1
|
Du P, Tao X, Harati J, Shi Y, Xiao L, Li X, Pan H, Wang PY. Human platelet lysate enhances small lipid droplet accumulation of human MSCs through MAPK phosphorylation. Stem Cell Res Ther 2024; 15:473. [PMID: 39696689 DOI: 10.1186/s13287-024-04085-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Human platelet lysate (hPL) has emerged as a promising serum substitute to enhance the self-renewal and multipotency of human mesenchymal stem cells (MSCs). Despite its potential, the specific biological mechanisms by which hPL influences MSC phenotypes remain inadequately understood. METHODS We investigated the biological signaling activated by hPL in two common types of human MSCs: bone marrow-derived MSCs (BMSCs) and adipose-derived MSCs (ASCs). Cell adhesion and cell-matrix interaction were assessed through immunofluorescence staining and western blotting. The impact of hPL on lipid droplet formation in MSCs was thoroughly examined using oil red O/BODIPY staining, semi-quantitative analysis, and qRT-PCR. RNA sequencing and intracellular inhibition assays were also performed to elucidate the mechanisms by which hPL modulates MSC behavior. RESULTS MSCs cultured in hPL medium demonstrated a reduction in cell size, spreading area, and vinculin puncta, while enhancing cell proliferation and lipid droplet accumulation compared to those cultured in control media. Notably, the lipid droplets in hPL-treated MSCs were significantly smaller than those in adipocyte-like cells differentiated from MSCs, highlighting hPL's distinctive role in lipid production. Gene and protein expression profiles of hPL-treated MSCs differed from those in adipocyte-like cells. An angiogenic factor array revealed that hPL-MSCs had a distinct angiogenic factor profile compared to FBS-MSCs, with VEGF expression closely linked to HIF-1α expression. RNA-seq data identified approximately 1,900 differentially expressed genes (DEGs) between hPL-MSCs and FBS-MSCs, with enrichment in focal adhesion, ECM-receptor interaction, and PI3K-Akt/MAPK signaling pathways. Inhibition of MAPK phosphorylation significantly hampered lipid formation in hPL-MSCs, underscoring the pivotal role of MAPK signaling in hPL-driven adipogenesis. CONCLUSION This study reveals the biological mechanisms by which hPL infleunces MSC behavior and differentiation, offering new insights into its potential application in regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Ping Du
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Xuelian Tao
- Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Javad Harati
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yue Shi
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Liang Xiao
- Department of Surgery and Oncology, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, China
| | - Xian Li
- Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Haobo Pan
- Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China.
| | - Peng-Yuan Wang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China.
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
2
|
Shaygani H, Mofrad YM, Demneh SMR, Hafezi S, Almasi-Jaf A, Shamloo A. Cartilage and bone injectable hydrogels: A review of injectability methods and treatment strategies for repair in tissue engineering. Int J Biol Macromol 2024; 282:136689. [PMID: 39447779 DOI: 10.1016/j.ijbiomac.2024.136689] [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: 12/27/2023] [Revised: 10/08/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024]
Abstract
Cartilage and bone are crucial tissues causing disability in the elderly population, often requiring prolonged treatment and surgical intervention due to limited regenerative capacity. Injectable hydrogels that closely mimic the extracellular matrix (ECM) of native hard tissue have attracted attention due to their minimally invasive application and ability to conform to irregular defect sites. These hydrogels facilitate key biological processes such as cell migration, chondrogenesis in cartilage repair, osteoinduction, angiogenesis, osteoconduction, and mineralization in bone repair. This review analyzes in-vitro and in-vivo biomedical databases over the past decade to identify advancements in hydrogel formulations, crosslinking mechanisms, and biomaterial selection for cartilage and bone tissue engineering. The review emphasizes the effectiveness of injectable hydrogels as carriers for cells, growth factors, and drugs, offering additional therapeutic benefits. The relevance of these findings is discussed in the context of their potential to serve as a robust alternative to current surgical and non-surgical treatments. This review also examines the advantages of injectable hydrogels, such as ease of administration, reduced patient recovery time, and enhanced bioactivity, thereby emphasizing their potential in clinical applications for cartilage and bone regeneration with emphasis on addressing the shortcomings of current treatments.
Collapse
Affiliation(s)
- Hossein Shaygani
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, Iran
| | - Yasaman Mozhdehbakhsh Mofrad
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, Iran; School of Mechanical Engineering, Iran University of Science and Technology, Tehran, Iran
| | - Seyed Mohammadhossein Rezaei Demneh
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, Iran
| | - Shayesteh Hafezi
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - Aram Almasi-Jaf
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, Iran
| | - Amir Shamloo
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, Iran.
| |
Collapse
|
3
|
Grivet-Brancot A, Buscemi M, Ciardelli G, Bronco S, Sartori S, Cassino C, Al Kayal T, Losi P, Soldani G, Boffito M. Cord Blood Platelet Lysate-Loaded Thermo-Sensitive Hydrogels for Potential Treatment of Chronic Skin Wounds. Pharmaceutics 2024; 16:1438. [PMID: 39598561 PMCID: PMC11597581 DOI: 10.3390/pharmaceutics16111438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/26/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND/OBJECTIVES Chronic skin wounds (CSWs) are a worldwide healthcare problem with relevant impacts on both patients and healthcare systems. In this context, innovative treatments are needed to improve tissue repair and patient recovery and quality of life. Cord blood platelet lysate (CB-PL) holds great promise in CSW treatment thanks to its high growth factors and signal molecule content. In this work, thermo-sensitive hydrogels based on an amphiphilic poly(ether urethane) (PEU) were developed as CB-PL carriers for CSW treatment. METHODS A Poloxamer 407®-based PEU was solubilized in aqueous medium (10 and 15% w/v) and added with CB-PL at a final concentration of 20% v/v. Hydrogels were characterized for their gelation potential, rheological properties, and swelling/dissolution behavior in a watery environment. CB-PL release was also tested, and the bioactivity of released CB-PL was evaluated through cell viability, proliferation, and migration assays. RESULTS PEU aqueous solutions with concentrations in the range 10-15% w/v exhibited quick (within a few minutes) sol-to-gel transition at around 30-37 °C and rheological properties modulated by the PEU concentration. Moreover, CB-PL loading within the gels did not affect the overall gel properties. Stability in aqueous media was dependent on the PEU concentration, and payload release was completed between 7 and 14 days depending on the polymer content. The CB-PL-loaded hydrogels also showed biocompatibility and released CB-PL induced keratinocyte migration and proliferation, with scratch wound recovery similar to the positive control (i.e., CB-PL alone). CONCLUSIONS The developed hydrogels represent promising tools for CSW treatment, with tunable gelation properties and residence time and the ability to encapsulate and deliver active biomolecules with sustained and controlled kinetics.
Collapse
Affiliation(s)
- Arianna Grivet-Brancot
- Institute for Chemical-Physical Processes, National Research Council, 56124 Pisa, Italy; (A.G.-B.); (S.B.)
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Torino, Italy;
| | - Marianna Buscemi
- Institute of Clinical Physiology, National Research Council, Massa, 56124 Pisa, Italy; (M.B.); (T.A.K.); (P.L.); (G.S.)
| | - Gianluca Ciardelli
- Institute for Chemical-Physical Processes, National Research Council, 56124 Pisa, Italy; (A.G.-B.); (S.B.)
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Torino, Italy;
| | - Simona Bronco
- Institute for Chemical-Physical Processes, National Research Council, 56124 Pisa, Italy; (A.G.-B.); (S.B.)
| | - Susanna Sartori
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Torino, Italy;
| | - Claudio Cassino
- Department of Science and Technological Innovation, Università del Piemonte Orientale, 15121 Alessandria, Italy;
| | - Tamer Al Kayal
- Institute of Clinical Physiology, National Research Council, Massa, 56124 Pisa, Italy; (M.B.); (T.A.K.); (P.L.); (G.S.)
| | - Paola Losi
- Institute of Clinical Physiology, National Research Council, Massa, 56124 Pisa, Italy; (M.B.); (T.A.K.); (P.L.); (G.S.)
| | - Giorgio Soldani
- Institute of Clinical Physiology, National Research Council, Massa, 56124 Pisa, Italy; (M.B.); (T.A.K.); (P.L.); (G.S.)
| | - Monica Boffito
- Institute for Chemical-Physical Processes, National Research Council, 56124 Pisa, Italy; (A.G.-B.); (S.B.)
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Torino, Italy;
| |
Collapse
|
4
|
Torabi Rahvar P, Abdekhodaie MJ, Jooybar E, Gantenbein B. An enzymatically crosslinked collagen type II/hyaluronic acid hybrid hydrogel: A biomimetic cell delivery system for cartilage tissue engineering. Int J Biol Macromol 2024; 279:134614. [PMID: 39127277 DOI: 10.1016/j.ijbiomac.2024.134614] [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: 12/27/2023] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
This study presents new injectable hydrogels based on hyaluronic acid and collagen type II that mimic the polysaccharide-protein structure of natural cartilage. After collagen isolation from chicken sternal cartilage, tyramine-grafted hyaluronic acid and collagen type II (HA-Tyr and COL-II-Tyr) were synthesized. Hybrid hydrogels were prepared with different ratios of HA-Tyr/COL-II-Tyr using horseradish peroxidase and noncytotoxic concentrations of hydrogen peroxide to encapsulate human bone marrow-derived mesenchymal stromal cells (hBM-MSCs). The findings showed that a higher HA-Tyr content resulted in a higher storage modulus and a lower hydrogel shrinkage, resulting in hydrogel swelling. Incorporating COL-II-Tyr into HA-Tyr hydrogels induced a more favorable microenvironment for hBM-MSCs chondrogenic differentiation. Compared to HA-Tyr alone, the hybrid HA-Tyr/COL-II-Tyr hydrogel promoted enhanced chondrocyte adhesion, spreading, proliferation, and upregulation of cartilage-related gene expression. These results highlight the promising potential of injectable HA-Tyr/COL-II-Tyr hybrid hydrogels to deliver cells for cartilage regeneration.
Collapse
Affiliation(s)
- Parisa Torabi Rahvar
- Department of Chemical Engineering, Sharif University of Technology, Tehran, Iran; Tissue Engineering for Orthopaedics & Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, Bern, Switzerland
| | - Mohammad J Abdekhodaie
- Department of Chemical Engineering, Sharif University of Technology, Tehran, Iran; Environmental and Applied Science Management, Yeates School of Graduate Studies, Toronto Metropolitan University, Toronto, Canada.
| | - Elaheh Jooybar
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopaedics & Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, Bern, Switzerland; Inselspital, Bern University Hospital, Department of Orthopedic Surgery & Traumatology, Bern, Switzerland
| |
Collapse
|
5
|
Park K, Gao WW, Zheng J, Oh KT, Kim IY, You S. Hydrogel-Mediated Local Delivery of Induced Nephron Progenitor Cell-Sourced Molecules as a Cell-Free Approach for Acute Kidney Injury. Int J Mol Sci 2024; 25:10615. [PMID: 39408943 PMCID: PMC11477367 DOI: 10.3390/ijms251910615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
Acute kidney injury (AKI) constitutes a severe condition characterized by a sudden decrease in kidney function. Utilizing lineage-restricted stem/progenitor cells, directly reprogrammed from somatic cells, is a promising therapeutic option in personalized medicine for serious and incurable diseases such as AKI. The present study describes the therapeutic potential of induced nephron progenitor cell-sourced molecules (iNPC-SMs) as a cell-free strategy against cisplatin (CP)-induced nephrotoxicity, employing hyaluronic acid (HA) hydrogel-mediated local delivery to minimize systemic leakage and degradation. iNPC-SMs exhibited anti-apoptotic effects on HK-2 cells by inhibiting CP-induced ROS generation. Additionally, the localized biodistribution facilitated by hydrogel-mediated iNPC-SM delivery contributed to enhanced renal function, anti-inflammatory response, and renal regeneration in AKI mice. This study could serve as a 'proof of concept' for injectable hydrogel-mediated iNPC-SM delivery in AKI and as a model for further exploration of the development of cell-free regenerative medicine strategies.
Collapse
Affiliation(s)
- Kyoungmin Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea; (K.P.); (W.-W.G.); (J.Z.); (K.T.O.)
| | - Wei-Wei Gao
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea; (K.P.); (W.-W.G.); (J.Z.); (K.T.O.)
| | - Jie Zheng
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea; (K.P.); (W.-W.G.); (J.Z.); (K.T.O.)
| | - Kyung Taek Oh
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea; (K.P.); (W.-W.G.); (J.Z.); (K.T.O.)
| | - In-Yong Kim
- Catholic High-Performance Cell Therapy Center & Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Seungkwon You
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea; (K.P.); (W.-W.G.); (J.Z.); (K.T.O.)
- Institute of Animal Molecular Biotechnology, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
6
|
An H, Zhang M, Gu Z, Jiao X, Ma Y, Huang Z, Wen Y, Dong Y, Zhang P. Advances in Polysaccharides for Cartilage Tissue Engineering Repair: A Review. Biomacromolecules 2024; 25:2243-2260. [PMID: 38523444 DOI: 10.1021/acs.biomac.3c01424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
Cartilage repair has been a significant challenge in orthopedics that has not yet been fully resolved. Due to the absence of blood vessels and the almost cell-free nature of mature cartilage tissue, the limited ability to repair cartilage has resulted in significant socioeconomic pressures. Polysaccharide materials have recently been widely used for cartilage tissue repair due to their excellent cell loading, biocompatibility, and chemical modifiability. They also provide a suitable microenvironment for cartilage repair and regeneration. In this Review, we summarize the techniques used clinically for cartilage repair, focusing on polysaccharides, polysaccharides for cartilage repair, and the differences between these and other materials. In addition, we summarize the techniques of tissue engineering strategies for cartilage repair and provide an outlook on developing next-generation cartilage repair and regeneration materials from polysaccharides. This Review will provide theoretical guidance for developing polysaccharide-based cartilage repair and regeneration materials with clinical applications for cartilage tissue repair and regeneration.
Collapse
Affiliation(s)
- Heng An
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Meng Zhang
- Department of Orthopaedics and Trauma Peking University People's Hospital, Beijing 100044, China
| | - Zhen Gu
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Xiangyu Jiao
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Yinglei Ma
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Zhe Huang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Yongqiang Wen
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | | | - Peixun Zhang
- Department of Orthopaedics and Trauma Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
7
|
Cai R, Shan Y, Du F, Miao Z, Zhu L, Hang L, Xiao L, Wang Z. Injectable hydrogels as promising in situ therapeutic platform for cartilage tissue engineering. Int J Biol Macromol 2024; 261:129537. [PMID: 38278383 DOI: 10.1016/j.ijbiomac.2024.129537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/01/2024] [Accepted: 01/14/2024] [Indexed: 01/28/2024]
Abstract
Injectable hydrogels are gaining prominence as a biocompatible, minimally invasive, and adaptable platform for cartilage tissue engineering. Commencing with their synthesis, this review accentuates the tailored matrix formulations and cross-linking techniques essential for fostering three-dimensional cell culture and melding with complex tissue structures. Subsequently, it spotlights the hydrogels' enhanced properties, highlighting their augmented functionalities and broadened scope in cartilage tissue repair applications. Furthermore, future perspectives are advocated, urging continuous innovation and exploration to surmount existing challenges and harness the full clinical potential of hydrogels in regenerative medicine. Such advancements are crucial for validating the long-term efficacy and safety of hydrogels, positioning them as a promising direction in regenerative medicine to address cartilage-related ailments.
Collapse
Affiliation(s)
- Rong Cai
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Yisi Shan
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Fengyi Du
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, 212013, China
| | - Zhiwei Miao
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Like Zhu
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Li Hang
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Long Xiao
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China.
| | - Zhirong Wang
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China.
| |
Collapse
|
8
|
Shan BH, Wu FG. Hydrogel-Based Growth Factor Delivery Platforms: Strategies and Recent Advances. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2210707. [PMID: 37009859 DOI: 10.1002/adma.202210707] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/25/2023] [Indexed: 06/19/2023]
Abstract
Growth factors play a crucial role in regulating a broad variety of biological processes and are regarded as powerful therapeutic agents in tissue engineering and regenerative medicine in the past decades. However, their application is limited by their short half-lives and potential side effects in physiological environments. Hydrogels are identified as having the promising potential to prolong the half-lives of growth factors and mitigate their adverse effects by restricting them within the matrix to reduce their rapid proteolysis, burst release, and unwanted diffusion. This review discusses recent progress in the development of growth factor-containing hydrogels for various biomedical applications, including wound healing, brain tissue repair, cartilage and bone regeneration, and spinal cord injury repair. In addition, the review introduces strategies for optimizing growth factor release including affinity-based delivery, carrier-assisted delivery, stimuli-responsive delivery, spatial structure-based delivery, and cellular system-based delivery. Finally, the review presents current limitations and future research directions for growth factor-delivering hydrogels.
Collapse
Affiliation(s)
- Bai-Hui Shan
- State Key Laboratory of Digital Medical Engineering Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| | - Fu-Gen Wu
- State Key Laboratory of Digital Medical Engineering Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| |
Collapse
|
9
|
Grzelak A, Hnydka A, Higuchi J, Michalak A, Tarczynska M, Gaweda K, Klimek K. Recent Achievements in the Development of Biomaterials Improved with Platelet Concentrates for Soft and Hard Tissue Engineering Applications. Int J Mol Sci 2024; 25:1525. [PMID: 38338805 PMCID: PMC10855389 DOI: 10.3390/ijms25031525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Platelet concentrates such as platelet-rich plasma, platelet-rich fibrin or concentrated growth factors are cost-effective autologous preparations containing various growth factors, including platelet-derived growth factor, transforming growth factor β, insulin-like growth factor 1 and vascular endothelial growth factor. For this reason, they are often used in regenerative medicine to treat wounds, nerve damage as well as cartilage and bone defects. Unfortunately, after administration, these preparations release growth factors very quickly, which lose their activity rapidly. As a consequence, this results in the need to repeat the therapy, which is associated with additional pain and discomfort for the patient. Recent research shows that combining platelet concentrates with biomaterials overcomes this problem because growth factors are released in a more sustainable manner. Moreover, this concept fits into the latest trends in tissue engineering, which include biomaterials, bioactive factors and cells. Therefore, this review presents the latest literature reports on the properties of biomaterials enriched with platelet concentrates for applications in skin, nerve, cartilage and bone tissue engineering.
Collapse
Affiliation(s)
- Agnieszka Grzelak
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki Street 1, 20-093 Lublin, Poland; (A.G.); (A.H.)
| | - Aleksandra Hnydka
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki Street 1, 20-093 Lublin, Poland; (A.G.); (A.H.)
| | - Julia Higuchi
- Laboratory of Nanostructures, Institute of High Pressure Physics, Polish Academy of Sciences, Prymasa Tysiaclecia Avenue 98, 01-142 Warsaw, Poland;
| | - Agnieszka Michalak
- Independent Laboratory of Behavioral Studies, Medical University of Lublin, Chodzki 4 a Street, 20-093 Lublin, Poland;
| | - Marta Tarczynska
- Department and Clinic of Orthopaedics and Traumatology, Medical University of Lublin, Jaczewskiego 8 Street, 20-090 Lublin, Poland; (M.T.); (K.G.)
- Arthros Medical Centre, Chodzki 31 Street, 20-093 Lublin, Poland
| | - Krzysztof Gaweda
- Department and Clinic of Orthopaedics and Traumatology, Medical University of Lublin, Jaczewskiego 8 Street, 20-090 Lublin, Poland; (M.T.); (K.G.)
- Arthros Medical Centre, Chodzki 31 Street, 20-093 Lublin, Poland
| | - Katarzyna Klimek
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki Street 1, 20-093 Lublin, Poland; (A.G.); (A.H.)
| |
Collapse
|
10
|
Wu H, Zhang X, Wang Z, Chen X, Li Y, Fang J, Zheng S, Zhang L, Li C, Hao L. Preparation, properties and in vitro osteogensis of self-reinforcing injectable hydrogel. Eur J Pharm Sci 2024; 192:106617. [PMID: 37865283 DOI: 10.1016/j.ejps.2023.106617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/22/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
As an attractive biomaterial for bone reconstruction, injectable biomaterials have many prominent characteristics such as good biocompatibility and bone-filling ability. However, there are weak as load-bearing scaffolds. In this study, polyvinyl alcohol (PVA) and bioactive glass (BAG) were interpenetrated into sodium alginate (SA) network to obtain self-enhanced injectable hydrogel. The optimum ratio of PVA/SA/BAG hydrogel was determined based on injectability, gelation time and chemical characterization. Results showed that the selected ratio had the shortest gelation time of 3.5min, and the hydrogel had a rough surface and good coagulation property. The hydrogel was capable of carrying 1kg of weight by mineralization for 14 d The compressive strength, compressive modulus, and fracture energy of the hydrogel reached 0.12MPa, 0.376MPa and 17.750kJ m-2, respectively. Meanwhile, the hydrogel had high moisture content and dissolution rate, and it was sensitive to temperature and ionic strength. Hydroxyapatite was generated on the hydrogel surface, and the hydrogel pores increased, and the pore size enlarged. The biocompatibility of PVA/SA/BAG hydrogel was analyzed using hemolysis and cytotoxicity assays. Results revealed its good biocompatibility with low hemolysis rate and no cytotoxicity to MC3T3-E1 cells. The hydrogel was also found to promote the differentiation of MC3T3-E1 cells with significantly increased in ALP activity and expression of relevant differentiation factors. In vitro mineralization assay showed an increase in calcium nodules and calcification area, indicating the ability of hydrogel to promote mineralization MC3T3-E1 cells. These findings indicated that PVA/SA/BAG hydrogel had potential uses in the field of irregular bone-defect repair due to its injectability, cytocompatibility, and tailorable functionality.
Collapse
Affiliation(s)
- Hongyan Wu
- College of Animal Science, Jilin University, Changchun, Jilin, China
| | - Xunming Zhang
- College of Animal Science, Jilin University, Changchun, Jilin, China
| | - Zhaoguo Wang
- College of Animal Science, Jilin University, Changchun, Jilin, China
| | - Xi Chen
- College of Animal Science, Jilin University, Changchun, Jilin, China
| | - Yi Li
- College of Animal Science, Jilin University, Changchun, Jilin, China
| | - Jiayuan Fang
- College of Animal Science, Jilin University, Changchun, Jilin, China
| | - Shuo Zheng
- College of Animal Science, Jilin University, Changchun, Jilin, China
| | - Libo Zhang
- College of Animal Science, Jilin University, Changchun, Jilin, China
| | - Changhong Li
- College of Life Sciences, Baicheng Normal University, Baicheng, Jilin, China.
| | - Linlin Hao
- College of Animal Science, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
11
|
Xie R, Pal V, Yu Y, Lu X, Gao M, Liang S, Huang M, Peng W, Ozbolat IT. A comprehensive review on 3D tissue models: Biofabrication technologies and preclinical applications. Biomaterials 2024; 304:122408. [PMID: 38041911 PMCID: PMC10843844 DOI: 10.1016/j.biomaterials.2023.122408] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/09/2023] [Accepted: 11/22/2023] [Indexed: 12/04/2023]
Abstract
The limitations of traditional two-dimensional (2D) cultures and animal testing, when it comes to precisely foreseeing the toxicity and clinical effectiveness of potential drug candidates, have resulted in a notable increase in the rate of failure during the process of drug discovery and development. Three-dimensional (3D) in-vitro models have arisen as substitute platforms with the capacity to accurately depict in-vivo conditions and increasing the predictivity of clinical effects and toxicity of drug candidates. It has been found that 3D models can accurately represent complex tissue structure of human body and can be used for a wide range of disease modeling purposes. Recently, substantial progress in biomedicine, materials and engineering have been made to fabricate various 3D in-vitro models, which have been exhibited better disease progression predictivity and drug effects than convention models, suggesting a promising direction in pharmaceutics. This comprehensive review highlights the recent developments in 3D in-vitro tissue models for preclinical applications including drug screening and disease modeling targeting multiple organs and tissues, like liver, bone, gastrointestinal tract, kidney, heart, brain, and cartilage. We discuss current strategies for fabricating 3D models for specific organs with their strengths and pitfalls. We expand future considerations for establishing a physiologically-relevant microenvironment for growing 3D models and also provide readers with a perspective on intellectual property, industry, and regulatory landscape.
Collapse
Affiliation(s)
- Renjian Xie
- Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering in Jiangxi Province, Gannan Medical University, Ganzhou, JX, 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, JX, China
| | - Vaibhav Pal
- Department of Chemistry, Pennsylvania State University, University Park, PA, USA; The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - Yanrong Yu
- School of Pharmaceutics, Nanchang University, Nanchang, JX, 330006, China
| | - Xiaolu Lu
- Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering in Jiangxi Province, Gannan Medical University, Ganzhou, JX, 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, JX, China
| | - Mengwei Gao
- School of Pharmaceutics, Nanchang University, Nanchang, JX, 330006, China
| | - Shijie Liang
- School of Pharmaceutics, Nanchang University, Nanchang, JX, 330006, China
| | - Miao Huang
- Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering in Jiangxi Province, Gannan Medical University, Ganzhou, JX, 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, JX, China
| | - Weijie Peng
- Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering in Jiangxi Province, Gannan Medical University, Ganzhou, JX, 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, JX, China; School of Pharmaceutics, Nanchang University, Nanchang, JX, 330006, China.
| | - Ibrahim T Ozbolat
- The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA; Engineering Science and Mechanics Department, Penn State University, University Park, PA, USA; Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA; Materials Research Institute, Pennsylvania State University, University Park, PA, USA; Department of Neurosurgery, Pennsylvania State College of Medicine, Hershey, PA, USA; Penn State Cancer Institute, Penn State University, Hershey, PA, 17033, USA; Department of Medical Oncology, Cukurova University, Adana, 01130, Turkey; Biotechnology Research and Application Center, Cukurova University, Adana, 01130, Turkey.
| |
Collapse
|
12
|
Atwal A, Dale TP, Snow M, Forsyth NR, Davoodi P. Injectable hydrogels: An emerging therapeutic strategy for cartilage regeneration. Adv Colloid Interface Sci 2023; 321:103030. [PMID: 37907031 DOI: 10.1016/j.cis.2023.103030] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 11/02/2023]
Abstract
The impairment of articular cartilage due to traumatic incidents or osteoarthritis has posed significant challenges for healthcare practitioners, researchers, and individuals suffering from these conditions. Due to the absence of an approved treatment strategy for the complete restoration of cartilage defects to their native state, the tissue condition often deteriorates over time, leading to osteoarthritic (OA). However, recent advancements in the field of regenerative medicine have unveiled promising prospects through the utilization of injectable hydrogels. This versatile class of biomaterials, characterized by their ability to emulate the characteristics of native articular cartilage, offers the distinct advantage of minimally invasive administration directly to the site of damage. These hydrogels can also serve as ideal delivery vehicles for a diverse range of bioactive agents, including growth factors, anti-inflammatory drugs, steroids, and cells. The controlled release of such biologically active molecules from hydrogel scaffolds can accelerate cartilage healing, stimulate chondrogenesis, and modulate the inflammatory microenvironment to halt osteoarthritic progression. The present review aims to describe the methods used to design injectable hydrogels, expound upon their applications as delivery vehicles of biologically active molecules, and provide an update on recent advances in leveraging these delivery systems to foster articular cartilage regeneration.
Collapse
Affiliation(s)
- Arjan Atwal
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom
| | - Tina P Dale
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom
| | - Martyn Snow
- Department of Arthroscopy, Royal Orthopaedic Hospital NHS Foundation Trust, Birmingham B31 2AP, United Kingdom; The Robert Jones and Agnes Hunt Hospital, Oswestry, Shropshire SY10 7AG, United Kingdom
| | - Nicholas R Forsyth
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom; Vice Principals' Office, University of Aberdeen, Kings College, Aberdeen AB24 3FX, United Kingdom
| | - Pooya Davoodi
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom.
| |
Collapse
|
13
|
Deng QS, Gao Y, Rui BY, Li XR, Liu PL, Han ZY, Wei ZY, Zhang CR, Wang F, Dawes H, Zhu TH, Tao SC, Guo SC. Double-network hydrogel enhanced by SS31-loaded mesoporous polydopamine nanoparticles: Symphonic collaboration of near-infrared photothermal antibacterial effect and mitochondrial maintenance for full-thickness wound healing in diabetes mellitus. Bioact Mater 2023; 27:409-428. [PMID: 37152712 PMCID: PMC10160601 DOI: 10.1016/j.bioactmat.2023.04.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/24/2023] [Accepted: 04/02/2023] [Indexed: 05/09/2023] Open
Abstract
Diabetic wound healing has become a serious healthcare challenge. The high-glucose environment leads to persistent bacterial infection and mitochondrial dysfunction, resulting in chronic inflammation, abnormal vascular function, and tissue necrosis. To solve these issues, we developed a double-network hydrogel, constructed with pluronic F127 diacrylate (F127DA) and hyaluronic acid methacrylate (HAMA), and enhanced by SS31-loaded mesoporous polydopamine nanoparticles (MPDA NPs). As components, SS31, a mitochondria-targeted peptide, maintains mitochondrial function, reduces mitochondrial reactive oxygen species (ROS) and thus regulates macrophage polarization, as well as promoting cell proliferation and migration, while MPDA NPs not only scavenge ROS and exert an anti-bacterial effect by photothermal treatment under near-infrared light irradiation, but also control release of SS31 in response to ROS. This F127DA/HAMA-MPDA@SS31 (FH-M@S) hydrogel has characteristics of adhesion, superior biocompatibility and mechanical properties which can adapt to irregular wounds at different body sites and provide sustained release of MPDA@SS31 (M@S) NPs. In addition, in a diabetic rat full thickness skin defect model, the FH-M@S hydrogel promoted macrophage M2 polarization, collagen deposition, neovascularization and wound healing. Therefore, the FH-M@S hydrogel exhibits promising therapeutic potential for skin regeneration.
Collapse
Affiliation(s)
- Qing-Song Deng
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Yuan Gao
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Bi-Yu Rui
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China
| | - Xu-Ran Li
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Po-Lin Liu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Zi-Yin Han
- Department of Rheumatology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, No.29, Xinglongxiang, Tianning District, Changzhou, 213000, China
| | - Zhan-Ying Wei
- Shanghai Clinical Research Centre of Bone Diseases, Department of Osteoporosis and Bone Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Chang-Ru Zhang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
| | - Fei Wang
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China
| | - Helen Dawes
- Faculty of Health and Life Science, Oxford Brookes University, Headington Road, Oxford, OX3 0BP, UK
- NIHR Oxford Health Biomedical Research Centre, Oxford, OX3 7JX, UK
- College of Medicine and Health, St Lukes Campus, University of Exeter, Heavitree Road, Exeter, EX1 2LU, UK
| | - Tong-He Zhu
- School of Chemistry and Chemical Engineering, Shanghai Engineering Research Center of Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Non-Coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, China
| | - Shi-Cong Tao
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China
- Corresponding author. Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
| | - Shang-Chun Guo
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- Corresponding author. Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
14
|
Goldshmid R, Simaan-Yameen H, Ifergan L, Loebel C, Burdick JA, Seliktar D. Modulus-dependent effects on neurogenic, myogenic, and chondrogenic differentiation of human mesenchymal stem cells in three-dimensional hydrogel cultures. J Biomed Mater Res A 2023; 111:1441-1458. [PMID: 37066837 DOI: 10.1002/jbm.a.37545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 03/23/2023] [Accepted: 03/25/2023] [Indexed: 04/18/2023]
Abstract
Human mesenchymal stromal cells (hMSCs) are of significant interest as a renewable source of therapeutically useful cells. In tissue engineering, hMSCs are implanted within a scaffold to provide enhanced capacity for tissue repair. The present study evaluates how mechanical properties of that scaffold can alter the phenotype and genotype of the cells, with the aim of augmenting hMSC differentiation along the myogenic, neurogenic or chondrogenic linages. The hMSCs were grown three-dimensionally (3D) in a hydrogel comprised of poly(ethylene glycol) (PEG)-conjugated to fibrinogen. The hydrogel's shear storage modulus (G'), which was controlled by increasing the amount of PEG-diacrylate cross-linker in the matrix, was varied in the range of 100-2000 Pascal (Pa). The differentiation into each lineage was initiated by a defined culture medium, and the hMSCs grown in the different modulus hydrogels were characterized using gene and protein expression. Materials having lower storage moduli (G' = 100 Pa) exhibited more hMSCs differentiating to neurogenic lineages. Myogenesis was favored in materials having intermediate modulus values (G' = 500 Pa), whereas chondrogenesis was favored in materials with a higher modulus (G' = 1000 Pa). Enhancing the differentiation pathway of hMSCs in 3D hydrogel scaffolds using simple modifications to mechanical properties represents an important achievement toward the effective application of these cells in tissue engineering.
Collapse
Affiliation(s)
- Revital Goldshmid
- The Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
- The Interdisciplinary Program for Biotechnology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Haneen Simaan-Yameen
- The Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
- The Interdisciplinary Program for Biotechnology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Liaura Ifergan
- The Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Claudia Loebel
- Materials Science & Engineering Department, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason A Burdick
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado, USA
| | - Dror Seliktar
- The Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
15
|
Toropitsyn E, Ščigalková I, Pravda M, Toropitsyna J, Velebný V. Enzymatically cross-linked hyaluronic acid hydrogels as in situ forming carriers of platelet-rich plasma: Mechanical properties and bioactivity levels evaluation. J Mech Behav Biomed Mater 2023; 143:105916. [PMID: 37224645 DOI: 10.1016/j.jmbbm.2023.105916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/14/2023] [Accepted: 05/15/2023] [Indexed: 05/26/2023]
Abstract
New studies have shown the great potential of the combination of in situ enzymatically cross-linked hydrogels based on tyramine derivative of hyaluronic acid (HA-TA) with platelet-rich plasma (PRP) and platelet lysate in regenerative medicine. This study describes how the presence of PRP and platelet lysate affects the kinetics of gelation, viscoelastic properties, swelling ratio, and the network structure of HA-TA hydrogels and how the encapsulation of PRP in hydrogels affects the bioactivity of released PRP determined as the ability to induce cell proliferation. The properties of hydrogels were tuned by a degree of substitution and concentration of HA-TA derivatives. The addition of platelet derivatives to the reaction mixture slowed down the cross-linking reaction and reduced elastic modulus (G') and thus cross-linking efficiency. However, low-swellable hydrogels (7-190%) suitable for soft tissue engineering with G' 200-1800 Pa were prepared with a gelation time within 1 min. It was confirmed that tested cross-linking reaction conditions are suitable for PRP incorporation because the total bioactivity level of PRP released from HA-TA hydrogels was ≥87% and HA-TA content in the hydrogels and thus mesh size (285-482 nm) has no significant effect on the bioactivity level of released PRP.
Collapse
Affiliation(s)
- Evgeniy Toropitsyn
- Contipro a.s., Dolní Dobrouč 401, 56102, Dolní Dobrouč, Czech Republic; Biocev, First Faculty of Medicine Charles University, Průmyslová 595, 25250, Vestec, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, Prague, 120 00, Czech Republic.
| | - Ivana Ščigalková
- Contipro a.s., Dolní Dobrouč 401, 56102, Dolní Dobrouč, Czech Republic
| | - Martin Pravda
- Contipro a.s., Dolní Dobrouč 401, 56102, Dolní Dobrouč, Czech Republic
| | - Jelena Toropitsyna
- Department of Power Engineering, University of Chemistry and Technology, Prague, Technická 5, Prague, 166 28, Czech Republic
| | - Vladimír Velebný
- Contipro a.s., Dolní Dobrouč 401, 56102, Dolní Dobrouč, Czech Republic
| |
Collapse
|
16
|
Tian B, Liu J, Guo S, Li A, Wan JB. Macromolecule-based hydrogels nanoarchitectonics with mesenchymal stem cells for regenerative medicine: A review. Int J Biol Macromol 2023:125161. [PMID: 37270118 DOI: 10.1016/j.ijbiomac.2023.125161] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/25/2023] [Accepted: 05/28/2023] [Indexed: 06/05/2023]
Abstract
The role of regenerative medicine in clinical therapies is becoming increasingly vital. Under specific conditions, mesenchymal stem cells (MSCs) are capable of differentiating into mesoblastema (i.e., adipocytes, chondrocytes, and osteocytes) and other embryonic lineages. Their application in regenerative medicine has attracted a great deal of interest among researchers. To maximize the potential applications of MSCs, materials science could provide natural extracellular matrices and provide an effective means to understand the various mechanisms of differentiation for the growth of MSCs. Pharmaceutical fields are represented among the research on biomaterials by macromolecule-based hydrogel nanoarchitectonics. Various biomaterials have been used to prepare hydrogels with their unique chemical and physical properties to provide a controlled microenvironment for the culture of MSCs, laying the groundwork for future practical applications in regenerative medicine. This article currently describes and summarizes the sources, characteristics, and clinical trials of MSCs. In addition, it describes the differentiation of MSCs in various macromolecule-based hydrogel nanoarchitectonics and highlights the preclinical studies of MSCs-loaded hydrogel materials in regenerative medicine conducted over the past few years. Finally, the challenges and prospects of MSC-loaded hydrogels are discussed, and the future development of macromolecule-based hydrogel nanoarchitectonics is outlined by comparing the current literature.
Collapse
Affiliation(s)
- Bingren Tian
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China.
| | - Jiayue Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao
| | - Songlin Guo
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Aiqin Li
- Department of Day-care Unit, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Jian-Bo Wan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao.
| |
Collapse
|
17
|
Enhanced Drug Delivery System Using Mesenchymal Stem Cells and Membrane-Coated Nanoparticles. Molecules 2023; 28:molecules28052130. [PMID: 36903399 PMCID: PMC10004171 DOI: 10.3390/molecules28052130] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/14/2023] [Accepted: 02/18/2023] [Indexed: 03/02/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have newly developed as a potential drug delivery system. MSC-based drug delivery systems (MSCs-DDS) have made significant strides in the treatment of several illnesses, as shown by a plethora of research. However, as this area of research rapidly develops, several issues with this delivery technique have emerged, most often as a result of its intrinsic limits. To increase the effectiveness and security of this system, several cutting-edge technologies are being developed concurrently. However, the advancement of MSC applicability in clinical practice is severely hampered by the absence of standardized methodologies for assessing cell safety, effectiveness, and biodistribution. In this work, the biodistribution and systemic safety of MSCs are highlighted as we assess the status of MSC-based cell therapy at this time. We also examine the underlying mechanisms of MSCs to better understand the risks of tumor initiation and propagation. Methods for MSC biodistribution are explored, as well as the pharmacokinetics and pharmacodynamics of cell therapies. We also highlight various promising technologies, such as nanotechnology, genome engineering technology, and biomimetic technology, to enhance MSC-DDS. For statistical analysis, we used analysis of variance (ANOVA), Kaplan Meier, and log-rank tests. In this work, we created a shared DDS medication distribution network using an extended enhanced optimization approach called enhanced particle swarm optimization (E-PSO). To identify the considerable untapped potential and highlight promising future research paths, we highlight the use of MSCs in gene delivery and medication, also membrane-coated MSC nanoparticles, for treatment and drug delivery.
Collapse
|
18
|
Canceill T, Jourdan G, Kémoun P, Guissard C, Monsef YA, Bourdens M, Chaput B, Cavalie S, Casteilla L, Planat-Bénard V, Monsarrat P, Raymond-Letron I. Characterization and Safety Profile of a New Combined Advanced Therapeutic Medical Product Platelet Lysate-Based Fibrin Hydrogel for Mesenchymal Stromal Cell Local Delivery in Regenerative Medicine. Int J Mol Sci 2023; 24:ijms24032206. [PMID: 36768532 PMCID: PMC9916739 DOI: 10.3390/ijms24032206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/14/2023] [Accepted: 01/16/2023] [Indexed: 01/25/2023] Open
Abstract
Adipose-derived mesenchymal stromal cells (ASC) transplant to recover the optimal tissue structure/function relationship is a promising strategy to regenerate tissue lesions. Because filling local tissue defects by injection alone is often challenging, designing adequate cell carriers with suitable characteristics is critical for in situ ASC delivery. The aim of this study was to optimize the generation phase of a platelet-lysate-based fibrin hydrogel (PLFH) as a proper carrier for in situ ASC implantation and (1) to investigate in vitro PLFH biomechanical properties, cell viability, proliferation and migration sustainability, and (2) to comprehensively assess the local in vivo PLFH/ASC safety profile (local tolerance, ASC fate, biodistribution and toxicity). We first defined the experimental conditions to enhance physicochemical properties and microscopic features of PLFH as an adequate ASC vehicle. When ASC were mixed with PLFH, in vitro assays exhibited hydrogel supporting cell migration, viability and proliferation. In vivo local subcutaneous and subgingival PLFH/ASC administration in nude mice allowed us to generate biosafety data, including biodegradability, tolerance, ASC fate and engraftment, and the absence of biodistribution and toxicity to non-target tissues. Our data strongly suggest that this novel combined ATMP for in situ administration is safe with an efficient local ASC engraftment, supporting the further development for human clinical cell therapy.
Collapse
Affiliation(s)
- Thibault Canceill
- CIRIMAT, Université Toulouse III Paul Sabatier, CNRS UMR 5085, INPT, Faculté de Pharmacie, 35 Chemin des Maraichers, CEDEX 09, 31062 Toulouse, France
- Department of Oral Medicine and Toulouse University Hospital (CHU of Toulouse)—Toulouse Institute of Oral Medicine and Science, CEDEX 09, 31062 Toulouse, France
| | - Géraldine Jourdan
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Batiment INCERE, 4bis Avenue Hubert Curien, 31100 Toulouse, France
| | - Philippe Kémoun
- Department of Oral Medicine and Toulouse University Hospital (CHU of Toulouse)—Toulouse Institute of Oral Medicine and Science, CEDEX 09, 31062 Toulouse, France
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Batiment INCERE, 4bis Avenue Hubert Curien, 31100 Toulouse, France
| | - Christophe Guissard
- Department of Oral Medicine and Toulouse University Hospital (CHU of Toulouse)—Toulouse Institute of Oral Medicine and Science, CEDEX 09, 31062 Toulouse, France
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Batiment INCERE, 4bis Avenue Hubert Curien, 31100 Toulouse, France
| | - Yanad Abou Monsef
- LabHPEC, Histology and Pathology Department, Université de Toulouse, ENVT, CEDEX 03, 31076 Toulouse, France
| | - Marion Bourdens
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Batiment INCERE, 4bis Avenue Hubert Curien, 31100 Toulouse, France
| | - Benoit Chaput
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Batiment INCERE, 4bis Avenue Hubert Curien, 31100 Toulouse, France
- Service de Chirurgie Plastique, Reconstructrice et Esthétique, Centre Hospitalier Universitaire Rangueil, Avenue du Professeur Jean Poulhès, CEDEX 09, 31059 Toulouse, France
| | - Sandrine Cavalie
- CIRIMAT, Université Toulouse III Paul Sabatier, CNRS UMR 5085, INPT, Faculté de Pharmacie, 35 Chemin des Maraichers, CEDEX 09, 31062 Toulouse, France
| | - Louis Casteilla
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Batiment INCERE, 4bis Avenue Hubert Curien, 31100 Toulouse, France
| | - Valérie Planat-Bénard
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Batiment INCERE, 4bis Avenue Hubert Curien, 31100 Toulouse, France
| | - Paul Monsarrat
- Department of Oral Medicine and Toulouse University Hospital (CHU of Toulouse)—Toulouse Institute of Oral Medicine and Science, CEDEX 09, 31062 Toulouse, France
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Batiment INCERE, 4bis Avenue Hubert Curien, 31100 Toulouse, France
- Artificial and Natural Intelligence Toulouse Institute ANITI, 31400 Toulouse, France
- Correspondence:
| | - Isabelle Raymond-Letron
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Batiment INCERE, 4bis Avenue Hubert Curien, 31100 Toulouse, France
- LabHPEC, Histology and Pathology Department, Université de Toulouse, ENVT, CEDEX 03, 31076 Toulouse, France
| |
Collapse
|
19
|
Strecanska M, Danisovic L, Ziaran S, Cehakova M. The Role of Extracellular Matrix and Hydrogels in Mesenchymal Stem Cell Chondrogenesis and Cartilage Regeneration. LIFE (BASEL, SWITZERLAND) 2022; 12:life12122066. [PMID: 36556431 PMCID: PMC9784885 DOI: 10.3390/life12122066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022]
Abstract
Diseases associated with articular cartilage disintegration or loss are still therapeutically challenging. The traditional treatment approaches only alleviate the symptoms while potentially causing serious side effects. The limited self-renewal potential of articular cartilage provides opportunities for advanced therapies involving mesenchymal stem cells (MSCs) that are characterized by a remarkable regenerative capacity. The chondrogenic potential of MSCs is known to be regulated by the local environment, including soluble factors and the less discussed extracellular matrix (ECM) components. This review summarizes the process of chondrogenesis, and also the biological properties of the ECM mediated by mechanotransduction as well as canonical and non-canonical signaling. Our focus is also on the influence of the ECM's physical parameters, molecular composition, and chondrogenic factor affinity on the adhesion, survival, and chondrogenic differentiation of MSCs. These basic biological insights are crucial for a more precise fabrication of ECM-mimicking hydrogels to improve cartilage tissue reconstruction. Lastly, we provide an overview of hydrogel classification and characterization. We also include the results from preclinical models combining MSCs with hydrogels for the treatment of cartilage defects, to support clinical application of this construct. Overall, it is believed that the proper combination of MSCs, hydrogels, and chondrogenic factors can lead to complex cartilage regeneration.
Collapse
Affiliation(s)
- Magdalena Strecanska
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia
- Institute of Medical Biology, Genetics, and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Lubos Danisovic
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia
- Institute of Medical Biology, Genetics, and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Stanislav Ziaran
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia
- Department of Urology, Faculty of Medicine, Comenius University, Limbova 5, 833 05 Bratislava, Slovakia
| | - Michaela Cehakova
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia
- Institute of Medical Biology, Genetics, and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
- Correspondence: ; Tel.: +421-2-5935-7215
| |
Collapse
|
20
|
Toropitsyn E, Pravda M, Rebenda D, Ščigalková I, Vrbka M, Velebný V. A composite device for viscosupplementation treatment resistant to degradation by reactive oxygen species and hyaluronidase. J Biomed Mater Res B Appl Biomater 2022; 110:2595-2611. [PMID: 35727166 DOI: 10.1002/jbm.b.35114] [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: 07/29/2021] [Revised: 05/02/2022] [Accepted: 06/08/2022] [Indexed: 12/15/2022]
Abstract
Osteoarthritis (OA) is one of the most common musculoskeletal disorders in the world. OA is often associated with the loss of viscoelastic and tribological properties of synovial fluid (SF) due to degradation of hyaluronic acid (HA) by reactive oxygen species (ROS) and hyaluronidases. Viscosupplementation is one of the ways how to effectively restore SF functions. However, current viscosupplementation products provide only temporal therapeutic effect because of short biological half-life. In this article we describe a novel device for viscosupplementation (NV) based on the cross-linked tyramine derivative of HA, chondroitin sulfate (CS), and high molecular weight HA by online determination of viscoelastic properties loss during degradation by ROS and hyaluronidase. Rheological and tribological properties of developed viscosupplement were compared with HA solutions with different molecular weights in the range 500-2000 kDa, which are currently commonly used as medical devices for viscosupplementation treatment. Moreover, based on clinical practice and scientific literature all samples were also diluted by model OA SF in the ratio 1:1 (vol/vol) to better predict final properties after injection to the joint. The observed results confirmed that NV exhibits appropriate rheological properties (viscosity, elastic, and viscous moduli) comparable with healthy SF and maintain them during degradation for a significantly longer time than HA solutions with molecular weight in the range 500-2000 kDa and cross-linked material without CS.
Collapse
Affiliation(s)
- Evgeniy Toropitsyn
- Contipro a.s., Dolní Dobrouč, Czech Republic.,Biocev, First Faculty of Medicine Charles University, Vestec, Czech Republic
| | | | - David Rebenda
- Faculty of Mechanical Engineering, Brno University of Technology, Brno, Czech Republic
| | | | - Martin Vrbka
- Faculty of Mechanical Engineering, Brno University of Technology, Brno, Czech Republic
| | | |
Collapse
|
21
|
Wang M, Deng Z, Guo Y, Xu P. Designing functional hyaluronic acid-based hydrogels for cartilage tissue engineering. Mater Today Bio 2022; 17:100495. [PMID: 36420054 PMCID: PMC9676212 DOI: 10.1016/j.mtbio.2022.100495] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 11/14/2022] Open
Abstract
Damage to cartilage tissues is often difficult to repair owing to chronic inflammation and a lack of bioactive factors. Therefore, developing bioactive materials, such as hydrogels acting as extracellular matrix mimics, that can inhibit the inflammatory microenvironment and promote cartilage repair is crucial. Hyaluronic acid, which exists in cartilage and synovial fluid, has been extensively investigated for cartilage tissue engineering because of its promotion of cell adhesion and proliferation, regulation of inflammation, and enhancement of cartilage regeneration. However, hyaluronic acid-based hydrogels have poor degradation rates and unfavorable mechanical properties, limiting their application in cartilage tissue engineering. Recently, various multifunctional hyaluronic acid-based hydrogels, including alkenyl, aldehyde, thiolated, phenolized, hydrazide, and host–guest group-modified hydrogels, have been extensively studied for use in cartilage tissue engineering. In this review, we summarize the recent progress in the multifunctional design of hyaluronic acid-based hydrogels and their application in cartilage tissue engineering. Moreover, we outline the future research prospects and directions in cartilage tissue regeneration. This would provide theoretical guidance for developing hyaluronic acid-based hydrogels with specific properties to satisfy the requirements of cartilage tissue repair.
Collapse
|
22
|
Markazi R, Soltani-Zangbar MS, Zamani M, Eghbal-Fard S, Motavalli R, Kamrani A, Dolati S, Ahmadi M, Aghebati-Maleki L, Mehdizadeh A, Eslamian F, Pishgahi A, Yousefi M. Platelet lysate and tendon healing: comparative analysis of autologous frozen-thawed PRP and ketorolac tromethamine in the treatment of patients with rotator cuff tendinopathy. Growth Factors 2022; 40:163-174. [PMID: 36026559 DOI: 10.1080/08977194.2022.2093198] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Platelet-rich blood derivatives are being nowadays increasingly used in the treatment of tendon-related pathologies as a rich source of growth factors. We sought to ascertain if local application of platelet lysate (PL) to augment rotator cuff repair ameliorates patient outcomes compared to ketorolac tromethamine treated group. A total of forty patients, with clinical diagnosis of Rotator Cuff Tendinopathy were randomized to receive sub acromial injections of PL every week for a total of 3 injections and two injection of ketorolac tromethamine once every two weeks. Subjective assessments included VAS, SPADI and shoulder range of motion were assessed at baseline and at 1 and 6 months after injection. Taking both control and PL groups, it was vividly seen that the outcomes were identical at the initial state, as well as the short-term one; whereas, when considering the 6-month period, there is a seemingly remarkable superiority in PL group in all parameters.
Collapse
Affiliation(s)
- Raha Markazi
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Sadegh Soltani-Zangbar
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Zamani
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Shadi Eghbal-Fard
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roza Motavalli
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Kamrani
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanam Dolati
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fariba Eslamian
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Pishgahi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
23
|
Vieira S, Silva-Correia J, Reis RL, Oliveira JM. Engineering Hydrogels for Modulation of Material-Cell Interactions. Macromol Biosci 2022; 22:e2200091. [PMID: 35853666 DOI: 10.1002/mabi.202200091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/29/2022] [Indexed: 11/06/2022]
Abstract
Hydrogels are a recurrent platform for Tissue Engineering (TE) strategies. Their versatility and the variety of available methods for tuning their properties highly contribute to hydrogels' success. As a result, the design of advanced hydrogels has been thoroughly studied, in the quest for better solutions not only for drugs- and cell-based therapies but also for more fundamental studies. The wide variety of sources, crosslinking strategies, and functionalization methods, and mostly the resemblance of hydrogels to the natural extracellular matrix, make this 3D hydrated structures an excellent tool for TE approaches. The state-of-the-art information regarding hydrogel design, processing methods, and the influence of different hydrogel formulations on the final cell-biomaterial interactions are overviewed herein. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Sílvia Vieira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joana Silva-Correia
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - J Miguel Oliveira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
24
|
Lehoczky G, Trofin RE, Vallmajo-Martin Q, Chawla S, Pelttari K, Mumme M, Haug M, Egloff C, Jakob M, Ehrbar M, Martin I, Barbero A. In Vitro and Ectopic In Vivo Studies toward the Utilization of Rapidly Isolated Human Nasal Chondrocytes for Single-Stage Arthroscopic Cartilage Regeneration Therapy. Int J Mol Sci 2022; 23:ijms23136900. [PMID: 35805907 PMCID: PMC9267018 DOI: 10.3390/ijms23136900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/15/2022] [Accepted: 06/19/2022] [Indexed: 02/05/2023] Open
Abstract
Nasal chondrocytes (NCs) have a higher and more reproducible chondrogenic capacity than articular chondrocytes, and the engineered cartilage tissue they generate in vitro has been demonstrated to be safe in clinical applications. Here, we aimed at determining the feasibility for a single-stage application of NCs for cartilage regeneration under minimally invasive settings. In particular, we assessed whether NCs isolated using a short collagenase digestion protocol retain their potential to proliferate and chondro-differentiate within an injectable, swiftly cross-linked and matrix-metalloproteinase (MMP)-degradable polyethylene glycol (PEG) gel enriched with human platelet lysate (hPL). NC-hPL-PEG gels were additionally tested for their capacity to generate cartilage tissue in vivo and to integrate into cartilage/bone compartments of human osteochondral plugs upon ectopic subcutaneous implantation into nude mice. NCs isolated with a rapid protocol and embedded in PEG gels with hPL at low cell density were capable of efficiently proliferating and of generating tissue rich in glycosaminoglycans and collagen II. NC-hPL-PEG gels developed into hyaline-like cartilage tissues upon ectopic in vivo implantation and integrated with surrounding native cartilage and bone tissues. The delivery of NCs in PEG gels containing hPL is a feasible strategy for cartilage repair and now requires further validation in orthotopic in vivo models.
Collapse
Affiliation(s)
- Gyözö Lehoczky
- Department of Orthopaedic Surgery and Traumatology, University Hospital of Basel, 4031 Basel, Switzerland; (G.L.); (M.M.); (C.E.)
- Department of Biomedicine, Tissue Engineering Laboratory, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (R.E.T.); (S.C.); (K.P.); (A.B.)
| | - Raluca Elena Trofin
- Department of Biomedicine, Tissue Engineering Laboratory, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (R.E.T.); (S.C.); (K.P.); (A.B.)
| | - Queralt Vallmajo-Martin
- Department of Obstetrics, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland; (Q.V.-M.); (M.E.)
| | - Shikha Chawla
- Department of Biomedicine, Tissue Engineering Laboratory, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (R.E.T.); (S.C.); (K.P.); (A.B.)
| | - Karoliina Pelttari
- Department of Biomedicine, Tissue Engineering Laboratory, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (R.E.T.); (S.C.); (K.P.); (A.B.)
| | - Marcus Mumme
- Department of Orthopaedic Surgery and Traumatology, University Hospital of Basel, 4031 Basel, Switzerland; (G.L.); (M.M.); (C.E.)
- Department of Biomedicine, Tissue Engineering Laboratory, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (R.E.T.); (S.C.); (K.P.); (A.B.)
- Department of Orthopaedic Surgery, University Children’s Hospital of Basel, 4056 Basel, Switzerland
| | - Martin Haug
- Department of Plastic, Reconstructive and Aesthetic Surgery and Hand Surgery, University Hospital of Basel, 4031 Basel, Switzerland;
| | - Christian Egloff
- Department of Orthopaedic Surgery and Traumatology, University Hospital of Basel, 4031 Basel, Switzerland; (G.L.); (M.M.); (C.E.)
| | | | - Martin Ehrbar
- Department of Obstetrics, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland; (Q.V.-M.); (M.E.)
| | - Ivan Martin
- Department of Biomedicine, Tissue Engineering Laboratory, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (R.E.T.); (S.C.); (K.P.); (A.B.)
- Correspondence: ; Tel.: +41-61-2652384; Fax: +41-61-2653990
| | - Andrea Barbero
- Department of Biomedicine, Tissue Engineering Laboratory, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (R.E.T.); (S.C.); (K.P.); (A.B.)
| |
Collapse
|
25
|
Wu S, Guo W, Li R, Zhang X, Qu W. Progress of Platelet Derivatives for Cartilage Tissue Engineering. Front Bioeng Biotechnol 2022; 10:907356. [PMID: 35782516 PMCID: PMC9243565 DOI: 10.3389/fbioe.2022.907356] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/05/2022] [Indexed: 11/13/2022] Open
Abstract
Articular cartilage has limited self-regeneration ability for lacking of blood vessels, nerves, and lymph that makes it a great challenge to repair defects of the tissue and restore motor functions of the injured or aging population. Platelet derivatives, such as platelet-rich plasma, have been proved effective, safe, and economical in musculoskeletal diseases for their autologous origin and rich in growth factors. The combination of platelet derivatives with biomaterials provides both mechanical support and localized sustained release of bioactive molecules in cartilage tissue engineering and low-cost efficient approaches of potential treatment. In this review, we first provide an overview of platelet derivatives and their application in clinical and experimental therapies, and then we further discuss the techniques of the addition of platelet derivatives and their influences on scaffold properties. Advances in cartilage tissue engineering with platelet derivatives as signal factors and structural components are also introduced before prospects and concerns in this research field. In short, platelet derivatives have broad application prospects as an economical and effective enhancement for tissue engineering–based articular cartilage repair.
Collapse
Affiliation(s)
- Siyu Wu
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Wenlai Guo
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Rui Li
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Xi Zhang
- Department of Burn Surgery, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Xi Zhang, ; Wenrui Qu,
| | - Wenrui Qu
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Xi Zhang, ; Wenrui Qu,
| |
Collapse
|
26
|
Naskou M, Tyma J, Gordon J, Berezny A, Kemelmakher H, Chocallo Richey A, Peroni J. Equine platelet lysate gel: a matrix for mesenchymal stem cell delivery. Stem Cells Dev 2022; 31:569-578. [PMID: 35678071 DOI: 10.1089/scd.2022.0097] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
A variety of bio-scaffolds have been developed as carriers for the delivery of Mesenchymal Stem Cells (MSCs) however many of them are unable to provide direct cell nourishment, a critical factor for survival and retention of MSCs at the site of delivery. Platelet lysate (PL) is a plasma derived product rich in growth factors, that can be turned into a gel matrix following the addition of calcium chloride. Our objective was to characterize growth factor and cytokine release of equine PL gel (ePL gel) encapsulated with MSCs over time and to measure the viability and proliferation of ePL gel-encapsulated MSCs for up to 14 days. Release of interleukin-1β (IL-1β), interleukin-10 (IL-10), transforming growth factor beta (TGF-β), vascular endothelial growth factor (VEGF), and platelet derived growth factor (PDGF-BB), as well as fibrinogen degradation, were measured from ePL gel with and without equine bone marrow derived MSCs and compared to MSCs in monolayer. MSC proliferation and viability within the gel were assessed up to 14 days. Compared to monolayer MSC cultures, significantly higher concentrations of IL-1β, IL-10, and TGF-β were measured from supernatants collected from ePL gel containing MSCs at various time points. Significantly lower concentrations of PDGF-BB were measured in the supernatant when MSCs were incorporated in ePL gel while VEGF tended to be increased compared to MSCs in monolayer. Incorporation in ePL gel for up to 14 days did not appear to affect viability and proliferation rates of MSCs as these were found to be similar to those measured in monolayer cell culture. ePL gel may have the potential to serve as bio-scaffold for MSC delivery since it appears to support the proliferation and viability of MSCs for up to 14 days.
Collapse
Affiliation(s)
- Maria Naskou
- Auburn University College of Veterinary Medicine, 70721, Pathobiology and Scott-Ritchey Research Center , Auburn, Alabama, United States;
| | - Jesse Tyma
- Mid-Atlantic Equine Medical Center, Ringoes, New Jersey, United States;
| | - Julie Gordon
- University of Georgia, 1355, Department of Large Animal Medicine , Athens, Georgia, United States;
| | - Alysha Berezny
- Duke Clinical Research Institute (DCRI), Durham, North Carolina, United States;
| | - Hannah Kemelmakher
- University of Georgia, 1355, Department of Large Animal Medicine , Athens, Georgia, United States;
| | - Anna Chocallo Richey
- University of Georgia, 1355, Department of Large Animal Medicine , Athens, Georgia, United States;
| | - John Peroni
- University of Georgia, 1355, Department of Large Animal Medicine , Athens, Georgia, United States;
| |
Collapse
|
27
|
Kuroda A, Saito W, Inoue G, Miyagi M, Shoji S, Sekiguchi H, Takaso M, Uchida K. Effect of Bone Morphogenetic Protein-2 (BMP-2)/Hydroxyapatite/In Situ-Formed Hyaluronan Hydrogel Composites on Bone Formation in a Murine Model of Posterolateral Lumbar Fusion. Cureus 2022; 14:e25509. [PMID: 35663656 PMCID: PMC9152467 DOI: 10.7759/cureus.25509] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2022] [Indexed: 11/18/2022] Open
Abstract
Several types of calcium phosphate (CaP) biomaterial carriers have been designed to deliver bone morphogenetic protein-2 (BMP-2) to augment spinal fusion in spinal surgery. Here, we evaluated an in situ-formed hydrogel (IFH) constructed from hyaluronan (IFH-HA) combined with a BMP2/hydroxyapatite (HAP) composite in bone formation in a murine model of posterolateral lumbar fusion (PLF). HAP was submerged in HA-tyramine (TA) polymer solution containing horseradish peroxidase (HRP) and 2 µg BMP-2 (BMP2/HA-TA/HRP solution). H2O2 was added to initiate the curing reaction (BMP-2/IFH-HA). phosphate-buffered saline (PBS) was added to the BMP2/HA-TA/HRP solution (BMP-2/HA-TA) instead of H2O2 to evaluate the effectiveness of the curing reaction. HAP immersed in PBS was used as a control. PLF model mice were randomly assigned to receive one these composites (n = 10 each). X-ray images were taken to assess the bone fusion, and microcomputed tomography analysis was conducted to examine new bone formation at the graft site four weeks following surgery. No evidence of fusion was observed four weeks after surgery in the Control or BMP2/HA-TA group. In contrast, the BMP2/IFH-HA group exhibited newly formed bone between the transverse processes and bone union in coronal sections. Relative to the Control and BMP2/HA-TA groups, the BMP2/IFH-HA group showed significantly greater bone volume. The BMP2/IFH-HA group also showed significantly elevated bone mineral content relative to the BMP2/HA-TA group. A composite comprising BMP2/HAP and IFH-HA, thus, enhanced the new bone formation in a murine model of PLF, suggesting its promise for augmenting spinal fusion.
Collapse
|
28
|
Edwards SD, Hou S, Brown JM, Boudreau RD, Lee Y, Kim YJ, Jeong KJ. Fast-Curing Injectable Microporous Hydrogel for In Situ Cell Encapsulation. ACS APPLIED BIO MATERIALS 2022; 5:2786-2794. [DOI: 10.1021/acsabm.2c00214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Seth D. Edwards
- Department of Chemical Engineering, University of New Hampshire, Durham, New Hampshire 03824, United States
| | - Shujie Hou
- Department of Chemical Engineering, University of New Hampshire, Durham, New Hampshire 03824, United States
| | - Jason M. Brown
- Department of Chemical Engineering, University of New Hampshire, Durham, New Hampshire 03824, United States
| | - Ryann D. Boudreau
- Department of Chemical Engineering, University of New Hampshire, Durham, New Hampshire 03824, United States
| | - Yuhan Lee
- Engineering in Medicine, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Young Jo Kim
- Department of Chemical Engineering, University of New Hampshire, Durham, New Hampshire 03824, United States
| | - Kyung Jae Jeong
- Department of Chemical Engineering, University of New Hampshire, Durham, New Hampshire 03824, United States
| |
Collapse
|
29
|
Ladeira BMF, Gomes MC, Custódio CA, Mano JF. High-Throughput Production of Microsponges from Platelet Lysate for Tissue Engineering Applications. Tissue Eng Part C Methods 2022; 28:325-334. [PMID: 35343236 DOI: 10.1089/ten.tec.2022.0029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cell-based therapies require a large number of cells, as well as appropriate methods to deliver the cells to damaged tissue. Microcarriers provide an optimal platform for large-scale cell culture while also improving cell retention during cell delivery. However, this technology still presents significant challenges due to low-throughput fabrication methods and an inability of the microcarriers to recreate the properties of human tissue. This work proposes, for the first time, the use of methacryloyl platelet lysates (PLMA), a photocrosslinkable material derived from human platelet lysates, to produce porous microcarriers. Initially, high quantities of PLMA/alginate core-shell microcapsules are produced using coaxial electrospray. Subsequently, the microcapsules are collected, irradiated with ultraviolet light, washed, and freeze dried yielding PLMA microsponges. These microsponges are able to support the adhesion and proliferation of human adipose-derived stem cells, while also displaying potential in the assembly of autologous microtissues. Cell-laden microsponges were shown to self-organize into aggregates, suggesting possible applications in bottom-up tissue engineering applications. Impact Statement Microcarriers have increasingly been used as delivery platforms in cell therapy. Herein, the encapsulation of human-derived proteins in alginate microcapsules is proposed as a method to produce microcarriers from photopolymerizable materials. The capsules function as a template structure, which is then processed into spherical microparticles, which can be used in cell culture, cell delivery, and bottom-up assembly. As a proof of concept, this method was combined with lyophilization to process methacryloyl platelet lysates into injectable microsponges for cell delivery.
Collapse
Affiliation(s)
- Bruno M F Ladeira
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Maria C Gomes
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Catarina A Custódio
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - João F Mano
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
30
|
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: 0.5] [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
|
31
|
Nazarnezhad S, Kermani F, Askari VR, Hosseini SA, Ebrahimzadeh-Bideskan A, Moradi A, Oskuee RK, Mollazadeh S, Kargozar S. Preparation and Characterization of Platelet Lysate (PL)-Loaded Electrospun Nanofibers for Epidermal Wound Healing. J Pharm Sci 2022; 111:2531-2539. [DOI: 10.1016/j.xphs.2022.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/11/2022] [Accepted: 04/11/2022] [Indexed: 12/14/2022]
|
32
|
Recent Developments in Hyaluronic Acid-Based Hydrogels for Cartilage Tissue Engineering Applications. Polymers (Basel) 2022; 14:polym14040839. [PMID: 35215752 PMCID: PMC8963043 DOI: 10.3390/polym14040839] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/18/2022] [Accepted: 02/19/2022] [Indexed: 01/27/2023] Open
Abstract
Articular cartilage lesions resulting from injurious impact, recurring loading, joint malalignment, etc., are very common and encompass the risk of evolving to serious cartilage diseases such as osteoarthritis. To date, cartilage injuries are typically treated via operative procedures such as autologous chondrocyte implantation (ACI), matrix-associated autologous chondrocyte implantation (MACI) and microfracture, which are characterized by low patient compliance. Accordingly, cartilage tissue engineering (CTE) has received a lot of interest. Cell-laden hydrogels are favorable candidates for cartilage repair since they resemble the native tissue environment and promote the formation of extracellular matrix. Various types of hydrogels have been developed so far for CTE applications based on both natural and synthetic biomaterials. Among these materials, hyaluronic acid (HA), a principal component of the cartilage tissue which can be easily modified and biofunctionalized, has been favored for the development of hydrogels since it interacts with cell surface receptors, supports the growth of chondrocytes and promotes the differentiation of mesenchymal stem cells to chondrocytes. The present work reviews the various types of HA-based hydrogels (e.g., in situ forming hydrogels, cryogels, microgels and three-dimensional (3D)-bioprinted hydrogel constructs) that have been used for cartilage repair, specially focusing on the results of their preclinical and clinical assessment.
Collapse
|
33
|
Guo W, Douma L, Hu MH, Eglin D, Alini M, Šećerović A, Grad S, Peng X, Zou X, D'Este M, Peroglio M. Hyaluronic acid-based interpenetrating network hydrogel as a cell carrier for nucleus pulposus repair. Carbohydr Polym 2022; 277:118828. [PMID: 34893245 DOI: 10.1016/j.carbpol.2021.118828] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 10/08/2021] [Accepted: 10/27/2021] [Indexed: 01/19/2023]
Abstract
Hyaluronic acid (HA) is a key component of the intervertebral disc (IVD) that is widely investigated as an IVD biomaterial. One persisting challenge is introducing materials capable of supporting cell encapsulation and function, yet with sufficient mechanical stability. In this study, a hybrid interpenetrating polymer network (IPN) was produced as a non-covalent hydrogel, based on a covalently cross-linked HA (HA-BDDE) and HA-poly(N-isopropylacrylamide) (HA-pNIPAM). The hybrid IPN was investigated for its physicochemical properties, with histology and gene expression analysis to determine matrix deposition in vitro and in an ex vivo model. The IPN hydrogel displayed cohesiveness for at least one week and rheological properties resembling native nucleus pulposus (NP) tissue. When implanted in an ex vivo IVD organ culture model, the IPN supported cell viability, phenotype expression of encapsulated NP cells and IVD matrix production over four weeks under physiological loading. Overall, our results indicate the therapeutic potential of this HA-based IPN hydrogel for IVD regeneration.
Collapse
Affiliation(s)
- Wei Guo
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland; Department of Spinal Surgery, Orthopaedic Research Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China
| | - Luzia Douma
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Ming Hsien Hu
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - David Eglin
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Mauro Alini
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Amra Šećerović
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Sibylle Grad
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Xinsheng Peng
- Department of Spinal Surgery, Orthopaedic Research Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China
| | - Xuenong Zou
- Department of Spinal Surgery, Orthopaedic Research Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China
| | - Matteo D'Este
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland.
| | - Marianna Peroglio
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| |
Collapse
|
34
|
Garcia-Orue I, Santos-Vizcaino E, Sanchez P, Gutierrez FB, Aguirre JJ, Hernandez RM, Igartua M. Bioactive and degradable hydrogel based on human platelet-rich plasma fibrin matrix combined with oxidized alginate in a diabetic mice wound healing model. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 135:112695. [DOI: 10.1016/j.msec.2022.112695] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 01/13/2022] [Accepted: 01/31/2022] [Indexed: 01/22/2023]
|
35
|
Rikkers M, Dijkstra K, Terhaard BF, Admiraal J, Levato R, Malda J, Vonk LA. Platelet-Rich Plasma Does Not Inhibit Inflammation or Promote Regeneration in Human Osteoarthritic Chondrocytes In Vitro Despite Increased Proliferation. Cartilage 2021; 13:991S-1003S. [PMID: 32969277 PMCID: PMC8721607 DOI: 10.1177/1947603520961162] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE The aims of the study were to assess the anti-inflammatory properties of platelet-rich plasma (PRP) and investigate its regenerative potential in osteoarthritic (OA) human chondrocytes. We hypothesized that PRP can modulate the inflammatory response and stimulate cartilage regeneration. DESIGN Primary human chondrocytes from OA knees were treated with manually prepared PRP, after which cell migration and proliferation were assessed. Next, tumor necrosis factor-α-stimulated chondrocytes were treated with a range of concentrations of PRP. Expression of genes involved in inflammation and chondrogenesis was determined by real-time polymerase chain reaction. In addition, chondrocytes were cultured in PRP gels and fibrin gels consisting of increasing concentrations of PRP. The production of cartilage extracellular matrix (ECM) was assessed. Deposition and release of glycosaminoglycans (GAG) and collagen was quantitatively determined and visualized by (immuno)histochemistry. Proliferation was assessed by quantitative measurement of DNA. RESULTS Both migration and the inflammatory response were altered by PRP, while proliferation was stimulated. Expression of chondrogenic markers COL2A1 and ACAN was downregulated by PRP, independent of PRP concentration. Chondrocytes cultured in PRP gel for 28 days proliferated significantly more when compared with chondrocytes cultured in fibrin gels. This effect was dose dependent. Significantly less GAGs and collagen were produced by chondrocytes cultured in PRP gels when compared with fibrin gels. This was qualitatively confirmed by histology. CONCLUSIONS PRP stimulated chondrocyte proliferation, but not migration. Also, production of cartilage ECM was strongly downregulated by PRP. Furthermore, PRP did not act anti-inflammatory on chondrocytes in an in vitro inflammation model.
Collapse
Affiliation(s)
- Margot Rikkers
- Department of Orthopaedics, University
Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Koen Dijkstra
- Department of Orthopaedics, University
Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Bastiaan F. Terhaard
- Department of Orthopaedics, University
Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jon Admiraal
- Department of Orthopaedics, University
Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Riccardo Levato
- Department of Orthopaedics, University
Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands,Department of Equine Sciences, Faculty
of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jos Malda
- Department of Orthopaedics, University
Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands,Department of Equine Sciences, Faculty
of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Lucienne A. Vonk
- Department of Orthopaedics, University
Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands,Lucienne A. Vonk, Department of
Orthopaedics, University Medical Center Utrecht, Utrecht University, Scientific
Liaison, CO.DON AG, Warthestraße 21, D-14513 Teltow, Germany.
| |
Collapse
|
36
|
Wei W, Dai H. Articular cartilage and osteochondral tissue engineering techniques: Recent advances and challenges. Bioact Mater 2021; 6:4830-4855. [PMID: 34136726 PMCID: PMC8175243 DOI: 10.1016/j.bioactmat.2021.05.011] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/20/2021] [Accepted: 05/11/2021] [Indexed: 12/18/2022] Open
Abstract
In spite of the considerable achievements in the field of regenerative medicine in the past several decades, osteochondral defect regeneration remains a challenging issue among diseases in the musculoskeletal system because of the spatial complexity of osteochondral units in composition, structure and functions. In order to repair the hierarchical tissue involving different layers of articular cartilage, cartilage-bone interface and subchondral bone, traditional clinical treatments including palliative and reparative methods have showed certain improvement in pain relief and defect filling. It is the development of tissue engineering that has provided more promising results in regenerating neo-tissues with comparable compositional, structural and functional characteristics to the native osteochondral tissues. Here in this review, some basic knowledge of the osteochondral units including the anatomical structure and composition, the defect classification and clinical treatments will be first introduced. Then we will highlight the recent progress in osteochondral tissue engineering from perspectives of scaffold design, cell encapsulation and signaling factor incorporation including bioreactor application. Clinical products for osteochondral defect repair will be analyzed and summarized later. Moreover, we will discuss the current obstacles and future directions to regenerate the damaged osteochondral tissues.
Collapse
Affiliation(s)
- Wenying Wei
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
- International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, 430070, China
| | - Honglian Dai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
- Foshan Xianhu Laboratory of the Advanced Energy Science and Technology Guangdong Laboratory, Xianhu Hydrogen Valley, Foshan, 528200, China
| |
Collapse
|
37
|
Combinations of Hydrogels and Mesenchymal Stromal Cells (MSCs) for Cartilage Tissue Engineering-A Review of the Literature. Gels 2021; 7:gels7040217. [PMID: 34842678 PMCID: PMC8628761 DOI: 10.3390/gels7040217] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/11/2021] [Accepted: 11/13/2021] [Indexed: 01/17/2023] Open
Abstract
Cartilage offers limited regenerative capacity. Cell-based approaches have emerged as a promising alternative in the treatment of cartilage defects and osteoarthritis. Due to their easy accessibility, abundancy, and chondrogenic potential mesenchymal stromal cells (MSCs) offer an attractive cell source. MSCs are often combined with natural or synthetic hydrogels providing tunable biocompatibility, biodegradability, and enhanced cell functionality. In this review, we focused on the different advantages and disadvantages of various natural, synthetic, and modified hydrogels. We examined the different combinations of MSC-subpopulations and hydrogels used for cartilage engineering in preclinical and clinical studies and reviewed the effects of added growth factors or gene transfer on chondrogenesis in MSC-laden hydrogels. The aim of this review is to add to the understanding of the disadvantages and advantages of various combinations of MSC-subpopulations, growth factors, gene transfers, and hydrogels in cartilage engineering.
Collapse
|
38
|
Lee SY, Ma J, Khoo TS, Abdullah N, Nik Md Noordin Kahar NNF, Abdul Hamid ZA, Mustapha M. Polysaccharide-Based Hydrogels for Microencapsulation of Stem Cells in Regenerative Medicine. Front Bioeng Biotechnol 2021; 9:735090. [PMID: 34733829 PMCID: PMC8558675 DOI: 10.3389/fbioe.2021.735090] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/27/2021] [Indexed: 12/29/2022] Open
Abstract
Stem cell-based therapy appears as a promising strategy to induce regeneration of damaged and diseased tissues. However, low survival, poor engraftment and a lack of site-specificity are major drawbacks. Polysaccharide hydrogels can address these issues and offer several advantages as cell delivery vehicles. They have become very popular due to their unique properties such as high-water content, biocompatibility, biodegradability and flexibility. Polysaccharide polymers can be physically or chemically crosslinked to construct biomimetic hydrogels. Their resemblance to living tissues mimics the native three-dimensional extracellular matrix and supports stem cell survival, proliferation and differentiation. Given the intricate nature of communication between hydrogels and stem cells, understanding their interaction is crucial. Cells are incorporated with polysaccharide hydrogels using various microencapsulation techniques, allowing generation of more relevant models and further enhancement of stem cell therapies. This paper provides a comprehensive review of human stem cells and polysaccharide hydrogels most used in regenerative medicine. The recent and advanced stem cell microencapsulation techniques, which include extrusion, emulsion, lithography, microfluidics, superhydrophobic surfaces and bioprinting, are described. This review also discusses current progress in clinical translation of stem-cell encapsulated polysaccharide hydrogels for cell delivery and disease modeling (drug testing and discovery) with focuses on musculoskeletal, nervous, cardiac and cancerous tissues.
Collapse
Affiliation(s)
- Si-Yuen Lee
- Department of Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Jingyi Ma
- Duke-NUS Medical School, Singapore, Singapore
| | - Tze Sean Khoo
- UKM Medical Molecular Biology Institute, National University of Malaysia, Bangi, Malaysia
| | - Norfadhilatuladha Abdullah
- Advanced Membrane Technology Research Centre, Faculty of Chemical and Energy Engineering, Universiti Teknologi Malaysia, Skudai, Malaysia
| | | | - Zuratul Ain Abdul Hamid
- School of Materials and Mineral Resources Engineering, Universiti Sains Malaysia, Nibong Tebal, Malaysia
| | - Muzaimi Mustapha
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| |
Collapse
|
39
|
Fang Y, Shi L, Duan Z, Rohani S. Hyaluronic acid hydrogels, as a biological macromolecule-based platform for stem cells delivery and their fate control: A review. Int J Biol Macromol 2021; 189:554-566. [PMID: 34437920 DOI: 10.1016/j.ijbiomac.2021.08.140] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 11/27/2022]
Abstract
Stem cell-based therapies offer numerous potentials to repair damaged or defective organs. The therapeutic outcomes of human studies, however, fall far short from what is expected. Enhancing stem cells local density and longevity would possibly maximize their healing potential. One promising strategy is to administer stem cells via injectable hydrogels. However, stem cells differentiation process is a delicate matter which is easily affected by various factors such as their interaction with their surrounding materials. Among various biomaterial options for hydrogels' production, hyaluronic acid (HA) has shown great promise. HA is a naturally occurring biological macromolecule, a polysaccharide of large molecular weight which is involved in cell proliferation, cell migration, angiogenesis, fetal development, and tissue function. In the current study we will discuss the applications, prospects, and challenges of HA-based hydrogels in stem cell delivery and fate control.
Collapse
Affiliation(s)
- Yu Fang
- Henan Provincial Engineering and Technology Research Center for Precise Synthesis of Fluorine-Containing Drugs, College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang, Henan 455000, People's Republic of China; Key Laboratory of New Opto-Electronic Functional Materials of Henan Province, College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang, Henan 455000, People's Republic of China.
| | - Lele Shi
- Henan Provincial Engineering and Technology Research Center for Precise Synthesis of Fluorine-Containing Drugs, College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang, Henan 455000, People's Republic of China; Key Laboratory of New Opto-Electronic Functional Materials of Henan Province, College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang, Henan 455000, People's Republic of China
| | - Zhiwei Duan
- Henan Provincial Engineering and Technology Research Center for Precise Synthesis of Fluorine-Containing Drugs, College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang, Henan 455000, People's Republic of China; Key Laboratory of New Opto-Electronic Functional Materials of Henan Province, College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang, Henan 455000, People's Republic of China
| | - Saeed Rohani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
Rial-Hermida MI, Rey-Rico A, Blanco-Fernandez B, Carballo-Pedrares N, Byrne EM, Mano JF. Recent Progress on Polysaccharide-Based Hydrogels for Controlled Delivery of Therapeutic Biomolecules. ACS Biomater Sci Eng 2021; 7:4102-4127. [PMID: 34137581 PMCID: PMC8919265 DOI: 10.1021/acsbiomaterials.0c01784] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 06/02/2021] [Indexed: 12/24/2022]
Abstract
A plethora of applications using polysaccharides have been developed in recent years due to their availability as well as their frequent nontoxicity and biodegradability. These polymers are usually obtained from renewable sources or are byproducts of industrial processes, thus, their use is collaborative in waste management and shows promise for an enhanced sustainable circular economy. Regarding the development of novel delivery systems for biotherapeutics, the potential of polysaccharides is attractive for the previously mentioned properties and also for the possibility of chemical modification of their structures, their ability to form matrixes of diverse architectures and mechanical properties, as well as for their ability to maintain bioactivity following incorporation of the biomolecules into the matrix. Biotherapeutics, such as proteins, growth factors, gene vectors, enzymes, hormones, DNA/RNA, and antibodies are currently in use as major therapeutics in a wide range of pathologies. In the present review, we summarize recent progress in the development of polysaccharide-based hydrogels of diverse nature, alone or in combination with other polymers or drug delivery systems, which have been implemented in the delivery of biotherapeutics in the pharmaceutical and biomedical fields.
Collapse
Affiliation(s)
- M. Isabel Rial-Hermida
- Department
of Chemistry, CICECO−Aveiro Institute of Materials, University of Aveiro 3810-193 Aveiro, Portugal
| | - Ana Rey-Rico
- Cell
Therapy and Regenerative Medicine
Unit, Centro de Investigacións Científicas Avanzadas
(CICA), Universidade da Coruña, 15071 A Coruña, Spain
| | - Barbara Blanco-Fernandez
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute of
Science and Technology, 08028 Barcelona, Spain
- CIBER
en Bioingeniería, Biomateriales y
Nanomedicina, CIBER-BBN, 28029 Madrid, Spain
| | - Natalia Carballo-Pedrares
- Cell
Therapy and Regenerative Medicine
Unit, Centro de Investigacións Científicas Avanzadas
(CICA), Universidade da Coruña, 15071 A Coruña, Spain
| | - Eimear M. Byrne
- Wellcome-Wolfson
Institute For Experimental Medicine, Queen’s
University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - João F. Mano
- Department
of Chemistry, CICECO−Aveiro Institute of Materials, University of Aveiro 3810-193 Aveiro, Portugal
| |
Collapse
|
41
|
Qiu G, Wu H, Huang M, Ma T, Schneider A, Oates TW, Weir MD, Xu HHK, Zhao L. Novel calcium phosphate cement with biofilm-inhibition and platelet lysate delivery to enhance osteogenesis of encapsulated human periodontal ligament stem cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112306. [PMID: 34474857 DOI: 10.1016/j.msec.2021.112306] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 01/09/2023]
Abstract
Osteomyelitis is caused by Staphylococcus aureus (S. aureus), with associated progressive bone loss. This study developed for the first time a calcium phosphate cement (CPC) for delivery of doxycycline (DOX) and human platelet lysate (hPL) to fight against S. aureus infection and enhance the osteogenesis of human periodontal ligament stem cells (hPDLSCs). Chitosan-containing CPC scaffolds were fabricated in the absence (CPCC) or presence of DOX (CPCC+DOX). In addition, hPL was encapsulated in alginate microbeads and incorporated into CPCC+DOX (CPCC+DOX+ hPL). Flexural strength of CPCC+DOX + hPL was (5.56 ± 0.55) MPa, lower than (8.26 ± 1.6) MPa of CPCC+DOX (p < 0.05), but exceeding the reported strength of cancellous bone. CPCC+DOX and CPCC+DOX + hPL exhibited strong antibacterial activity against S. aureus, reducing biofilm CFU by 4 orders of magnitude. The hPDLSCs encapsulated in microbeads were co-cultured with the CPCs. The hPDLSCs were able to be released from the microbeads and showed a high proliferation rate, increasing by about 8 folds at 14 days for all groups. The hPL was released from the scaffold and promoted the osteogenic differentiation of hPDLSCs. ALP activity was 28.07 ± 5.15 mU/mg for CPCC+DOX + hPL, higher than 17.36 ± 2.37 mU/mg and 1.34 ± 0.37 mU/mg of CPCC+DOX and CPCC, respectively (p < 0.05). At 7 days, osteogenic genes (ALP, RUNX2, COL-1, and OPN) in CPCC+DOX + hPL were 3-10 folds those of control. The amount of hPDLSC-synthesized bone mineral with CPCC+DOX + hPL was 3.8 folds that of CPCC (p < 0.05). In summary, the novel CPC + DOX + hPL-hPDLSCs scaffold exhibited strong antibacterial activity, excellent cytocompatibility and hPDLSC osteogenic differentiation, showing a promising approach for treatment and prevention of bone infection and enhancement of bone regeneration.
Collapse
Affiliation(s)
- Gengtao Qiu
- Department of Trauma and Joint Surgery, Shunde Hospital, Southern Medical University, Foshan, Guangdong, China; Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA; Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hansen Wu
- General Administration Office, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mingguang Huang
- Department of Trauma and Joint Surgery, Shunde Hospital, Southern Medical University, Foshan, Guangdong, China
| | - Tao Ma
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD 21201, USA; Member, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Thomas W Oates
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Michael D Weir
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA.
| | - Hockin H K Xu
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA; Member, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Liang Zhao
- Department of Trauma and Joint Surgery, Shunde Hospital, Southern Medical University, Foshan, Guangdong, China; Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
42
|
Mallis P, Sokolis DP, Katsimpoulas M, Kostakis A, Stavropoulos-Giokas C, Michalopoulos E. Improved Repopulation Efficacy of Decellularized Small Diameter Vascular Grafts Utilizing the Cord Blood Platelet Lysate. Bioengineering (Basel) 2021; 8:bioengineering8090118. [PMID: 34562940 PMCID: PMC8467559 DOI: 10.3390/bioengineering8090118] [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: 07/29/2021] [Revised: 08/18/2021] [Accepted: 08/24/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The development of functional bioengineered small-diameter vascular grafts (SDVGs), represents a major challenge of tissue engineering. This study aimed to evaluate the repopulation efficacy of biological vessels, utilizing the cord blood platelet lysate (CBPL). METHODS Human umbilical arteries (hUAs, n = 10) were submitted to decellularization. Then, an evaluation of decellularized hUAs, involving histological, biochemical and biomechanical analysis, was performed. Wharton's Jelly (WJ) Mesenchymal Stromal Cells (MSCs) were isolated and characterized for their properties. Then, WJ-MSCs (1.5 × 106 cells) were seeded on decellularized hUAs (n = 5) and cultivated with (Group A) or without the presence of the CBPL, (Group B) for 30 days. Histological analysis involving immunohistochemistry (against Ki67, for determination of cell proliferation) and indirect immunofluorescence (against activated MAP kinase, additional marker for cell growth and proliferation) was performed. RESULTS The decellularized hUAs retained their initial vessel's properties, in terms of key-specific proteins, the biochemical and biomechanical characteristics were preserved. The evaluation of the repopulation process indicated a more uniform distribution of WJ-MSCs in group A compared to group B. The repopulated vascular grafts of group B were characterized by greater Ki67 and MAP kinase expression compared to group A. CONCLUSION The results of this study indicated that the CBPL may improve the repopulation efficacy, thus bringing the biological SDVGs one step closer to clinical application.
Collapse
Affiliation(s)
- Panagiotis Mallis
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece; (C.S.-G.); (E.M.)
- Correspondence: ; Tel.: +30-2106597331 or +30-6971616467; Fax: +30-210-6597345
| | - Dimitrios P. Sokolis
- Laboratory of Biomechanics, Center for Experimental Surgery, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece;
| | - Michalis Katsimpoulas
- Center of Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece; (M.K.); (A.K.)
| | - Alkiviadis Kostakis
- Center of Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece; (M.K.); (A.K.)
| | - Catherine Stavropoulos-Giokas
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece; (C.S.-G.); (E.M.)
| | - Efstathios Michalopoulos
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece; (C.S.-G.); (E.M.)
| |
Collapse
|
43
|
Min SJ, Lee JS, Nah H, Kim SH, Moon HJ, Reis RL, Kwon IK, Heo DN. Development of photo-crosslinkable platelet lysate-based hydrogels for 3D printing and tissue engineering. Biofabrication 2021; 13. [PMID: 34330124 DOI: 10.1088/1758-5090/ac1993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 07/30/2021] [Indexed: 12/18/2022]
Abstract
Three-dimensional (3D) printing shows potential for use as an advanced technology for forming biomimetic tissue and other complex structures. However, there are limits and restrictions on selection of conventional bioinks. Here we report the first 3D-printable platelet lysate (PLMA)-based hydrogel, which consists of platelet lysate from whole blood of humans that can simulate the 3D structure of tissues and can be formed into a crosslinked hydrogel layer-by-layer to build cell-laden hydrogel constructs through methacrylated photo-polymerization. Furthermore, it can be customized for use with various tissues by controlling the physical properties according to irradiation time and concentration. In particular, different cells can be mixed and printed, and the integrity of the 3D printed structure can maintain its shape after crosslinking. The bio-ink exhibits excellent cell diffusion and proliferation at low concentrations, which improves moldability and biocompatibility. The 3D-printable PLMA bioinks may constitute a new strategy to create customized microenvironments for the repair of various tissuesin vivousing materials derived from the human body.
Collapse
Affiliation(s)
- Sung Jun Min
- Department of Dentistry, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Jae Seo Lee
- Department of Dentistry, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Haram Nah
- Department of Dentistry, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Seung Hyeon Kim
- Department of Dentistry, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Ho-Jin Moon
- Department of Dental Materials, School of Dentistry, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Rui L Reis
- Department of Dental Materials, School of Dentistry, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.,3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark-Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco/Guimarães, Portugal
| | - Il Keun Kwon
- Department of Dental Materials, School of Dentistry, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.,Kyung Hee University Medical Science Research Institute, Kyung Hee University, 23 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Dong Nyoung Heo
- Department of Dental Materials, School of Dentistry, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.,Biofriends Inc., 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| |
Collapse
|
44
|
An S, Choi S, Min S, Cho SW. Hyaluronic Acid-based Biomimetic Hydrogels for Tissue Engineering and Medical Applications. BIOTECHNOL BIOPROC E 2021. [DOI: 10.1007/s12257-020-0343-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
45
|
Chahal AS, Gómez-Florit M, Domingues RMA, Gomes ME, Tiainen H. Human Platelet Lysate-Loaded Poly(ethylene glycol) Hydrogels Induce Stem Cell Chemotaxis In Vitro. Biomacromolecules 2021; 22:3486-3496. [PMID: 34314152 PMCID: PMC8382254 DOI: 10.1021/acs.biomac.1c00573] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
![]()
Platelet lysates
(PL) contain a selection of proteins and growth
factors (GFs) that are known to mediate cell activity. Many of these
biomolecules have been identified as chemoattractants with the capacity
to induce cell migration. In order to effectively deliver and retain
these biomolecules to the site of injury, a scaffold containing PL
could be an option. We use poly(ethylene glycol) (PEG) hydrogels consisting
of 90 vol % PL to investigate their migratory potential on human mesenchymal
stem cells (hMSCs). Cells exposed to these hydrogels were tracked,
resulting in cell trajectories and detailed migratory parameters (velocity,
Euclidean distance, directness, and forward migration index). Volumetric
swelling ratios, hydrogel mechanical properties, and the release kinetics
of proteins and GFs from hydrogels were also assessed. Furthermore,
hMSC spheroids were encapsulated within the hydrogels to qualitatively
assess cell invasion by means of sprouting and disintegration of the
spheroid. Cell spheroids encapsulated within the PL-PEG gels exhibited
initial outgrowths and eventually colonized the 3D matrix successfully.
Results from this study confirmed that hMSCs exhibit directional migration
toward the PL-loaded hydrogel with increased velocity and directness,
compared to the controls. Overall, the incorporation of PL renders
the PEG hydrogel bioactive. This study demonstrates the capacity of
PL-loaded hydrogel constructs to attract stem cells for endogenous
tissue engineering purposes.
Collapse
Affiliation(s)
- Aman S Chahal
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, Geitmyrsveien 69-71, 0455 Oslo, Norway
| | - Manuel Gómez-Florit
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark-Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
| | - Rui M A Domingues
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark-Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
| | - Manuela E Gomes
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark-Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
| | - Hanna Tiainen
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, Geitmyrsveien 69-71, 0455 Oslo, Norway
| |
Collapse
|
46
|
Jiang WC, Hsu WY, Ao-Ieong WS, Wang CY, Wang J, Yet SF. A novel engineered vascular construct of stem cell-laden 3D-printed PGSA scaffold enhances tissue revascularization. Biofabrication 2021; 13. [PMID: 34233298 DOI: 10.1088/1758-5090/ac1259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 07/07/2021] [Indexed: 12/26/2022]
Abstract
Development of transplantable engineered tissue has been hampered by lacking vascular network within the engineered tissue. Three-dimensional (3D) printing has emerged as a new technology with great potential in fabrication and customization of geometric microstructure. In this study, utilizing digital light processing system, we manufactured a recently designed novel 3D architecture scaffold with poly(glycerol sebacate) acrylate (PGSA). Vascular construct was subsequently generated by seeding stem cells within this scaffold. PGSA provided inductive substrate in terms of supporting three-germ layer differentiation of embryonic stem cells (ESCs) and also promoting ESCs-derived vascular progenitor cells (VPCs) differentiation into endothelial cells (ECs). Furthermore, the differentiation efficiency of VPCs into ECs on PGSA was much higher than that on collagen IV or fibronectin. The results from seeding VPCs in the rotating hexagonal PGSA scaffold suggest that this architectural framework is highly efficient for cell engraftment in 3D structures. After long-term suspension culture of the VPCs in scaffold under directed EC differentiation condition, VPC-differentiated ECs were populated in the scaffold and expressed EC markers. Transplantation of the vascular construct in mice resulted in formation of new vascular network and integration of the microvasculature within the scaffold into the existing vasculature of host tissue. Importantly, in a mouse model of wound healing, ECs from the transplanted vascular construct directly contributed to revascularization and enhanced blood perfusion at the injured site. Collectively, this transplantable vascular construct provides an innovative alternative therapeutic strategy for vascular tissue engineering.
Collapse
Affiliation(s)
- Wei-Cheng Jiang
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Wan-Yuan Hsu
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Wai-Sam Ao-Ieong
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chun-Yen Wang
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Jane Wang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Shaw-Fang Yet
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 35053, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
47
|
Haghwerdi F, Khozaei Ravari M, Taghiyar L, Shamekhi MA, Jahangir S, Haririan I, Baghaban Eslaminejad M. Application of bone and cartilage extracellular matrices in articular cartilage regeneration. Biomed Mater 2021; 16. [PMID: 34102624 DOI: 10.1088/1748-605x/ac094b] [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: 12/07/2020] [Accepted: 06/08/2021] [Indexed: 01/02/2023]
Abstract
Articular cartilage has an avascular structure with a poor ability for self-repair; therefore, many challenges arise in cases of trauma or disease. It is of utmost importance to identify the proper biomaterial for tissue repair that has the capability to direct cell recruitment, proliferation, differentiation, and tissue integration by imitating the natural microenvironment of cells and transmitting an orchestra of intracellular signals. Cartilage extracellular matrix (cECM) is a complex nanostructure composed of divergent proteins and glycosaminoglycans (GAGs), which regulate many functions of resident cells. Numerous studies have shown the remarkable capacity of ECM-derived biomaterials for tissue repair and regeneration. Moreover, given the importance of biodegradability, biocompatibility, 3D structure, porosity, and mechanical stability in the design of suitable scaffolds for cartilage tissue engineering, demineralized bone matrix (DBM) appears to be a promising biomaterial for this purpose, as it possesses the aforementioned characteristics inherently. To the best of the authors' knowledge, no comprehensive review study on the use of DBM in cartilage tissue engineering has previously been published. Since so much work is needed to address DBM limitations such as pore size, cell retention, and so on, we decided to draw the attention of researchers in this field by compiling a list of recent publications. This review discusses the implementation of composite scaffolds of natural or synthetic origin functionalized with cECM or DBM in cartilage tissue engineering. Cutting-edge advances and limitations are also discussed in an attempt to provide guidance to researchers and clinicians.
Collapse
Affiliation(s)
- Fatemeh Haghwerdi
- Department of Pharmaceutical Biomaterials, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mojtaba Khozaei Ravari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran Iran
| | - Leila Taghiyar
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran Iran
| | - Mohammad Amin Shamekhi
- Department of Polymer Engineering, Islamic Azad University, Sarvestan Branch, Sarvestan, Iran
| | - Shahrbano Jahangir
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran Iran
| | - Ismaeil Haririan
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center (MBRC), Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran Iran
| |
Collapse
|
48
|
Antich C, Jiménez G, de Vicente J, López‐Ruiz E, Chocarro‐Wrona C, Griñán‐Lisón C, Carrillo E, Montañez E, Marchal JA. Development of a Biomimetic Hydrogel Based on Predifferentiated Mesenchymal Stem-Cell-Derived ECM for Cartilage Tissue Engineering. Adv Healthc Mater 2021; 10:e2001847. [PMID: 33646595 PMCID: PMC11468687 DOI: 10.1002/adhm.202001847] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/06/2021] [Indexed: 12/20/2022]
Abstract
The use of decellularized extracellular matrix (dECM) as a biomaterial has been an important step forward for the development of functional tissue constructs. In addition to tissues and organs, cell cultures are gaining a lot of attention as an alternative source of dECM. In this work, a novel biomimetic hydrogel is developed based on dECM obtained from mesenchymal stem cells (mdECM) for cartilage tissue engineering. To this end, cells are seeded under specific culture conditions to generate an early chondrogenic extracellular matrix (ECM) providing cues and elements necessary for cartilage development. The composition is determined by quantitative, histological, and mass spectrometry techniques. Moreover, the decellularization process is evaluated by measuring the DNA content and compositional analyses, and the hydrogel is formulated at different concentrations (3% and 6% w/v). Results show that mdECM derived hydrogels possess excellent biocompatibility and suitable physicochemical and mechanical properties for their injectability. Furthermore, it is evidenced that this hydrogel is able to induce chondrogenesis of mesenchymal stem cells (MSCs) without supplemental factors and, furthermore, to form hyaline cartilage-like tissue after in vivo implantation. These findings demonstrate for the first time the potential of this hydrogel based on mdECM for applications in cartilage repair and regeneration.
Collapse
Affiliation(s)
- Cristina Antich
- Department of Human Anatomy and EmbryologyFaculty of MedicineUniversity of GranadaGranada18016Spain
- Instituto de Investigación Biosanitaria ibs. GRANADAUniversity of GranadaGranada18014Spain
- Biopathology and Regenerative Medicine Institute (IBIMER)Centre for Biomedical ResearchUniversity of GranadaGranada18100Spain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranada18016Spain
| | - Gema Jiménez
- Instituto de Investigación Biosanitaria ibs. GRANADAUniversity of GranadaGranada18014Spain
- Biopathology and Regenerative Medicine Institute (IBIMER)Centre for Biomedical ResearchUniversity of GranadaGranada18100Spain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranada18016Spain
- Department of Health ScienceFaculty of Experimental ScienceUniversity of JaénJaén23071Spain
| | - Juan de Vicente
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranada18016Spain
- Biocolloid and Fluid Physics GroupDepartment of Applied PhysicsFaculty of SciencesUniversity of GranadaGranada18071Spain
| | - Elena López‐Ruiz
- Instituto de Investigación Biosanitaria ibs. GRANADAUniversity of GranadaGranada18014Spain
- Biopathology and Regenerative Medicine Institute (IBIMER)Centre for Biomedical ResearchUniversity of GranadaGranada18100Spain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranada18016Spain
- Department of Health ScienceFaculty of Experimental ScienceUniversity of JaénJaén23071Spain
| | - Carlos Chocarro‐Wrona
- Department of Human Anatomy and EmbryologyFaculty of MedicineUniversity of GranadaGranada18016Spain
- Instituto de Investigación Biosanitaria ibs. GRANADAUniversity of GranadaGranada18014Spain
- Biopathology and Regenerative Medicine Institute (IBIMER)Centre for Biomedical ResearchUniversity of GranadaGranada18100Spain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranada18016Spain
| | - Carmen Griñán‐Lisón
- Department of Human Anatomy and EmbryologyFaculty of MedicineUniversity of GranadaGranada18016Spain
- Instituto de Investigación Biosanitaria ibs. GRANADAUniversity of GranadaGranada18014Spain
- Biopathology and Regenerative Medicine Institute (IBIMER)Centre for Biomedical ResearchUniversity of GranadaGranada18100Spain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranada18016Spain
| | - Esmeralda Carrillo
- Department of Human Anatomy and EmbryologyFaculty of MedicineUniversity of GranadaGranada18016Spain
- Instituto de Investigación Biosanitaria ibs. GRANADAUniversity of GranadaGranada18014Spain
- Biopathology and Regenerative Medicine Institute (IBIMER)Centre for Biomedical ResearchUniversity of GranadaGranada18100Spain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranada18016Spain
| | - Elvira Montañez
- Department of Orthopedic Surgery and TraumatologyVirgen de la Victoria University HospitalMálaga29010Spain
- Biomedical Research Institute of Malaga (IBIMA)Virgen de la Victoria University HospitalMálaga29010Spain
| | - Juan A. Marchal
- Department of Human Anatomy and EmbryologyFaculty of MedicineUniversity of GranadaGranada18016Spain
- Instituto de Investigación Biosanitaria ibs. GRANADAUniversity of GranadaGranada18014Spain
- Biopathology and Regenerative Medicine Institute (IBIMER)Centre for Biomedical ResearchUniversity of GranadaGranada18100Spain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranada18016Spain
| |
Collapse
|
49
|
Weitkamp JT, Wöltje M, Nußpickel B, Schmidt FN, Aibibu D, Bayer A, Eglin D, Armiento AR, Arnold P, Cherif C, Lucius R, Smeets R, Kurz B, Behrendt P. Silk Fiber-Reinforced Hyaluronic Acid-Based Hydrogel for Cartilage Tissue Engineering. Int J Mol Sci 2021; 22:ijms22073635. [PMID: 33807323 PMCID: PMC8036422 DOI: 10.3390/ijms22073635] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/21/2021] [Accepted: 03/25/2021] [Indexed: 12/17/2022] Open
Abstract
A continuing challenge in cartilage tissue engineering for cartilage regeneration is the creation of a suitable synthetic microenvironment for chondrocytes and tissue regeneration. The aim of this study was to develop a highly tunable hybrid scaffold based on a silk fibroin matrix (SM) and a hyaluronic acid (HA) hydrogel. Human articular chondrocytes were embedded in a porous 3-dimensional SM, before infiltration with tyramine modified HA hydrogel. Scaffolds were cultured in chondropermissive medium with and without TGF-β1. Cell viability and cell distribution were assessed using CellTiter-Blue assay and Live/Dead staining. Chondrogenic marker expression was detected using qPCR. Biosynthesis of matrix compounds was analyzed by dimethylmethylene blue assay and immuno-histology. Differences in biomaterial stiffness and stress relaxation were characterized using a one-step unconfined compression test. Cell morphology was investigated by scanning electron microscopy. Hybrid scaffold revealed superior chondro-inductive and biomechanical properties compared to sole SM. The presence of HA and TGF-β1 increased chondrogenic marker gene expression and matrix deposition. Hybrid scaffolds offer cytocompatible and highly tunable properties as cell-carrier systems, as well as favorable biomechanical properties.
Collapse
Affiliation(s)
- Jan-Tobias Weitkamp
- Department of Anatomy, Christian-Albrechts-University Kiel, 24118 Kiel, Germany; (B.N.); (A.B.); (P.A.); (R.L.); (B.K.)
- Department of Oral and Maxillofacial Surgery, Division of Regenerative Orofacial Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany;
- Correspondence:
| | - Michael Wöltje
- Institute of Textile Machinery and High Performance Material Technology, 01069 Dresden, Germany; (M.W.); (D.A.); (C.C.)
| | - Bastian Nußpickel
- Department of Anatomy, Christian-Albrechts-University Kiel, 24118 Kiel, Germany; (B.N.); (A.B.); (P.A.); (R.L.); (B.K.)
| | - Felix N. Schmidt
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 22529 Hamburg, Germany;
| | - Dilbar Aibibu
- Institute of Textile Machinery and High Performance Material Technology, 01069 Dresden, Germany; (M.W.); (D.A.); (C.C.)
| | - Andreas Bayer
- Department of Anatomy, Christian-Albrechts-University Kiel, 24118 Kiel, Germany; (B.N.); (A.B.); (P.A.); (R.L.); (B.K.)
| | - David Eglin
- Mines Saint-Etienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059 Sainbiose, Centre CIS, F-42023 Saint-Etienne, France;
| | | | - Philipp Arnold
- Department of Anatomy, Christian-Albrechts-University Kiel, 24118 Kiel, Germany; (B.N.); (A.B.); (P.A.); (R.L.); (B.K.)
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Chokri Cherif
- Institute of Textile Machinery and High Performance Material Technology, 01069 Dresden, Germany; (M.W.); (D.A.); (C.C.)
| | - Ralph Lucius
- Department of Anatomy, Christian-Albrechts-University Kiel, 24118 Kiel, Germany; (B.N.); (A.B.); (P.A.); (R.L.); (B.K.)
| | - Ralf Smeets
- Department of Oral and Maxillofacial Surgery, Division of Regenerative Orofacial Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany;
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, 20251 Ham-burg, Germany
| | - Bodo Kurz
- Department of Anatomy, Christian-Albrechts-University Kiel, 24118 Kiel, Germany; (B.N.); (A.B.); (P.A.); (R.L.); (B.K.)
| | - Peter Behrendt
- Clinic for Orthopedic and Trauma Surgery, University Medical Center Schleswig-Holstein, Campus Kiel, 24015 Kiel, Germany;
| |
Collapse
|
50
|
Chen J, Qiu T, Guo L, He L, Li X. Topology Reliable LCST-Type Behavior of ABA Triblock Polymer and Influence on Water Condensation and Crystallization. Macromol Rapid Commun 2021; 42:e2100024. [PMID: 33768621 DOI: 10.1002/marc.202100024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/15/2021] [Indexed: 01/26/2023]
Abstract
As a kind of smart material, thermoresponsive hydrogels are widely investigated and applied in many fields. Due to the limitation of the freezing temperature of the water, it is a challenge to further broaden their sol-gel transition temperature (Tgel ) range, especially below 0 °C. Herein, the lower critical solution temperature type of amphiphilic ABA triblock copolymers, synthesized via two-step reversible addition-fragmentation chain transfer (RAFT) polymerization is demonstrated. The hydrophilic A-block and the hydrophobic B-block are composed of poly(N,N-dimethylacrylamide) (PDMAA) and poly(diacetone acrylamide) (PDAAM), respectively. The degree of polymerization (DP) of both A-block and B-block shows a significant influence on the Tgel of triblock copolymer dispersion. By changing the length of these two blocks or physically blending these copolymers dispersions, the Tgel can be well adjusted in a temperature range from 45 to -10 °C. Moreover, When the Tgel is higher than 4 °C, the triblock copolymer coatings show a good anti-fogging property. And when the Tgel is around or lower than the freezing temperature of the water, aqueous dispersions of the triblock copolymer have an ice recrystallization inhibition activity, resulting in the decrease of average maximum grain size (MLGS) of ice crystal.
Collapse
Affiliation(s)
- Jing Chen
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Teng Qiu
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Longhai Guo
- Key Laboratory of Carbon Fiber and Functional Polymers, Ministry of Education, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Lifan He
- Beijing Engineering Research Center of Synthesis and Application of Waterborne Polymer, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Xiaoyu Li
- Key Laboratory of Carbon Fiber and Functional Polymers, Ministry of Education, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| |
Collapse
|