the growth factor dosage required for human pluripotent stem cell expansion. Biomaterials 2020; 248:120007. [PMID: 32302801 PMCID: PMC8445021 DOI: 10.1016/j.biomaterials.2020.120007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 02/24/2020] [Accepted: 03/24/2020] [Indexed: 12/17/2022]
Abstract
Translation of human pluripotent stem cell (hPSC)-derived therapies to the clinic demands scalable, cost-effective methods for cell expansion. Culture media currently used for hPSC expansion rely on high concentrations and frequent supplementation of recombinant growth factors due to their short half-life at physiological temperatures. Here, we developed a biomaterial strategy using mineral-coated microparticles (MCMs) to sustain delivery of basic fibroblast growth factor (bFGF), a thermolabile protein critical for hPSC pluripotency and proliferation. We show that the MCMs stabilize bFGF against thermally induced activity loss and provide more efficient sustained release of active growth factor compared to polymeric carriers commonly used for growth factor delivery. Using a statistically driven optimization approach called Design of Experiments, we generated a bFGF-loaded MCM formulation that supported hPSC expansion over 25 passages without the need for additional bFGF supplementation to the media, resulting in greater than 80% reduction in bFGF usage compared to standard approaches. This materials-based strategy to stabilize and sustain delivery of a thermolabile growth factor has broad potential to reduce costs associated with recombinant protein supplements in scalable biomanufacturing of emerging cell therapies.
Collapse
Affiliation(s)
- Andrew S Khalil
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Angela W Xie
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Hunter J Johnson
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA; Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
29
|
Tu S, Wu J, Chen L, Tian Y, Qin W, Huang S, Wang R, Lin Z, Song Z. LncRNA CALB2 sponges miR-30b-3p to promote odontoblast differentiation of human dental pulp stem cells via up-regulating RUNX2. Cell Signal 2020; 73:109695. [PMID: 32565162 DOI: 10.1016/j.cellsig.2020.109695] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 06/10/2020] [Accepted: 06/10/2020] [Indexed: 12/21/2022]
Abstract
Illuminating the mechanisms of odontoblast differentiation of human dental pulp stem cells (hDPSCs) is the key to find therapeutic clues to promote odontogenesis. LncRNAs play a regulatory role in odontoblast differentiation. Here, we identified a novel lncRNA, named lncRNA CALB2. It was up-regulated in odontoblast-differentiated hDPSCs and potentially interacted with miR-30b-3p and RUNX2. Via gain- and loss-of-function approaches, we found lncRNA CALB2 significantly promoted the odontoblast differentiation of hDPSCs. Then, dual luciferase reporter assay and RNA immunoprecipitation assay revealed that both lncRNA CALB2 and RUNX2 mRNA could directly bind to miR-30b-3p via the same binding sites. Interestingly, miR-30b-3p in hDPSCs was down-regulated and RUNX2 was up-regulated during odontoblast differentiation. Moreover, lncRNA CALB2 knockdown significantly reduced the protein level of RUNX2, DSPP and DMP-1, while miR-30b-3p inhibitor rescued the reduction. Furthermore, miR-30b-3p exerted an inhibitory effect on odontoblast differentiation, which could be reversed by lncRNA CALB2. Collectively, these findings indicate that the newly identified lncRNA CALB2 acts as a miR-30b-3p sponge to regulate RUNX2 expression, thus promoting the odontoblast differentiation of hDPSCs. LncRNA CALB2/miR-30b-3p/RUNX2 axis could be a novel therapeutic target for accelerating odontogenesis.
Collapse
Affiliation(s)
- Shaoqin Tu
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong, China
| | - Jinyan Wu
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong, China
| | - Lingling Chen
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong, China
| | - Yaguang Tian
- Department of Stomatology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou 570311, Hainan, China
| | - Wei Qin
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong, China
| | - Shuheng Huang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong, China
| | - Runfu Wang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong, China
| | - Zhengmei Lin
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong, China.
| | - Zhi Song
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong, China.
| |
Collapse
|
30
|
Shanjani Y, Siebert SM, Ker DFE, Mercado-Pagán AE, Yang YP. Acoustic Patterning of Growth Factor for Three-Dimensional Tissue Engineering. Tissue Eng Part A 2020; 26:602-612. [PMID: 31950880 PMCID: PMC7310194 DOI: 10.1089/ten.tea.2019.0271] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 01/07/2020] [Indexed: 02/06/2023] Open
Abstract
Temporal and spatial presentations of biological cues are critical for tissue engineering. There is a great need in improving the incorporation of bioagent(s) (specifically growth factor(s) [GF(s)]) onto three-dimensional scaffolds. In this study, we developed a process to combine additive manufacturing (AM) technology with acoustic droplet ejection (ADE) technology to control GF distribution. More specifically, we implemented ADE to control the distribution of recombinant human bone morphogenetic protein-2 (rhBMP-2) onto polycaprolactone (PCL)-based tissue engineering constructs (TECs). Three substrates were used in this study: (1) succinimide-terminated PCL (PCL-N-hydroxysuccinimide [NHS]) as model material, (2) alkali-treated PCL (PCL-NaOH) as first control material, and (3) fibrin-coated PCL (PCL-Fibrin) as second control material. It was shown that our process enables a pattern of BMP-2 spots of ∼250 μm in diameter with ∼700 μm center-to-center spacing. An initial concentration of BMP-2 higher than 300 μg/L was required to retain a detectable amount of GF on the substrate after a wash with phosphate-buffered solution. However, to obtain detectable osteogenic differentiation of C2C12 cells, the initial concentration of BMP-2 higher than 750 μg/L was needed. The cells on PCL-NHS samples showed spatial alkaline phosphatase staining correlating with local patterns of BMP-2, although the intensity was lower than the controls (PCL-NaOH and PCL-Fibrin). Our results have demonstrated that the developed AM-ADE process holds great promise in creating TECs with highly controlled GF patterning. Impact statement The combined process of additive manufacturing with acoustic droplet ejection to control growth factor (GF) distribution across three-dimensional (3D) porous scaffolds that is presented in this study enables creating 3D tissue engineering constructs with highly controlled GF patterning. Such constructs enable temporal and spatial presentations of biological cues for enhancing cell migration and differentiation and eventually the formation of targeted tissues in vitro and in vivo.
Collapse
Affiliation(s)
- Yaser Shanjani
- Department of Orthopedic Surgery, School of Medicine, Stanford University, Stanford, California
| | - Sean Michael Siebert
- Department of Orthopedic Surgery, School of Medicine, Stanford University, Stanford, California
| | - Dai Fei Elmer Ker
- Department of Orthopedic Surgery, School of Medicine, Stanford University, Stanford, California
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, New Territories, Hong Kong SAR
| | - Angel E. Mercado-Pagán
- Department of Orthopedic Surgery, School of Medicine, Stanford University, Stanford, California
| | - Yunzhi Peter Yang
- Department of Orthopedic Surgery, School of Medicine, Stanford University, Stanford, California
- Department of Materials Science and Engineering, Stanford University, Stanford, California
- Department of Bioengineering, Stanford University, Stanford, California
| |
Collapse
|
31
|
Basiri H, Abouei Mehrizi A, Ghaee A, Farokhi M, Chekini M, Kumacheva E. Carbon Dots Conjugated with Vascular Endothelial Growth Factor for Protein Tracking in Angiogenic Therapy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:2893-2900. [PMID: 32125865 DOI: 10.1021/acs.langmuir.9b03980] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
One of the challenges of using growth factors for tissue regeneration is to monitor their biodistributions and delivery to injured tissues for minimally invasive detection. In the present study, tracking of human vascular endothelial growth factor (VEGF) was achieved by chemically linking it to photoluminescent carbon dots (CDs). Carbon dots were synthesized by the hydrothermal method and, subsequently, conjugated with VEGF using carbodiimide coupling. ELISA and western blot analysis revealed that VEGF-conjugated CDs preserve the binding affinity of VEGF to its antibodies. We also show that VEGF-conjugated CDs maintain the functionality of VEGF for tube formation and cell migration. The VEGF-conjugated CDs were also used for in vitro imaging of human umbilical vein endothelial cells. The results of this work suggest that cell-penetrating VEGF-conjugated CDs can be used for growth factor protein tracking in therapeutic and tissue engineering applications.
Collapse
Affiliation(s)
- Hamideh Basiri
- Faculty of New Sciences and Technologies, Department of Life Science Engineering, University of Tehran, Tehran 1439957131, Iran
- Department of Chemistry, University of Toronto, 80 Saint George Street, Toronto, Ontario M5S 3H6, Canada
| | - Ali Abouei Mehrizi
- Faculty of New Sciences and Technologies, Department of Life Science Engineering, University of Tehran, Tehran 1439957131, Iran
| | - Azadeh Ghaee
- Faculty of New Sciences and Technologies, Department of Life Science Engineering, University of Tehran, Tehran 1439957131, Iran
| | - Mehdi Farokhi
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Mahshid Chekini
- Department of Chemistry, University of Toronto, 80 Saint George Street, Toronto, Ontario M5S 3H6, Canada
| | - Eugenia Kumacheva
- Department of Chemistry, University of Toronto, 80 Saint George Street, Toronto, Ontario M5S 3H6, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 4 Taddle Creek Road, Toronto, Ontario M5S 3G9, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| |
Collapse
|
32
|
Kuterbekov M, Jonas AM, Glinel K, Picart C. Osteogenic Differentiation of Adipose-Derived Stromal Cells: From Bench to Clinics. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:461-474. [PMID: 32098603 DOI: 10.1089/ten.teb.2019.0225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In addition to mesenchymal stem cells, adipose-derived stem/stromal cells (ASCs) are an attractive source for a large variety of cell-based therapies. One of their most important potential applications is related to the regeneration of bone tissue thanks to their capacity to differentiate in bone cells. However, this requires a proper control of their osteogenic differentiation, which depends not only on the initial characteristics of harvested cells but also on the conditions used for their culture. In this review, we first briefly describe the preclinical and clinical trials using ASCs for bone regeneration and present the quantitative parameters used to characterize the osteogenic differentiation of ASCs. We then focus on the soluble factors influencing the osteogenic differentiation of ACS, including the steroid hormones and various growth factors, notably the most osteoinductive ones, the bone morphogenetic proteins (BMPs). Impact statement Adipose-derived stromal/stem cells are reviewed for their use in bone regeneration.
Collapse
Affiliation(s)
- Mirasbek Kuterbekov
- Institute of Condensed Matter & Nanosciences (Bio & Soft Matter), Université Catholique de Louvain, Louvain-la-Neuve, Belgium.,Grenoble Institute of Technology, University Grenoble Alpes, LMGP, Grenoble, France
| | - Alain M Jonas
- Institute of Condensed Matter & Nanosciences (Bio & Soft Matter), Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Karine Glinel
- Institute of Condensed Matter & Nanosciences (Bio & Soft Matter), Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Catherine Picart
- Grenoble Institute of Technology, University Grenoble Alpes, LMGP, Grenoble, France.,Biomimetism and Regenerative Medicine Lab, CEA, Institute of Interdisciplinary Research of Grenoble (IRIG), Université Grenoble-Alpes/CEA/CNRS, Grenoble, France
| |
Collapse
|
33
|
Natural polymeric biomaterials in growth factor delivery for treating diabetic foot ulcers. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2019.101385] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
34
|
Wang F, Hou K, Chen W, Wang Y, Wang R, Tian C, Xu S, Ji Y, Yang Q, Zhao P, Yu L, Lu Z, Zhang H, Li F, Wang H, He B, Kaplan DL, Xia Q. Transgenic PDGF-BB/sericin hydrogel supports for cell proliferation and osteogenic differentiation. Biomater Sci 2020; 8:657-672. [DOI: 10.1039/c9bm01478k] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The present study demonstrates fabrication of PDGF-BB functionalized sericin hydrogel to explore biomaterials-related utility in bone tissue engineering.
Collapse
|
35
|
Amirsadeghi A, Jafari A, Eggermont LJ, Hashemi SS, Bencherif SA, Khorram M. Vascularization strategies for skin tissue engineering. Biomater Sci 2020; 8:4073-4094. [DOI: 10.1039/d0bm00266f] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lack of proper vascularization after skin trauma causes delayed wound healing. This has sparked the development of various tissue engineering strategies to improve vascularization.
Collapse
Affiliation(s)
- Armin Amirsadeghi
- Department of Chemical Engineering
- School of Chemical and Petroleum Engineering
- Shiraz University
- Shiraz 71348-51154
- Iran
| | - Arman Jafari
- Department of Chemical Engineering
- School of Chemical and Petroleum Engineering
- Shiraz University
- Shiraz 71348-51154
- Iran
| | | | - Seyedeh-Sara Hashemi
- Burn & Wound Healing Research Center
- Shiraz University of Medical Science
- Shiraz 71345-1978
- Iran
| | - Sidi A. Bencherif
- Department of Chemical Engineering
- Northeastern University
- Boston
- USA
- Department of Bioengineering
| | - Mohammad Khorram
- Department of Chemical Engineering
- School of Chemical and Petroleum Engineering
- Shiraz University
- Shiraz 71348-51154
- Iran
| |
Collapse
|
36
|
Vijayan A, A S, Kumar GSV. PEG grafted chitosan scaffold for dual growth factor delivery for enhanced wound healing. Sci Rep 2019; 9:19165. [PMID: 31844069 PMCID: PMC6915706 DOI: 10.1038/s41598-019-55214-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/04/2019] [Indexed: 01/06/2023] Open
Abstract
Application of growth factors at wound site has improved the efficiency and quality of healing. Basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF) induce proliferation of various cells in wound healing. Delivery of growth factor from controlled release systems protect it from degradation and also result in sustained delivery of it at the site of injury. The goal of the study was to develop a Polyethylene glycol (PEG) cross-linked cotton-like chitosan scaffold (CS-PEG-H) by freeze-drying method and chemically conjugate heparin to the scaffold to which the growth factors can be electrostatically bound and evaluate its wound healing properties in vitro and in vivo. The growth factor containing scaffolds induced increased proliferation of HaCaT cells, increased neovascularization and collagen formation seen by H and E and Masson's trichrome staining. Immunohistochemistry was performed using the Ki67 marker which increased proliferation of cells in growth factor containing scaffold treated group. Frequent dressing changes are a major deterrent to proper wound healing. Our system was found to release both VEGF and bFGF in a continuous manner and attained stability after 7 days. Thus our system can maintain therapeutic levels of growth factor at the wound bed thereby avoiding the need for daily applications and frequent dressing changes. Thus, it can be a promising candidate for wound healing.
Collapse
Affiliation(s)
- Amritha Vijayan
- Cancer Biology, Nano Drug Delivery Systems (NDDS), Bio-Innovation Center (BIC), Rajiv Gandhi Centre for Biotechnology, Thycaud P.O, Thiruvananthapuram, Kerala, 695014, India
- Research Scholar, Department of Biotechnology, Faculty of Applied Science & Technology, University of Kerala, Trivandrum, Kerala, 695581, India
| | - Sabareeswaran A
- Histopathology laboratory, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, 695011, India
| | - G S Vinod Kumar
- Cancer Biology, Nano Drug Delivery Systems (NDDS), Bio-Innovation Center (BIC), Rajiv Gandhi Centre for Biotechnology, Thycaud P.O, Thiruvananthapuram, Kerala, 695014, India.
| |
Collapse
|
37
|
Development of hydrogel-like biomaterials via nanoparticle assembly and solid-hydrogel transformation. J Control Release 2019; 318:185-196. [PMID: 31857102 DOI: 10.1016/j.jconrel.2019.12.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/07/2019] [Accepted: 12/15/2019] [Indexed: 11/22/2022]
Abstract
Hydrogels for biomedical applications such as controlled drug release are usually synthesized with the chemical or physical crosslinking of monomers or macromers. In this work, we used gelatin to prepare hydrogel nanoparticles and studied whether gelatin nanoparticles (GNPs) could assemble to form a solid biomaterial and whether this solid biomaterial was capable of transforming into a hydrogel upon introduction to a hydrated environment. The data show that GNPs with or without aptamer functionalization could form a nanoparticle-assembled porous solid biomaterial after freezing and lyophilization treatment. This formation did not need any additional crosslinking reactions. More importantly, this solid biomaterial could undergo solid-to-hydrogel transition after contacting a solution and this transformation was tunable to match different shapes and geometries of defined molds. The formed hydrogel could also sequester and release growth factors for the promotion of skin wound healing. Thus, GNP-assembled solid biomaterials hold great potential as an off-the-shelf therapy for biomedical application such as drug delivery and regenerative medicine.
Collapse
|
38
|
Gaspar D, Peixoto R, De Pieri A, Striegl B, Zeugolis DI, Raghunath M. Local pharmacological induction of angiogenesis: Drugs for cells and cells as drugs. Adv Drug Deliv Rev 2019; 146:126-154. [PMID: 31226398 DOI: 10.1016/j.addr.2019.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 05/12/2019] [Accepted: 06/16/2019] [Indexed: 12/12/2022]
Abstract
The past decades have seen significant advances in pro-angiogenic strategies based on delivery of molecules and cells for conditions such as coronary artery disease, critical limb ischemia and stroke. Currently, three major strategies are evolving. Firstly, various pharmacological agents (growth factors, interleukins, small molecules, DNA/RNA) are locally applied at the ischemic region. Secondly, preparations of living cells with considerable bandwidth of tissue origin, differentiation state and preconditioning are delivered locally, rarely systemically. Thirdly, based on the notion, that cellular effects can be attributed mostly to factors secreted in situ, the cellular secretome (conditioned media, exosomes) has come into the spotlight. We review these three strategies to achieve (neo)angiogenesis in ischemic tissue with focus on the angiogenic mechanisms they tackle, such as transcription cascades, specific signalling steps and cellular gases. We also include cancer-therapy relevant lymphangiogenesis, and shall seek to explain why there are often conflicting data between in vitro and in vivo. The lion's share of data encompassing all three approaches comes from experimental animal work and we shall highlight common technical obstacles in the delivery of therapeutic molecules, cells, and secretome. This plethora of preclinical data contrasts with a dearth of clinical studies. A lack of adequate delivery vehicles and standardised assessment of clinical outcomes might play a role here, as well as regulatory, IP, and manufacturing constraints of candidate compounds; in addition, completed clinical trials have yet to reveal a successful and efficacious strategy. As the biology of angiogenesis is understood well enough for clinical purposes, it will be a matter of time to achieve success for well-stratified patients, and most probably with a combination of compounds.
Collapse
Affiliation(s)
- Diana Gaspar
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Rita Peixoto
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Andrea De Pieri
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Proxy Biomedical Ltd., Coilleach, Spiddal, Galway, Ireland
| | - Britta Striegl
- Competence Centre Tissue Engineering for Drug Development (TEDD), Centre for Cell Biology & Tissue Engineering, Institute for Chemistry and Biotechnology, Zurich University of Applied Sciences, Zurich, Switzerland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Michael Raghunath
- Competence Centre Tissue Engineering for Drug Development (TEDD), Centre for Cell Biology & Tissue Engineering, Institute for Chemistry and Biotechnology, Zurich University of Applied Sciences, Zurich, Switzerland.
| |
Collapse
|
39
|
Thermo-sensitive gellan maleate/N-isopropylacrylamide hydrogels: initial “in vitro” and “in vivo” evaluation as ocular inserts. Polym Bull (Berl) 2019. [DOI: 10.1007/s00289-019-02772-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
40
|
Orth M, Shenar AK, Scheuer C, Braun BJ, Herath SC, Holstein JH, Histing T, Yu X, Murphy WL, Pohlemann T, Laschke MW, Menger MD. VEGF-loaded mineral-coated microparticles improve bone repair and are associated with increased expression of epo and RUNX-2 in murine non-unions. J Orthop Res 2019; 37:821-831. [PMID: 30835895 DOI: 10.1002/jor.24267] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 02/12/2019] [Indexed: 02/04/2023]
Abstract
A poor vascular supply of the fracture gap is a key factor for the development of atrophic non-unions. Mineral-coated microparticles (MCM) represent a sophisticated carrier system for the delivery of vascular endothelial growth factor (VEGF). Hence, we investigated whether VEGF-loaded MCM improve bone repair in non-unions. For this purpose, we analyzed binding and release kinetics of MCM for VEGF in vitro. Moreover, we applied VEGF-loaded or -unloaded MCM in a murine non-union model in vivo and studied the process of bone healing by means of biomechanical, radiological, histomorphometric, and Western blot techniques. MCM-free non-unions served as controls. The binding efficiency of MCM for VEGF was 46 ± 3% and the release profile revealed an initial minor burst release followed by a sustained release over a 50-day study period, thus, mimicking the physiological expression profile of VEGF during bone healing. In vivo, bone defects treated with VEGF-loaded MCM exhibited a higher bending stiffness, a higher fraction of bone volume/tissue volume and a larger callus area on days 14 and 70 when compared to the other groups. Western blot analyses on day 14 revealed a higher expression of VEGF, erythropoietin (EPO), and runt-related transcription factor 2, but not of EPO-receptor in bone defects treated with VEGF-loaded MCM. These findings demonstrate that the use of MCM for VEGF delivery shows great potential due to the ability to maintain protein stability and functionality in vivo. Moreover, the application of VEGF-loaded MCM represent a promising strategy for the treatment of non-unions. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
- Marcel Orth
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany.,Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Amira K Shenar
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany.,Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Claudia Scheuer
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Benedikt J Braun
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Steven C Herath
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Jörg H Holstein
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany.,Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Tina Histing
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany.,Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Xiaohua Yu
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | - Tim Pohlemann
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| |
Collapse
|
41
|
Vijayan A, James PP, Nanditha CK, Kumar GSV. Multiple cargo deliveries of growth factors and antimicrobial peptide using biodegradable nanopolymer as a potential wound healing system. Int J Nanomedicine 2019; 14:2253-2263. [PMID: 30992665 PMCID: PMC6445221 DOI: 10.2147/ijn.s190321] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Treatment of wounds with the help of nanoparticles (NPs) is more effective and superior in comparison to traditional wound healing methods as it protects and sustains active drug release at the wound site thus enhancing the safety of the drug and reducing the possibility of side effects. The advantages of this method are the possibility of allowing a reduction in administered dose, limiting toxicity levels to the minimum, and increasing safety of topical delivery of the drug. Materials and methods We report the synthesis of a novel poly (lactic-co-glycolic acid) (PLGA) NP-based multicargo delivery system for growth factors and antimicrobial peptide. Growth factors vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) were entrapped in PLGA NPs by solvent diffusion method and an antimicrobial peptide (K4) was conjugated to the NP by carbodiimide chemistry. The developed multiple cargo delivery systems with growth factors (VEGF and bFGF) and an antimicrobial peptide (K4) were investigated and optimized for potential wound healing. Results The system showed a sustained release of growth factors and was evaluated for cytotoxicity by MTT and live/dead assay, which revealed that the bioactivity of the growth factor-entrapped NPs was higher than that of free growth factors, and it also induced enhanced cell proliferation in vitro. Conclusion The development of a system for the codelivery of growth factors (VEGF and bFGF) and an antimicrobial peptide (K4) was investigated for potential wound healing application. The entrapment of growth factors with very high efficiency is an advantage in this method along with its sustained release from the nanoparticulate system, which will enhance the angiogenesis. Our system also displayed broad-spectrum antimicrobial activity against both gram-positive and gram-negative bacteria.
Collapse
Affiliation(s)
- Amritha Vijayan
- Chemical Biology, Nano Drug Delivery Systems (NDDS), Bio-Innovation Center (BIC), Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala 695014, India,
| | - Pinky Prabha James
- Chemical Biology, Nano Drug Delivery Systems (NDDS), Bio-Innovation Center (BIC), Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala 695014, India,
| | - C K Nanditha
- Chemical Biology, Nano Drug Delivery Systems (NDDS), Bio-Innovation Center (BIC), Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala 695014, India,
| | - G S Vinod Kumar
- Chemical Biology, Nano Drug Delivery Systems (NDDS), Bio-Innovation Center (BIC), Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala 695014, India,
| |
Collapse
|
42
|
Nakayama KH, Shayan M, Huang NF. Engineering Biomimetic Materials for Skeletal Muscle Repair and Regeneration. Adv Healthc Mater 2019; 8:e1801168. [PMID: 30725530 PMCID: PMC6589032 DOI: 10.1002/adhm.201801168] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/21/2018] [Indexed: 11/12/2022]
Abstract
Although skeletal muscle is highly regenerative following injury or disease, endogenous self-regeneration is severely impaired in conditions of volume traumatic muscle loss. Consequently, tissue engineering approaches are a promising means to regenerate skeletal muscle. Biological scaffolds serve as not only structural support for the promotion of cellular ingrowth but also impart potent modulatory signaling cues that may be beneficial for tissue regeneration. In this work, the progress of tissue engineering approaches for skeletal muscle engineering and regeneration is overviewed, with a focus on the techniques to create biomimetic engineered tissue using extracellular cues. These factors include mechanical and electrical stimulation, geometric patterning, and delivery of growth factors or other bioactive molecules. The progress of evaluating the therapeutic efficacy of these approaches in preclinical models of muscle injury is further discussed.
Collapse
Affiliation(s)
- Karina H Nakayama
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Veterans Affairs Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA, 94304, USA
- The Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
| | - Mahdis Shayan
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Veterans Affairs Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA, 94304, USA
- The Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
| | - Ngan F Huang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Veterans Affairs Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA, 94304, USA
- The Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
43
|
Rousselle SD, Ramot Y, Nyska A, Jackson ND. Pathology of Bioabsorbable Implants in Preclinical Studies. Toxicol Pathol 2019; 47:358-378. [DOI: 10.1177/0192623318816681] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Bioabsorbable implants can be advantageous for certain surgical tissue bioengineering applications and implant-assisted tissue repair. They offer the obvious benefits of nonpermanence and eventual restoration of the native tissue’s biomechanical and immunological properties, while providing a structural scaffold for healing and a route for additional therapies (i.e., drug elution). They present unique developmental, imaging, and histopathological challenges in the conduct of preclinical animal studies and in interpretation of pathology data. The bioabsorption process is typically associated with a gradual decline (over months to years) in structural strength and integrity and may also be associated with cellular responses such as phagocytosis that may confound interpretation of efficacy and safety end points. Additionally, as these implants bioabsorb, they become increasingly difficult to isolate histologically and thus imaging modalities such as microCT become very valuable to determine the original location of the implants and to assess the remodeling response in tandem with histopathology. In this article, we will review different types of bioabsorbable implants and commonly used bioabsorbable materials; additionally, we will address some of the most common challenges and pitfalls confronting histologists and pathologists in collecting, handling, imaging, preparing tissues through histology, evaluating, and interpreting study data associated with bioabsorbable implants.
Collapse
Affiliation(s)
| | - Yuval Ramot
- Hadassah—Hebrew University Medical Center, Jerusalem, Israel
| | | | | |
Collapse
|
44
|
Omidi M, Hashemi M, Tayebi L. Microfluidic synthesis of PLGA/carbon quantum dot microspheres for vascular endothelial growth factor delivery. RSC Adv 2019; 9:33246-33256. [PMID: 35529135 PMCID: PMC9073357 DOI: 10.1039/c9ra06279c] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 10/08/2019] [Indexed: 11/21/2022] Open
Abstract
In this study, vascular endothelial growth factor (VEGF) loaded poly(d,l-lactide-co-glycolide) (PLGA) – carbon quantum dot microspheres were produced using microfluidic platforms.
Collapse
Affiliation(s)
- Meisam Omidi
- Marquette University School of Dentistry
- Milwaukee
- USA
| | - Mohadeseh Hashemi
- Department of Biomedical Engineering
- The University of Texas at Austin
- Austin
- USA
- Division of Pharmaceutics
| | - Lobat Tayebi
- Marquette University School of Dentistry
- Milwaukee
- USA
| |
Collapse
|
45
|
|
46
|
Fabrication of the FGF1-functionalized sericin hydrogels with cell proliferation activity for biomedical application using genetically engineered Bombyx mori (B. mori) silk. Acta Biomater 2018; 79:239-252. [PMID: 30149211 DOI: 10.1016/j.actbio.2018.08.031] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/21/2018] [Accepted: 08/23/2018] [Indexed: 12/28/2022]
Abstract
Sericin, as the major component of Bombyx mori silk, is a useful biomaterial for tissue engineering due to its hydrophilicity, biocompatibility and biodegradability. Here, we report the fabrication of a human acidic fibroblast growth factor (FGF1)-functionalized sericin hydrogel using a transgenic silkworm spun silk with FGF1 incorporated in its sericin layer. Sericin, together with FGF1, were simultaneously extracted from the silk fiber and then exposed to cold-induced hydrogel formation without additional crosslinking. The fabricated FGF1 sericin hydrogels demonstrated injectability, useful mechanical properties and a porous microstructure, which contributed to cell adhesion and survival. In addition, FGF1 achieved long-term storage in the sericin hydrogels over a wide range of temperatures. Further, the sericin-FGF1 demonstrated sustained release to promote cell proliferation and wound healing. Furthermore, cellular inflammatory responses showed that the FGF1 sericin hydrogels exhibited biocompatibility and no immunogenicity. This study revealed the successful exploration of FGF1-functionalized sericin hydrogels as a new protein-based biomaterial to expand applications of FGF1 and sericin in tissue and medical engineering. Further, we demonstrated a strategy for the predesign of exogenous protein-functionalized sericin hydrogels through genetically modifying silk fibers as sources for their cost effective production at a large scale. STATEMENT OF SIGNIFICANCE Sericin from the Bombyx mori silk, is regarded as a desirable biomaterial for tissue engineering due to its hydrophilicity, biocompatibility and biodegradability. Genetically engineering the sericin with functional exogenous proteins would enhance its biofunctions and further expand its application in tissue engineering. In this study, we demonstrated a method to fabricate a human acidic fibroblast growth factor (FGF1)-functionalized sericin hydrogel using a transgenic silkworm spun silk with FGF1 incorporated in its sericin layer. The fabricated FGF1 sericin hydrogels demonstrated injectability, porous microstructure, biocompatibility and no immunogenicity which contributed to cell adhesion and survival. Remarkably, FGF1 could achieve a long-term stability in the sericin hydrogels over a wide range of temperatures and sustained release to promote cell proliferation and wound healing. This study revealed the successful exploration of FGF1-functionalized sericin hydrogels as a new protein-based biomaterial in tissue and medical engineering application, and provided a strategy for the predesign of exogenous protein-functionalized sericin hydrogels through genetically modifying silk fibers as sources for their cost effective production at a large scale.
Collapse
|
47
|
Abstract
Programmable hydrogels are defined as hydrogels that are able to change their properties and functions periodically, reversibly and/or sequentially on demand. They are different from those responsive hydrogels whose changes are passive or cannot be stopped or reversed once started and vice versa. The purpose of this review is to summarize major progress in developing programmable hydrogels from the viewpoints of principles, functions and biomedical applications. The principles are first introduced in three categories including biological, chemical and physical stimulation. With the stimulation, programmable hydrogels can undergo functional changes in dimension, mechanical support, cell attachment and molecular sequestration, which are introduced in the middle of this review. The last section is focused on the introduction and discussion of four biomedical applications including mechanistic studies in mechanobiology, tissue engineering, cell separation and protein delivery.
Collapse
Affiliation(s)
- Yong Wang
- Department of Biomedical Engineering, The Pennsylvania State University University Park, PA 16802, USA.
| |
Collapse
|
48
|
Abstract
Polymeric chains crosslinked through supramolecular interactions-directional and reversible non-covalent interactions-compose an emerging class of modular and tunable biomaterials. The choice of chemical moiety utilized in the crosslink affords different thermodynamic and kinetic parameters of association, which in turn illustrate the connectivity and dynamics of the system. These parameters, coupled with the choice of polymeric architecture, can then be engineered to control environmental responsiveness, viscoelasticity, and cargo diffusion profiles, yielding advanced biomaterials which demonstrate rapid shear-thinning, self-healing, and extended release. In this review we examine the relationship between supramolecular crosslink chemistry and biomedically relevant macroscopic properties. We then describe how these properties are currently leveraged in the development of materials for drug delivery, immunology, regenerative medicine, and 3D-bioprinting (253 references).
Collapse
Affiliation(s)
- Joseph L Mann
- Department of Materials Science and Engineering, Stanford University, 476 Lomita Mall, Stanford, CA 94305, USA.
| | | | | | | |
Collapse
|
49
|
Rahman SU, Nagrath M, Ponnusamy S, Arany PR. Nanoscale and Macroscale Scaffolds with Controlled-Release Polymeric Systems for Dental Craniomaxillofacial Tissue Engineering. MATERIALS (BASEL, SWITZERLAND) 2018; 11:E1478. [PMID: 30127246 PMCID: PMC6120038 DOI: 10.3390/ma11081478] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/03/2018] [Accepted: 08/10/2018] [Indexed: 12/11/2022]
Abstract
Tremendous progress in stem cell biology has resulted in a major current focus on effective modalities to promote directed cellular behavior for clinical therapy. The fundamental principles of tissue engineering are aimed at providing soluble and insoluble biological cues to promote these directed biological responses. Better understanding of extracellular matrix functions is ensuring optimal adhesive substrates to promote cell mobility and a suitable physical niche to direct stem cell responses. Further, appreciation of the roles of matrix constituents as morphogen cues, termed matrikines or matricryptins, are also now being directly exploited in biomaterial design. These insoluble topological cues can be presented at both micro- and nanoscales with specific fabrication techniques. Progress in development and molecular biology has described key roles for a range of biological molecules, such as proteins, lipids, and nucleic acids, to serve as morphogens promoting directed behavior in stem cells. Controlled-release systems involving encapsulation of bioactive agents within polymeric carriers are enabling utilization of soluble cues. Using our efforts at dental craniofacial tissue engineering, this narrative review focuses on outlining specific biomaterial fabrication techniques, such as electrospinning, gas foaming, and 3D printing used in combination with polymeric nano- or microspheres. These avenues are providing unprecedented therapeutic opportunities for precision bioengineering for regenerative applications.
Collapse
Affiliation(s)
- Saeed Ur Rahman
- Departments of Oral Biology and Biomedical Engineering, School of Dentistry, University at Buffalo, Buffalo, NY 14214, USA.
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad, Lahore Campus, Lahore 54000, Pakistan.
| | - Malvika Nagrath
- Departments of Oral Biology and Biomedical Engineering, School of Dentistry, University at Buffalo, Buffalo, NY 14214, USA.
- Department of Biomedical Engineering, Ryerson University, Toronto, ON M5B 2K3, Canada.
| | - Sasikumar Ponnusamy
- Departments of Oral Biology and Biomedical Engineering, School of Dentistry, University at Buffalo, Buffalo, NY 14214, USA.
| | - Praveen R Arany
- Departments of Oral Biology and Biomedical Engineering, School of Dentistry, University at Buffalo, Buffalo, NY 14214, USA.
| |
Collapse
|
50
|
Hassani Besheli N, Damoogh S, Zafar B, Mottaghitalab F, Motasadizadeh H, Rezaei F, Shokrgozar MA, Farokhi M. Preparation of a Codelivery System Based on Vancomycin/Silk Scaffold Containing Silk Nanoparticle Loaded VEGF. ACS Biomater Sci Eng 2018; 4:2836-2846. [DOI: 10.1021/acsbiomaterials.8b00149] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Negar Hassani Besheli
- School of Chemical Engineering, Collage of Engineering, University of Tehran, P.O. Box 11155-4563, Tehran 1417466191, Iran
| | - Sheyda Damoogh
- National Cell Bank of Iran, Pasteur Institute of Iran, P.O. Box 1316943551, Tehran 1316943551, Iran
| | - Bahareh Zafar
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 14177-55469, Iran
| | - Fatemeh Mottaghitalab
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran 1417613151, Iran
| | - Hamidreza Motasadizadeh
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Fatemeh Rezaei
- Faculty of Biomedical Engineering, Amirkabir University of Technology, Tehran 15875/4413, Iran
| | - Mohammad Ali Shokrgozar
- National Cell Bank of Iran, Pasteur Institute of Iran, P.O. Box 1316943551, Tehran 1316943551, Iran
| | - Mehdi Farokhi
- National Cell Bank of Iran, Pasteur Institute of Iran, P.O. Box 1316943551, Tehran 1316943551, Iran
| |
Collapse
|