1
|
Vallmajo-Martin Q, Kiveliö AS, Metzger S, Milleret V, Lienemann PS, Carrara BM, Millan C, Ghayor C, Ochsenbein-Koelble N, Ehrbar M. Undifferentiated Human Amniotic Fluid Progenitor Cells Promote Bone Regeneration in Vivo. Adv Healthc Mater 2025; 14:e2300843. [PMID: 39930929 DOI: 10.1002/adhm.202300843] [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: 08/20/2024] [Revised: 01/22/2025] [Indexed: 04/18/2025]
Abstract
The treatment of large bone defects requires bone tissue substitutes. However, the lack of accessible autologous bone, especially in newborns with spina bifida or cleft palate conditions, severely limits therapeutic options involving bone grafts. Here, an engineering approach to reconstruct bone is presented by combining human amniocentesis-derived amniotic fluid progenitor cells (hAFCs) and a biomimetic, injectable, and fully synthetic poly(ethylene glycol) hydrogel that is crosslinked enzymatically by transglutaminase FXIII (TG-PEG). hAFCs are isolated by their colony-forming capacity, expanded in vitro, and undergo osteogenic, chondrogenic, or adipogenic differentiation under appropriate stimulation. When encapsulated in TG-PEG hydrogels, hAFCs rapidly deposit endogenous extracellular matrix (ECM) in vitro. hAFC-laden TG-PEG hydrogels containing low concentrations of bone morphogenetic protein (BMP-2) promote formation of ectopic bone organoids in vivo in a murine model without requiring prior in vitro differentiation. Strikingly, hAFC-induced constructs form as much bone in this model as adult bone marrow-derived stromal cells (hBMSCs), and significantly more than adipose-derived stromal cells (hASCs). Utilization of autologous hAFCs embedded in TG-PEG hydrogels presents a promising therapeutic strategy for bone replacement, particularly in fetuses and newborns where limited stem cell availability can be overcome through minimally invasive harvest of amniotic fluid.
Collapse
Affiliation(s)
- Queralt Vallmajo-Martin
- Department of Obstetrics, University Hospital Zürich (USZ), Zürich, 8091, Switzerland
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, 1015, Switzerland
| | - Anna-Sofia Kiveliö
- Department of Obstetrics, University Hospital Zürich (USZ), Zürich, 8091, Switzerland
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, 1015, Switzerland
| | - Stéphanie Metzger
- Department of Obstetrics, University Hospital Zürich (USZ), Zürich, 8091, Switzerland
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, 1015, Switzerland
| | - Vincent Milleret
- Department of Obstetrics, University Hospital Zürich (USZ), Zürich, 8091, Switzerland
| | - Philipp S Lienemann
- Department of Obstetrics, University Hospital Zürich (USZ), Zürich, 8091, Switzerland
| | - Bianca M Carrara
- Department of Obstetrics, University Hospital Zürich (USZ), Zürich, 8091, Switzerland
| | - Christopher Millan
- Department of Urology, University Hospital Zürich (USZ), Zürich, 8091, Switzerland
| | - Chafik Ghayor
- Center of Dental Medicine, Oral Biotechnology & Bioengineering, University of Zürich (UZH), Zürich, 8006, Switzerland
| | | | - Martin Ehrbar
- Department of Obstetrics, University Hospital Zürich (USZ), Zürich, 8091, Switzerland
| |
Collapse
|
2
|
Moghaddam MM, Jooybar E, Imani R, Ehrbar M. Development of injectable microgel-based scaffolds via enzymatic cross-linking of hyaluronic acid-tyramine/gelatin-tyramine for potential bone tissue engineering. Int J Biol Macromol 2024; 279:135176. [PMID: 39214205 DOI: 10.1016/j.ijbiomac.2024.135176] [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: 05/08/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Currently, the healing of large bone defects relies on invasive surgeries and the transplantation of autologous bone. As a less invasive treatment option, the provision of microenvironments that promote the regeneration of defective bones holds great promise. Here, we developed hyaluronic acid (HA)/gelatin (Ge) microgel-based scaffolds to guide bone regeneration. To enable the formation of microgels by enzymatic cross-linking in the presence of horseradish peroxidase (HRP) and hydrogen peroxide (H2O2), we modified the polymers with tyramine (TA). Spectrophotometry and proton nuclear magnetic resonance (1H NMR) spectroscopy analysis confirmed successful tyramine substitution on polymer backbones. To enable the formation of microgels by a water-in-oil emulsion approach, the HRP and H2O2 concentrations were tuned to achieve the gelation in a few seconds. By varying the stirring speed from 600 to 1000 rpm, spherical microgels were produced with an average size of 116 ± 8.7 and 68 ± 4.7 μm, respectively. The results showed that microgels were injectable through needles and showed good biocompatibility with the cultured human osteosarcoma cell line (MG-63). HA/Ge-TA microgels served as a promising substrate for MG-63 cells since they improved the alkaline phosphatase activity and level of calcium deposition. In summary, the developed HA/Ge-TA microgels are promising injectable microgel-based scaffolds in bone tissue engineering.
Collapse
Affiliation(s)
- Melika Mansouri Moghaddam
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Elaheh Jooybar
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.
| | - Rana Imani
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.
| | - Martin Ehrbar
- Department of Obstetrics, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
3
|
Niu X, Xiao S, Huang R, Huang D, Aifantis KE, Yu H, Xue C, Yin L, Dunne N, Li X. ZIF-8-modified hydrogel sequentially delivers angiogenic and osteogenic growth factors to accelerate vascularized bone regeneration. J Control Release 2024; 374:154-170. [PMID: 39127448 DOI: 10.1016/j.jconrel.2024.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
To realize high-quality vascularized bone regeneration, we developed a multifunctional hydrogel (SHPP-ZB) by incorporating BMP-2@ZIF-8/PEG-NH2 nanoparticles (NPs) into a sodium alginate/hydroxyapatite/polyvinyl alcohol hydrogel loaded with PDGF-BB, allowing for the sequential release of angiogenic and osteogenic growth factors (GFs) during bone repair. ZIF-8 served as a protective host for BMP-2 from degradation, ensuring high encapsulation efficiency and long-term bioactivity. The SHPP-ZB hydrogel exhibited enhanced mechanical strength and injectability, making it suitable for complex bone defects. It provided a swelling interface for tissue interlocking and the early release of Zn2+ and tannin acid (TA) to exert antioxidant and antibacterial effects, followed by the sequential release of angiogenic and osteogenic GFs to promote high-quality vascularized bone regeneration. In vitro experiments demonstrated the superior angiogenic and osteogenic properties of SHPP-ZB compared to other groups. In vivo experiments indicated that the sequential delivery of GFs via SHPP-ZB hydrogel could improve vascularized bone regeneration. Further, RNA sequencing analysis of regenerative bone tissue revealed that SHPP-ZB hydrogel promoted vascularized bone regeneration by regulating JUN, MAPK, Wnt, and calcium signaling pathways in vivo. This study presented a promising approach for efficient vascularized bone regeneration in large-scale bone defects.
Collapse
Affiliation(s)
- Xiaolian Niu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Shengzhao Xiao
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai 200011, China.
| | - Ruoyu Huang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Di Huang
- Department of Biomedical Engineering, Research Center for Nano-Biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Katerina E Aifantis
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Han Yu
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Chao Xue
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Lan Yin
- Key Laboratory of Advanced Materials of Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Nicholas Dunne
- Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Stokes Building, Collins Avenue, Dublin 9, Ireland
| | - Xiaoming Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| |
Collapse
|
4
|
Jiang Z, Chen L, Huang L, Yu S, Lin J, Li M, Gao Y, Yang L. Bioactive Materials That Promote the Homing of Endogenous Mesenchymal Stem Cells to Improve Wound Healing. Int J Nanomedicine 2024; 19:7751-7773. [PMID: 39099796 PMCID: PMC11297574 DOI: 10.2147/ijn.s455469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 04/23/2024] [Indexed: 08/06/2024] Open
Abstract
Endogenous stem cell homing refers to the transport of endogenous mesenchymal stem cells (MSCs) to damaged tissue. The paradigm of using well-designed biomaterials to induce resident stem cells to home in to the injured site while coordinating their behavior and function to promote tissue regeneration is known as endogenous regenerative medicine (ERM). ERM is a promising new avenue in regenerative therapy research, and it involves the mobilizing of endogenous stem cells for homing as the principal means through which to achieve it. Comprehending how mesenchymal stem cells home in and grasp the influencing factors of mesenchymal stem cell homing is essential for the understanding and design of tissue engineering. This review summarizes the process of MSC homing, the factors influencing the homing process, analyses endogenous stem cell homing studies of interest in the field of skin tissue repair, explores the integration of endogenous homing promotion strategies with cellular therapies and details tissue engineering strategies that can be used to modulate endogenous homing of stem cells. In addition to providing more systematic theories and ideas for improved materials for endogenous tissue repair, this review provides new perspectives to explore the complex process of tissue remodeling to enhance the rational design of biomaterial scaffolds and guide tissue regeneration strategies.
Collapse
Affiliation(s)
- Ziwei Jiang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Lianglong Chen
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Lei Huang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Shengxiang Yu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Jiabao Lin
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Mengyao Li
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Yanbin Gao
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Lei Yang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
5
|
Li X, Cheng Y, Gu P, Zhao C, Li Z, Tong L, Zeng W, Liang J, Luo E, Jiang Q, Zhou Z, Fan Y, Zhang X, Sun Y. Engineered Microchannel Scaffolds with Instructive Niches Reinforce Endogenous Bone Regeneration by Regulating CSF-1/CSF-1R Pathway. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310876. [PMID: 38321645 DOI: 10.1002/adma.202310876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/30/2024] [Indexed: 02/08/2024]
Abstract
Structural and physiological cues provide guidance for the directional migration and spatial organization of endogenous cells. Here, a microchannel scaffold with instructive niches is developed using a circumferential freeze-casting technique with an alkaline salting-out strategy. Thereinto, polydopamine-coated nano-hydroxyapatite is employed as a functional inorganic linker to participate in the entanglement and crystallization of chitosan molecules. This scaffold orchestrates the advantage of an oriented porous structure for rapid cell infiltration and satisfactory immunomodulatory capacity to promote stem cell recruitment, retention, and subsequent osteogenic differentiation. Transcriptomic analysis as well as its in vitro and in vivo verification demonstrates that essential colony-stimulating factor-1 (CSF-1) factor is induced by this scaffold, and effectively bound to the target colony-stimulating factor-1 receptor (CSF-1R) on the macrophage surface to activate the M2 phenotype, achieving substantial endogenous bone regeneration. This strategy provides a simple and efficient approach for engineering inducible bone regenerative biomaterials.
Collapse
Affiliation(s)
- Xing Li
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Yaling Cheng
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Peiyang Gu
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Chengkun Zhao
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Zhulian Li
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Lei Tong
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Weinan Zeng
- Department of Orthopedic Surgery and Orthopedic Research Institution, West China Hospital, Sichuan University, 17# Gaopeng Avenue, Chengdu, 610041, P. R. China
| | - Jie Liang
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- Sichuan Testing Center for Biomaterials and Medical Devices, Sichuan University, 29# Wangjiang Road, Chengdu, 610064, P. R. China
| | - En Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, 14#, 3rd, Section of Renmin South Road, Chengdu, 610041, P. R. China
| | - Qing Jiang
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Zongke Zhou
- Department of Orthopedic Surgery and Orthopedic Research Institution, West China Hospital, Sichuan University, 17# Gaopeng Avenue, Chengdu, 610041, P. R. China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Yong Sun
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| |
Collapse
|
6
|
Yang J, Tan Q, Li K, Liao J, Hao Y, Chen Y. Advances and Trends of Photoresponsive Hydrogels for Bone Tissue Engineering. ACS Biomater Sci Eng 2024; 10:1921-1945. [PMID: 38457377 DOI: 10.1021/acsbiomaterials.3c01485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2024]
Abstract
The development of static hydrogels as an optimal choice for bone tissue engineering (BTE) remains a difficult challenge primarily due to the intricate nature of bone healing processes, continuous physiological functions, and pathological changes. Hence, there is an urgent need to exploit smart hydrogels with programmable properties that can effectively enhance bone regeneration. Increasing evidence suggests that photoresponsive hydrogels are promising bioscaffolds for BTE due to their advantages such as controlled drug release, cell fate modulation, and the photothermal effect. Here, we review the current advances in photoresponsive hydrogels. The mechanism of photoresponsiveness and its advanced applications in bone repair are also elucidated. Future research would focus on the development of more efficient, safer, and smarter photoresponsive hydrogels for BTE. This review is aimed at offering comprehensive guidance on the trends of photoresponsive hydrogels and shedding light on their potential clinical application in BTE.
Collapse
Affiliation(s)
- Juan Yang
- West China School of Nursing/West China Hospital, Sichuan University, Chengdu 610041, PR China
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Qingqing Tan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Ka Li
- West China School of Nursing/West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Jinfeng Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Ying Hao
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yuwen Chen
- West China School of Nursing/West China Hospital, Sichuan University, Chengdu 610041, PR China
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu 610041, PR China
| |
Collapse
|
7
|
Yu Y, You Z, Li X, Lou F, Xiong D, Ye L, Wang Z. Injectable Nanocomposite Hydrogels with Strong Antibacterial, Osteoinductive, and ROS-Scavenging Capabilities for Periodontitis Treatment. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38497587 DOI: 10.1021/acsami.3c16577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Injectable antibacterial and osteoinductive hydrogels have received considerable attention for promoting bone regeneration owing to their versatile functionalities. However, a current hydrogel with antibacterial, osteoinductive, and antioxidant properties by a facile method for periodontitis treatment is still missing. To overcome this issue, we designed an injectable hydrogel system (GPM) composed of gelatin, Ti3C2Tx MXene nanosheets, and poly-l-lysine using a simple enzymatic cross-linking technique. Physicochemical characterization demonstrated that the GPM hydrogel matrix exhibited excellent stability, moderate tissue adhesion ability, and good mechanical behavior. The GPM hydrogels significantly inhibited the growth of Porphyromonas gingivalis, scavenged reactive oxygen species, attenuated inflammatory responses, and enhanced bone tissue regeneration. Intriguingly, the arrangement of the junctional epithelium, alveolar bone volume, and alveolar bone height in the GPM-treated periodontal disease group recovered to that of the healthy group. Therefore, our injectable hydrogel system with versatile functions may serve as an excellent tissue scaffold for the treatment of periodontitis.
Collapse
Affiliation(s)
- Yue Yu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ziying You
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xin Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Feng Lou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ding Xiong
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhenming Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
8
|
Lee J, Lee E, Huh SJ, Kang JI, Park KM, Byun H, Lee S, Kim E, Shin H. Composite Spheroid-Laden Bilayer Hydrogel for Engineering Three-Dimensional Osteochondral Tissue. Tissue Eng Part A 2024; 30:225-243. [PMID: 38062771 DOI: 10.1089/ten.tea.2023.0299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2024] Open
Abstract
A combination of hydrogels and stem cell spheroids has been used to engineer three-dimensional (3D) osteochondral tissue, but precise zonal control directing cell fate within the hydrogel remains a challenge. In this study, we developed a composite spheroid-laden bilayer hydrogel to imitate osteochondral tissue by spatially controlled differentiation of human adipose-derived stem cells. Meticulous optimization of the spheroid-size and mechanical strength of gelatin methacryloyl (GelMA) hydrogel enables the cells to homogeneously sprout within the hydrogel. Moreover, fibers immobilizing transforming growth factor beta-1 (TGF-β1) or bone morphogenetic protein-2 (BMP-2) were incorporated within the spheroids, which induced chondrogenic or osteogenic differentiation of cells in general media, respectively. The spheroids-filled GelMA solution was crosslinked to create the bilayer hydrogel, which demonstrated a strong interfacial adhesion between the two layers. The cell sprouting enhanced the adhesion of each hydrogel, demonstrated by increase in tensile strength from 4.8 ± 0.4 to 6.9 ± 1.2 MPa after 14 days of culture. Importantly, the spatially confined delivery of BMP-2 within the spheroids increased mineral deposition and more than threefold enhanced osteogenic genes of cells in the bone layer while the cells induced by TGF-β1 signals were apparently differentiated into chondrocytes within the cartilage layer. The results suggest that our composite spheroid-laden hydrogel could be used for the biofabrication of osteochondral tissue, which can be applied to engineer other complex tissues by delivery of appropriate biomolecules.
Collapse
Affiliation(s)
- Jinkyu Lee
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
- Department of Bioengineering, BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul, Republic of Korea
| | - Eunjin Lee
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
- Department of Bioengineering, BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul, Republic of Korea
| | - Seung Jae Huh
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
- Department of Bioengineering, BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul, Republic of Korea
| | - Jeon Il Kang
- Department of Bioengineering and Nano-Bioengineering, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Kyung Min Park
- Department of Bioengineering and Nano-Bioengineering, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Hayeon Byun
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
| | - Sangmin Lee
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
| | - Eunhyung Kim
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
- Department of Bioengineering, BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
- Department of Bioengineering, BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul, Republic of Korea
- Department of Bioengineering, Institute of Nano Science and Technology, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
9
|
Vallmajo-Martin Q, Millan C, Müller R, Weber FE, Ehrbar M, Ghayor C. Enhanced bone regeneration in rat calvarial defects through BMP2 release from engineered poly(ethylene glycol) hydrogels. Sci Rep 2024; 14:4916. [PMID: 38418564 PMCID: PMC10901800 DOI: 10.1038/s41598-024-55411-z] [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: 06/30/2023] [Accepted: 02/23/2024] [Indexed: 03/01/2024] Open
Abstract
The clinical standard therapy for large bone defects, typically addressed through autograft or allograft donor tissue, faces significant limitations. Tissue engineering offers a promising alternative strategy for the regeneration of substantial bone lesions. In this study, we harnessed poly(ethylene glycol) (PEG)-based hydrogels, optimizing critical parameters including stiffness, incorporation of arginine-glycine-aspartic acid (RGD) cell adhesion motifs, degradability, and the release of BMP2 to promote bone formation. In vitro we demonstrated that human bone marrow derived stromal cell (hBMSC) proliferation and spreading strongly correlates with hydrogel stiffness and adhesion to RGD peptide motifs. Moreover, the incorporation of the osteogenic growth factor BMP2 into the hydrogels enabled sustained release, effectively inducing bone regeneration in encapsulated progenitor cells. When used in vivo to treat calvarial defects in rats, we showed that hydrogels of low and intermediate stiffness optimally facilitated cell migration, proliferation, and differentiation promoting the efficient repair of bone defects. Our comprehensive in vitro and in vivo findings collectively suggest that the developed hydrogels hold significant promise for clinical translation for bone repair and regeneration by delivering sustained and controlled stimuli from active signaling molecules.
Collapse
Affiliation(s)
- Queralt Vallmajo-Martin
- Department of Obstetrics, University Hospital Zürich, University of Zürich, Schmelzbergstrasse 12, 8091, Zurich, Switzerland
- School of Life Sciences and School of Engineering, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Station 15, 1015, Lausanne, Switzerland
| | - Christopher Millan
- Department of Urology, University Hospital Zürich, University of Zürich, Wagistrasse 21, 8952, Zurich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, Eidgenössische Technische Hochschule Zürich, Leopold-Ruzicka-Weg 8093, 8049, Zurich, Switzerland
| | - Franz E Weber
- Center of Dental Medicine, Oral Biotechnology & Bioengineering, University of Zürich, Plattenstrasse 11, 8032, Zurich, Switzerland
| | - Martin Ehrbar
- Department of Obstetrics, University Hospital Zürich, University of Zürich, Schmelzbergstrasse 12, 8091, Zurich, Switzerland.
| | - Chafik Ghayor
- Center of Dental Medicine, Oral Biotechnology & Bioengineering, University of Zürich, Plattenstrasse 11, 8032, Zurich, Switzerland.
| |
Collapse
|
10
|
Liu T, You Z, Shen F, Yang P, Chen J, Meng S, Wang C, Xiong D, You C, Wang Z, Shi Y, Ye L. Tricarboxylic Acid Cycle Metabolite-Coordinated Biohydrogels Augment Cranial Bone Regeneration Through Neutrophil-Stimulated Mesenchymal Stem Cell Recruitment and Histone Acetylation-Mediated Osteogenesis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:5486-5503. [PMID: 38284176 DOI: 10.1021/acsami.3c15473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Cranial bone defects remain a major clinical challenge, increasing patients' life burdens. Tricarboxylic acid (TCA) cycle metabolites play crucial roles in facilitating bone tissue regeneration. However, the development of TCA cycle metabolite-modified biomimetic grafts for skull bone regeneration still needs to be improved. The mechanism underlying the release of TCA cycle metabolites from biomaterials in regulating immune responses and mesenchymal stem cell (MSC) fate (migration and differentiation) remains unknown. Herein, this work constructs biomimetic hydrogels composed of gelatin and chitosan networks covalently cross-linked by genipin (CGG hydrogels). A series of TCA cycle metabolite-coordinated CGG hydrogels with strong mechanical and antiswelling performances are subsequently developed. Remarkably, the citrate (Na3Cit, Cit)-coordinated CGG hydrogels (CGG-Cit hydrogels) with the highest mechanical modulus and strength significantly promote skull bone regeneration in rat and murine cranial defects. Mechanistically, using a transgenic mouse model, bulk RNA sequencing, and single-cell RNA sequencing, this work demonstrates that CGG-Cit hydrogels promote Gli1+ MSC migration via neutrophil-secreted oncostatin M. Results also indicate that citrate improves osteogenesis via enhanced histone H3K9 acetylation on osteogenic master genes. Taken together, the immune microenvironment- and MSC fate-regulated CGG-Cit hydrogels represent a highly efficient and facile approach toward skull bone tissue regeneration with great potential for bench-to-bedside translation.
Collapse
Affiliation(s)
- Tingjun Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ziying You
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Fangyuan Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Puying Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Junyu Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shuhuai Meng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chenglin Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ding Xiong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chengjia You
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhenming Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yu Shi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
11
|
Yang B, Lin Y, Huang Y, Zhu N, Shen YQ. Extracellular vesicles modulate key signalling pathways in refractory wound healing. BURNS & TRAUMA 2023; 11:tkad039. [PMID: 38026441 PMCID: PMC10654481 DOI: 10.1093/burnst/tkad039] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 05/10/2023] [Accepted: 06/22/2023] [Indexed: 12/01/2023]
Abstract
Chronic wounds are wounds that cannot heal properly due to various factors, such as underlying diseases, infection or reinjury, and improper healing of skin wounds and ulcers can cause a serious economic burden. Numerous studies have shown that extracellular vesicles (EVs) derived from stem/progenitor cells promote wound healing, reduce scar formation and have significant advantages over traditional treatment methods. EVs are membranous particles that carry various bioactive molecules from their cellular origins, such as cytokines, nucleic acids, enzymes, lipids and proteins. EVs can mediate cell-to-cell communication and modulate various physiological processes, such as cell differentiation, angiogenesis, immune response and tissue remodelling. In this review, we summarize the recent advances in EV-based wound healing, focusing on the signalling pathways that are regulated by EVs and their cargos. We discuss how EVs derived from different types of stem/progenitor cells can promote wound healing and reduce scar formation by modulating the Wnt/β-catenin, phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin, vascular endothelial growth factor, transforming growth factor β and JAK-STAT pathways. Moreover, we also highlight the challenges and opportunities for engineering or modifying EVs to enhance their efficacy and specificity for wound healing.
Collapse
Affiliation(s)
- Bowen Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renmin South Road, Wuhou District, Chengdu 610041, China
| | - Yumeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renmin South Road, Wuhou District, Chengdu 610041, China
| | - Yibo Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renmin South Road, Wuhou District, Chengdu 610041, China
| | - Nanxi Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renmin South Road, Wuhou District, Chengdu 610041, China
| | - Ying-Qiang Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renmin South Road, Wuhou District, Chengdu 610041, China
| |
Collapse
|
12
|
Gholami Farashah MS, Mohammadi A, Javadi M, Soleimani Rad J, Shakouri SK, Meshgi S, Roshangar L. Bone marrow mesenchymal stem cells' osteogenic potential: superiority or non-superiority to other sources of mesenchymal stem cells? Cell Tissue Bank 2023; 24:663-681. [PMID: 36622494 DOI: 10.1007/s10561-022-10066-w] [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: 06/02/2022] [Accepted: 12/14/2022] [Indexed: 01/10/2023]
Abstract
Skeletal problems are an increasing issue due to the increase in the global aging population. Different statistics reports show that today, the global population is aging that results in skeletal problems, increased health system costs, and even higher mortality associated with skeletal problems. Common treatments such as surgery and bone grafts are not always effective and in some cases, they can even cause secondary problems such as infections or improper repair. Cell therapy is a method that can be utilized along with common treatments independently. Mesenchymal stem cells (MSCs) are a very important and efficient source in terms of different diseases, especially bone problems. These cells are present in different tissues such as bone marrow, adipose tissue, umbilical cord, placenta, dental pulp, peripheral blood, amniotic fluid and others. Among the types of MSCs, bone marrow mesenchymal stem cells (BMMSCs) are the most widely used source of these cells, which have appeared to be very effective and promising in terms of skeletal diseases, especially compared to the other sources of MSCs. This study focuses on the specific potential and content of BMMSCs from which the specific capacity of these cells originates, and compares their osteogenic potential with other types of MSCs, and also the future directions in the application of BMMSCs as a source for cell therapy.
Collapse
Affiliation(s)
- Mohammad Sadegh Gholami Farashah
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirhossein Mohammadi
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Javadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Soleimani Rad
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Kazem Shakouri
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahla Meshgi
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
13
|
Gu L, Huang R, Ni N, Gu P, Fan X. Advances and Prospects in Materials for Craniofacial Bone Reconstruction. ACS Biomater Sci Eng 2023; 9:4462-4496. [PMID: 37470754 DOI: 10.1021/acsbiomaterials.3c00399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
The craniofacial region is composed of 23 bones, which provide crucial function in keeping the normal position of brain and eyeballs, aesthetics of the craniofacial complex, facial movements, and visual function. Given the complex geometry and architecture, craniofacial bone defects not only affect the normal craniofacial structure but also may result in severe craniofacial dysfunction. Therefore, the exploration of rapid, precise, and effective reconstruction of craniofacial bone defects is urgent. Recently, developments in advanced bone tissue engineering bring new hope for the ideal reconstruction of the craniofacial bone defects. This report, presenting a first-time comprehensive review of recent advances of biomaterials in craniofacial bone tissue engineering, overviews the modification of traditional biomaterials and development of advanced biomaterials applying to craniofacial reconstruction. Challenges and perspectives of biomaterial development in craniofacial fields are discussed in the end.
Collapse
Affiliation(s)
- Li Gu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Rui Huang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Ni Ni
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Ping Gu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Xianqun Fan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| |
Collapse
|
14
|
Puiggalí-Jou A, Asadikorayem M, Maniura-Weber K, Zenobi-Wong M. Growth factor-loaded sulfated microislands in granular hydrogels promote hMSCs migration and chondrogenic differentiation. Acta Biomater 2023; 166:69-84. [PMID: 37030622 DOI: 10.1016/j.actbio.2023.03.045] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/10/2023]
Abstract
Cell-based therapies for articular cartilage lesions are expensive and time-consuming; clearly, a one-step procedure to induce endogenous repair would have significant clinical benefits. Acellular heterogeneous granular hydrogels were explored for their injectability, cell-friendly cross-linking, and ability to promote migration, as well as to serve as a scaffold for depositing cartilage extracellular matrix. The hydrogels were prepared by mechanical sizing of bulk methacrylated hyaluronic acid (HAMA) and bulk HAMA incorporating sulfated HAMA (SHAMA). SHAMA's negative charges allowed for the retention of positively charged growth factors (GFs) (e.g., TGFB3 and PDGF-BB). Mixtures of HAMA and GF-loaded SHAMA microgels were annealed by enzymatic cross-linking, forming heterogeneous granular hydrogels with GF deposits. The addition of GF loaded sulfated microislands guided cell migration and enhanced chondrogenesis. Granular heterogeneous hydrogels showed increased matrix deposition and cartilage tissue maturation compared to bulk or homogeneous granular hydrogels. This advanced material provides an ideal 3D environment for guiding cell migration and differentiation into cartilage. STATEMENT OF SIGNIFICANCE: Acellular materials which promote regeneration are of great interest for repair of cartilage defects, and they are more cost- and time-effective compared to current cell-based therapies. Here we develop an injectable, granular hydrogel system which promotes cell migration from the surrounding tissue, facilitating endogenous repair. The hydrogel architecture and chemistry were optimized to increase cell migration and extracellular matrix deposition. The present study provides quantitative data on the effect of microgel size and chemical modification on cell migration, growth factor retention and tissue maturation.
Collapse
Affiliation(s)
- Anna Puiggalí-Jou
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - Maryam Asadikorayem
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - Katharina Maniura-Weber
- Laboratory for Biointerfaces, Empa, Swiss Federal Laboratories for Materials Science and Technology, 9014 St. Gallen, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland.
| |
Collapse
|
15
|
Co CM, Nguyen T, Vaish B, Izuagbe S, Borrelli J, Tang L. Biomolecule-releasing bioadhesive for glenoid labrum repair through induced host progenitor cell responses. J Orthop Res 2023; 41:1624-1636. [PMID: 36448179 PMCID: PMC10355087 DOI: 10.1002/jor.25494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/18/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022]
Abstract
Glenoid labral tears occur with repetitive dislocation events and are common injuries observed in shoulder arthroscopic procedures. Although surgery can restore shoulder anatomy, repair is associated with poor clinical outcomes, which may be attributed to the poor regenerative capability of glenoid labral fibrocartilage. Thus, this study was designed to assess whether in situ tissue regeneration via biomolecule-stimulated recruitment of progenitor cells is a viable approach for the regeneration of labral tears. We developed a click chemistry-based bioadhesive to improve labral repair and reduce local inflammatory responses due to trauma. Additionally, we previously identified the presence of progenitor cells in the human labrum, which can be recruited by platelet-derived growth factor (PDGF). Thus, we hypothesized that PDGF-releasing adhesives could induce the regenerative responses of progenitor cells at the injury site to improve labral healing. In a rat glenoid labral tear model, we evaluated the effect of PDGF-releasing adhesives on promoting progenitor cells to participate in labral tear healing. After 3 and 6 weeks, the labrum was histologically analyzed for inflammatory responses, progenitor cell recruitment, proliferation, and extracellular matrix (ECM) production (collagen and glycosaminoglycan). Our results showed that adhesives alone considerably reduced local inflammatory responses and labral tissue dissolution. PDGF-releasing adhesives significantly increased progenitor cell recruitment, proliferation, and ECM production. These results demonstrate that by accelerating autologous progenitor cell responses, PDGF-releasing adhesives represent a novel clinically relevant strategy to improve the healing of glenoid labral tears.
Collapse
Affiliation(s)
- Cynthia M Co
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Tam Nguyen
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Bhavya Vaish
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Samira Izuagbe
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Joseph Borrelli
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Liping Tang
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA
| |
Collapse
|
16
|
Lee JC, Brien HJ, Walton BL, Eidman ZM, Toda S, Lim WA, Brunger JM. Instructional materials that control cellular activity through synthetic Notch receptors. Biomaterials 2023; 297:122099. [PMID: 37023529 PMCID: PMC10320837 DOI: 10.1016/j.biomaterials.2023.122099] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 03/13/2023] [Accepted: 03/22/2023] [Indexed: 03/31/2023]
Abstract
The field of regenerative engineering relies primarily on the dual technical platforms of cell selection/conditioning and biomaterial fabrication to support directed cell differentiation. As the field has matured, an appreciation for the influence of biomaterials on cell behaviors has resulted in engineered matrices that meet biomechanical and biochemical demands of target pathologies. Yet, despite advances in methods to produce designer matrices, regenerative engineers remain unable to reliably orchestrate behaviors of therapeutic cells in situ. Here, we present a platform named MATRIX whereby cellular responses to biomaterials can be custom defined by combining engineered materials with cells expressing cognate synthetic biology control modules. Such privileged channels of material-to-cell communication can activate synthetic Notch receptors and govern activities as diverse as transcriptome engineering, inflammation attenuation, and pluripotent stem cell differentiation, all in response to materials decorated with otherwise bioinert ligands. Further, we show that engineered cellular behaviors are confined to programmed biomaterial surfaces, highlighting the potential to use this platform to spatially organize cellular responses to bulk, soluble factors. This integrated approach of co-engineering cells and biomaterials for orthogonal interactions opens new avenues for reproducible control of cell-based therapies and tissue replacements.
Collapse
Affiliation(s)
- Joanne C Lee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Hannah J Brien
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Bonnie L Walton
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Zachary M Eidman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Satoshi Toda
- WPI Nano Life Science Institute (NanoLSI), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Wendell A Lim
- Cell Design Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, 94158, USA.
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA; Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, 37212, USA.
| |
Collapse
|
17
|
Oliver-Cervelló L, Martin-Gómez H, Gonzalez-Garcia C, Salmeron-Sanchez M, Ginebra MP, Mas-Moruno C. Protease-degradable hydrogels with multifunctional biomimetic peptides for bone tissue engineering. Front Bioeng Biotechnol 2023; 11:1192436. [PMID: 37324414 PMCID: PMC10267393 DOI: 10.3389/fbioe.2023.1192436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/23/2023] [Indexed: 06/17/2023] Open
Abstract
Mimicking bone extracellular matrix (ECM) is paramount to develop novel biomaterials for bone tissue engineering. In this regard, the combination of integrin-binding ligands together with osteogenic peptides represents a powerful approach to recapitulate the healing microenvironment of bone. In the present work, we designed polyethylene glycol (PEG)-based hydrogels functionalized with cell instructive multifunctional biomimetic peptides (either with cyclic RGD-DWIVA or cyclic RGD-cyclic DWIVA) and cross-linked with matrix metalloproteinases (MMPs)-degradable sequences to enable dynamic enzymatic biodegradation and cell spreading and differentiation. The analysis of the intrinsic properties of the hydrogel revealed relevant mechanical properties, porosity, swelling and degradability to engineer hydrogels for bone tissue engineering. Moreover, the engineered hydrogels were able to promote human mesenchymal stem cells (MSCs) spreading and significantly improve their osteogenic differentiation. Thus, these novel hydrogels could be a promising candidate for applications in bone tissue engineering, such as acellular systems to be implanted and regenerate bone or in stem cells therapy.
Collapse
Affiliation(s)
- Lluís Oliver-Cervelló
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, Spain
| | - Helena Martin-Gómez
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, Spain
| | - Cristina Gonzalez-Garcia
- Centre for the Cellular Microenvironment, Advanced Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Manuel Salmeron-Sanchez
- Centre for the Cellular Microenvironment, Advanced Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Maria-Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Carlos Mas-Moruno
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, Spain
| |
Collapse
|
18
|
Doron G, Pearson JJ, Guldberg RE, Temenoff JS. Development and characterization of Factor Xa-responsive materials for applications in cell culture and biologics delivery. J Biomed Mater Res A 2023; 111:634-643. [PMID: 36794576 DOI: 10.1002/jbm.a.37513] [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: 11/07/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/17/2023]
Abstract
Stimuli-responsive biomaterials may be used to better control the release of bioactive molecules or cells for applications involving drug delivery and controlled cell release. In this study, we developed a Factor Xa (FXa)-responsive biomaterial capable of controlled release of pharmaceutical agents and cells from in vitro culture. FXa-cleavable substrates were formed as hydrogels that degraded in response to FXa enzyme over several hours. Hydrogels were shown to release both heparin and a model protein in response to FXa. Additionally, RGD-functionalized FXa-degradable hydrogels were used to culture mesenchymal stromal cells (MSCs), enabling FXa-mediated cell dissociation from hydrogels in a manner that preserved multicellular structures. Harvesting MSCs using FXa-mediated dissociation did not influence their differentiation capacity or indoleamine 2,3-dioxygenase (IDO) activity (a measure of immunomodulatory capacity). In all, this FXa-degradable hydrogel is a novel responsive biomaterial system that may be used for on-demand drug delivery, as well as for improving processes for in vitro culture of therapeutic cells.
Collapse
Affiliation(s)
- Gilad Doron
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Joseph J Pearson
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Robert E Guldberg
- Knight Campus for Accelerating Scientific Impact, 6231 University of Oregon, Eugene, Oregon, USA
| | - Johnna S Temenoff
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
19
|
Review on Bioinspired Design of ECM-Mimicking Scaffolds by Computer-Aided Assembly of Cell-Free and Cell Laden Micro-Modules. J Funct Biomater 2023; 14:jfb14020101. [PMID: 36826900 PMCID: PMC9964438 DOI: 10.3390/jfb14020101] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/31/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Tissue engineering needs bioactive drug delivery scaffolds capable of guiding cell biosynthesis and tissue morphogenesis in three dimensions. Several strategies have been developed to design and fabricate ECM-mimicking scaffolds suitable for directing in vitro cell/scaffold interaction, and controlling tissue morphogenesis in vivo. Among these strategies, emerging computer aided design and manufacturing processes, such as modular tissue unit patterning, promise to provide unprecedented control over the generation of biologically and biomechanically competent tissue analogues. This review discusses recent studies and highlights the role of scaffold microstructural properties and their drug release capability in cell fate control and tissue morphogenesis. Furthermore, the work highlights recent advances in the bottom-up fabrication of porous scaffolds and hybrid constructs through the computer-aided assembly of cell-free and/or cell-laden micro-modules. The advantages, current limitations, and future challenges of these strategies are described and discussed.
Collapse
|
20
|
Lv Z, Hu T, Bian Y, Wang G, Wu Z, Li H, Liu X, Yang S, Tan C, Liang R, Weng X. A MgFe-LDH Nanosheet-Incorporated Smart Thermo-Responsive Hydrogel with Controllable Growth Factor Releasing Capability for Bone Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2206545. [PMID: 36426823 DOI: 10.1002/adma.202206545] [Citation(s) in RCA: 100] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/27/2022] [Indexed: 06/16/2023]
Abstract
Although growth factor (GF)-loaded hydrogels have been explored as promising materials in repairing bone defects, it still remains challenging to construct smart hydrogels with excellent gelation/mechanical properties as well as controllable GF releasing capability. Herein, the incorporation of bone morphogenetic protein 2 (BMP-2)-functionalized MgFe-layered double hydroxide (LDH) nanosheets into chitosan/silk fibroin (CS) hydrogels loaded with platelet-derived growth factor-BB (PDGF-BB) to construct a smart injectable thermo-responsive hydrogel (denoted as CSP-LB), which can achieve a burst release of PDGF-BB and a sustained release of BMP-2, for highly efficient bone regeneration is reported. The incorporation of MgFe-LDH in CS hydrogel not only shortens the gelation time and decreases sol-gel transition temperature, but also enhances the mechanical property of the hydrogel. Because of the sequential release of dual-GFs and sustained release of bioactive Mg2+ /Fe3+ ions, the in vitro experiments prove that the CSP-LB hydrogel exhibits excellent angiogenic and osteogenic properties compared with the CS hydrogel. In vivo experiments further prove that the CSP-LB hydrogel can significantly enhance bone regeneration with higher bone volume and mineral density than that of the CS hydrogel. This smart thermo-sensitive CSP-LB hydrogel possesses excellent gelation capability and angiogenic and osteogenic properties, thus providing a promising minimally invasive solution for bone defect treatment.
Collapse
Affiliation(s)
- Zehui Lv
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, P. R. China
| | - Tingting Hu
- State Key Laboratory of Chemical Resource Engineering, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Yixin Bian
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, P. R. China
| | - Guanyun Wang
- State Key Laboratory of Chemical Resource Engineering, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Zhikang Wu
- Institute of Advanced Materials (IAM) and Key Laboratory of Flexible Electronics (KLoFE), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, P. R. China
| | - Hai Li
- Institute of Advanced Materials (IAM) and Key Laboratory of Flexible Electronics (KLoFE), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, P. R. China
| | - Xueyan Liu
- State Key Laboratory of Chemical Resource Engineering, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Shuqing Yang
- State Key Laboratory of Chemical Resource Engineering, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Chaoliang Tan
- Department of Chemistry and Center of Super-Diamond and Advanced Films (COSDAF), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, 999077, Hong Kong
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, 518057, P. R. China
| | - Ruizheng Liang
- State Key Laboratory of Chemical Resource Engineering, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Xisheng Weng
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, P. R. China
| |
Collapse
|
21
|
Chen M, Chen Y, Wei C. Nanoparticles based composite coatings with tunable vascular endothelial growth factor and bone morphogenetic protein-2 release for bone regeneration. J Biomed Mater Res A 2022; 111:1044-1053. [PMID: 36565172 DOI: 10.1002/jbm.a.37489] [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: 07/13/2022] [Revised: 12/06/2022] [Accepted: 12/14/2022] [Indexed: 12/25/2022]
Abstract
Bone healing is a complex cascade involving precisely coordinated spatiotemporal presentation of multiple growth factors (GFs), including osteogenic and angiogenic GFs, and each stage of bone healing requires varying types and content of GFs. In this study, we fabricated a composite nanocoating with tunable vascular endothelial growth factor (VEGF) and bone morphogenetic protein-2 (BMP-2) that was coated on the surface of a polydopamine (PDA)-decorated tertiary calcium phosphate (TCP) scaffold using VEGF-loaded chitosan/bovine serum albumin nanoparticles (CS/BSA-NPs) and BMP-2-loaded poly-L-lysine/oxidized alginate nanoparticles (PLL/OALG-NPs). It was found that VEGF could be efficiently released to promote vascularization in early bone repair stages due to the rapid biodegradation of CS/BSA-NPs, while bone formation can be promoted by a sustained release of BMP-2 from the slowly degrading PLL/OALG-NPs. The composite coating and TCP scaffold can be conjugated due to the excellent adhesive property of PDA. The composite coating can achieve the rapid release of VEGF and sustained release of BMP-2, which can activate GFs for accelerating bone healing.
Collapse
Affiliation(s)
- Mingcong Chen
- Department of Orthopaedics and Traumatology, Shenzhen University General Hospital, Shenzhen, China
| | - Yang Chen
- Department of Surgery, First People's Hospital of Foshan, Foshan, China
| | - Cheng Wei
- Department of Orthopaedics and Traumatology, Shenzhen University General Hospital, Shenzhen, China
| |
Collapse
|
22
|
Cao Y, Tan J, Zhao H, Deng T, Hu Y, Zeng J, Li J, Cheng Y, Tang J, Hu Z, Hu K, Xu B, Wang Z, Wu Y, Lobie PE, Ma S. Bead-jet printing enabled sparse mesenchymal stem cell patterning augments skeletal muscle and hair follicle regeneration. Nat Commun 2022; 13:7463. [PMID: 36460667 PMCID: PMC9718784 DOI: 10.1038/s41467-022-35183-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 11/22/2022] [Indexed: 12/05/2022] Open
Abstract
Transplantation of mesenchymal stem cells (MSCs) holds promise to repair severe traumatic injuries. However, current transplantation practices limit the potential of this technique, either by losing the viable MSCs or reducing the performance of resident MSCs. Herein, we design a "bead-jet" printer, specialized for high-throughput intra-operative formulation and printing of MSCs-laden Matrigel beads. We show that high-density encapsulation of MSCs in Matrigel beads is able to augment MSC function, increasing MSC proliferation, migration, and extracellular vesicle production, compared with low-density bead or high-density bulk encapsulation of the equivalent number of MSCs. We find that the high-density MSCs-laden beads in sparse patterns demonstrate significantly improved therapeutic performance, by regenerating skeletal muscles approaching native-like cell density with reduced fibrosis, and regenerating skin with hair follicle growth and increased dermis thickness. MSC proliferation within 1-week post-transplantation and differentiation at 3 - 4 weeks post-transplantation are suggested to contribute therapy augmentation. We expect this "bead-jet" printing system to strengthen the potential of MSC transplantation.
Collapse
Affiliation(s)
- Yuanxiong Cao
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Jiayi Tan
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Haoran Zhao
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Ting Deng
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Yunxia Hu
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Junhong Zeng
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Jiawei Li
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Yifan Cheng
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Jiyuan Tang
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Zhiwei Hu
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Keer Hu
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Bing Xu
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
- Shenzhen Bay Laboratory, 518055, Shenzhen, China
| | - Zitian Wang
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Yaojiong Wu
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
| | - Peter E Lobie
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China
- Shenzhen Bay Laboratory, 518055, Shenzhen, China
| | - Shaohua Ma
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, 518055, Shenzhen, China.
- Tsinghua-Berkeley Shenzhen Institute (TBSI), 518055, Shenzhen, China.
- Shenzhen Bay Laboratory, 518055, Shenzhen, China.
- Institute for Brain and Cognitive Sciences, Tsinghua University, 100084, Beijing, China.
| |
Collapse
|
23
|
A composite, off-the-shelf osteoinductive material for large, vascularized bone flap prefabrication. Acta Biomater 2022; 154:641-649. [PMID: 36261107 DOI: 10.1016/j.actbio.2022.10.023] [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: 06/08/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 12/14/2022]
Abstract
We previously described an immortalized, genetically-engineered human Mesenchymal stromal cell line to generate BMP2-enriched Chondrogenic Matrices (MB-CM), which after devitalization and storage could efficiently induce ectopic bone tissue by endochondral ossification. In order to increase the efficiency of MB-CM utilization towards engineering scaled-up bone structures, here we hypothesized that MB-CM can retain osteoinductive properties when combined with an osteoconductive material. We first tested different volumetric ratios of MB-CM:SmartBone® (as clinically used, osteoconductive reference material) and assessed the bone formation capacity of the resulting composites following ectopic mouse implantation. After 8 weeks, as little as 25% of MB-CM was sufficient to induce bone formation and fusion across SmartBone® granules, generating large interconnected bony structures. The same composite percentage was then further assessed in a scaled-up model, namely within an axially-vascularized, confined, ectopically prefabricated flap (0.8 cm3) in rats. The material efficiently induced the formation of new bone (31% of the cross-sectional area after 8 weeks), including bone marrow and vascular elements, throughout the flap volume. Our findings outline a strategy for efficient use of MB-CM as part of a composite material, thereby reducing the amount required to fill large spaces and enabling utilization in critically sized grafts, to address challenging clinical scenarios in bone reconstruction. STATEMENT OF SIGNIFICANCE: Most bone repair strategies rely either on osteconductive properties of ceramics and devitalized bone, or osteoinductive properties of growth factors and extracellular matrices (ECM). Here we designed a composite material made of a clinically accepted osteoconductive material and an off-the-shelf tissue engineered human cartilage ECM with strong osteoinductive properties. We showed that low amount of osteoinductive ECM potentiated host cells recruitment to form large vascularized bone structures in two different animal models, one being a challenging prefabricated bone-flap model targeting challenging clinical bone repair. Overall, this study highlights the use of a promising human off-the-shelf material for accelerated healing towards clinical applications.
Collapse
|
24
|
Lin S, Yin S, Shi J, Yang G, Wen X, Zhang W, Zhou M, Jiang X. Orchestration of energy metabolism and osteogenesis by Mg2+ facilitates low-dose BMP-2-driven regeneration. Bioact Mater 2022; 18:116-127. [PMID: 35387176 PMCID: PMC8961427 DOI: 10.1016/j.bioactmat.2022.03.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/26/2022] [Accepted: 03/13/2022] [Indexed: 12/03/2022] Open
Abstract
The clinical application of bone morphogenetic protein-2 (BMP-2) is limited by several factors, including ineffectiveness at low doses and severe adverse effects at high doses. To address these efficacy and safety limitations, we explored whether orchestration of energy metabolism and osteogenesis by magnesium ion (Mg2+) could reduce the dose and thereby improve the safety of BMP-2. Our results demonstrated that rapid metabolic activation triggered by BMP-2 was indispensable for subsequent osteogenesis. Moreover, inadequate metabolic stimulation was shown to be responsible for the ineffectiveness of low-dose BMP-2. Next, we identified that Mg2+, as an ''energy propellant", substantially increased cellular bioenergetic levels to support the osteogenesis via the Akt-glycolysis-Mrs2-mitochondrial axis, and consequently enhanced the osteoinductivity of BMP-2. Based on the mechanistic discovery, microgel composite hydrogels were fabricated as low-dose BMP-2/Mg2+ codelivery system through microfluidic and 3D printing technologies. An in vivo study further confirmed that rapid and robust bone regeneration was induced by the codelivery system. Collectively, these results suggest that this bioenergetic-driven, cost-effective, low-dose BMP-2-based strategy has substantial potential for bone repair. BMP-2 triggered rapid metabolic adaption, characterized by the successive activation of glycolysis and OxPhos. Inadequate activation of metabolic state led to the ineffectiveness of low-dose BMP-2. Mg2+ elevated the bioenergetic levels and enhance the efficacy of BMP-2 via the Akt-glycolysis-Mrs2-mitochondrial axis. Composite hydrogels with BMP-2 and "energy propellant" Mg2+ were fabricated to orchestrate metabolism and osteogenesis. The hydrogels achieved efficient low-dose BMP-2-driven regeneration in vivo.
Collapse
|
25
|
A dual-responsive polydopamine modified hydroxybutyl chitosan hydrogel for sequential regulation of bone regeneration. Carbohydr Polym 2022; 297:120027. [DOI: 10.1016/j.carbpol.2022.120027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 11/30/2022]
|
26
|
Teal CJ, Hettiaratchi MH, Ho MT, Ortin-Martinez A, Ganesh AN, Pickering AJ, Golinski AW, Hackel BJ, Wallace VA, Shoichet MS. Directed Evolution Enables Simultaneous Controlled Release of Multiple Therapeutic Proteins from Biopolymer-Based Hydrogels. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2202612. [PMID: 35790035 DOI: 10.1002/adma.202202612] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/28/2022] [Indexed: 06/15/2023]
Abstract
With the advent of increasingly complex combination strategies of biologics, independent control over their delivery is the key to their efficacy; however, current approaches are hindered by the limited independent tunability of their release rates. To overcome these limitations, directed evolution is used to engineer highly specific, low affinity affibody binding partners to multiple therapeutic proteins to independently control protein release rates. As a proof-of-concept, specific affibody binding partners for two proteins with broad therapeutic utility: insulin-like growth factor-1 (IGF-1) and pigment epithelium-derived factor (PEDF) are identified. Protein-affibody binding interactions specific to these target proteins with equilibrium dissociation constants (KD ) between 10-7 and 10-8 m are discovered. The affibodies are covalently bound to the backbone of crosslinked hydrogels using click chemistry, enabling sustained, independent, and simultaneous release of bioactive IGF-1 and PEDF over 7 days. The system is tested with C57BL/6J mice in vivo, and the affibody-controlled release of IGF-1 results in sustained activity when compared to bolus IGF-1 delivery. This work demonstrates a new, broadly applicable approach to tune the release of therapeutic proteins simultaneously and independently and thus the way for precise control over the delivery of multicomponent therapies is paved.
Collapse
Affiliation(s)
- Carter J Teal
- Institute of Biomedical Engineering, 164 College Street, Toronto, ON, M5S 3G9, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - Marian H Hettiaratchi
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada
| | - Margaret T Ho
- Institute of Biomedical Engineering, 164 College Street, Toronto, ON, M5S 3G9, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - Arturo Ortin-Martinez
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, 399 Bathurst Street, Toronto, ON, M5T 2S8, Canada
| | - Ahil N Ganesh
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada
| | - Andrew J Pickering
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada
| | - Alex W Golinski
- Department of Chemical Engineering and Materials Science, University of Minnesota-Twin Cities, 421 Washington Avenue Southeast, 356 Amundson Hall, Minneapolis, MN, 55455, USA
| | - Benjamin J Hackel
- Department of Chemical Engineering and Materials Science, University of Minnesota-Twin Cities, 421 Washington Avenue Southeast, 356 Amundson Hall, Minneapolis, MN, 55455, USA
| | - Valerie A Wallace
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, 399 Bathurst Street, Toronto, ON, M5T 2S8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 27 King's College Circle, Toronto, ON, M5S 1A1, Canada
- Department of Ophthalmology and Vision Sciences, University of Toronto, 340 College Street, Toronto, ON, M5T 3A9, Canada
| | - Molly S Shoichet
- Institute of Biomedical Engineering, 164 College Street, Toronto, ON, M5S 3G9, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada
- Department of Ophthalmology and Vision Sciences, University of Toronto, 340 College Street, Toronto, ON, M5T 3A9, Canada
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, ON, M5S 3H6, Canada
| |
Collapse
|
27
|
Famos F, Avilla-Royo E, Vonzun L, Ochsenbein-Kölble N, Ehrbar M. Miniaturized bioengineered models for preterm fetal membrane healing. Fetal Diagn Ther 2022; 49:235-244. [PMID: 35709687 DOI: 10.1159/000525559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/26/2022] [Indexed: 11/19/2022]
Abstract
INTRODUCTION The reason for the absence of fetal membrane (FM) healing after a fetoscopic intervention is not known. We hypothesize that the lack of robust miniaturized models to study preterm FM functions is currently hampering the development of new treatments for FM healing. Specifically, miniaturized models to study preterm FM healing with minimal amounts of tissue are currently lacking. METHODS In this study, we collected FMs from planned cesarean deliveries and developed different ex vivo models with an engineered biomaterial to study FM healing. Then, the effect of PDGF-BB on the migration of cells from preterm and term FMs was evaluated. RESULTS FMs could be viably cultured ex vivo for 14 days. In a model of punctured FMs, migration of cells into FM defects was less pronounced than migration out of the tissue into the biomaterial. In a miniaturized model of preterm cell migration, PDGF-BB promoted migration of preterm amnion cells into the biomaterial. DISCUSSION AND CONCLUSION By using a novel miniaturized model of preterm tissue, we here successfully demonstrate that PDGF-BB can promote preterm FM cell migration of microtissues encapsulated in a three-dimensional environment.
Collapse
Affiliation(s)
- Flurina Famos
- Department of Obstetrics, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Eva Avilla-Royo
- Department of Obstetrics, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Ladina Vonzun
- Department of Obstetrics, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- The Zurich Center for Fetal Diagnosis and Therapy, Zurich, Switzerland
| | - Nicole Ochsenbein-Kölble
- Department of Obstetrics, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- The Zurich Center for Fetal Diagnosis and Therapy, Zurich, Switzerland
| | - Martin Ehrbar
- Department of Obstetrics, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- The Zurich Center for Fetal Diagnosis and Therapy, Zurich, Switzerland
| |
Collapse
|
28
|
Ji C, Qiu M, Ruan H, Li C, Cheng L, Wang J, Li C, Qi J, Cui W, Deng L. Transcriptome Analysis Revealed the Symbiosis Niche of 3D Scaffolds to Accelerate Bone Defect Healing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105194. [PMID: 35040587 PMCID: PMC8922091 DOI: 10.1002/advs.202105194] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/17/2021] [Indexed: 05/04/2023]
Abstract
Three dimension (3D) printed scaffolds have been shown to be superior in promoting tissue repair, but the cell-level specific regulatory network activated by 3D printing scaffolds with different material components to form a symbiosis niche have not been systematically revealed. Here, three typical 3D printed scaffolds, including natural polymer hydrogel (gelatin-methacryloyl, GelMA), synthetic polymer material (polycaprolactone, PCL), and bioceramic (β-tricalcium phosphate, β-TCP), are fabricated to explore the regulating effect of the symbiotic microenvironment during bone healing. Enrichment analysis show that hydrogel promotes tissue regeneration and reconstruction by improving blood vessel generation by enhancing oxygen transport and red blood cell development. The PCL scaffold regulates cell proliferation and differentiation by promoting cellular senescence, cell cycle and deoxyribonucleic acid (DNA) replication pathways, accelerating the process of endochondral ossification, and the formation of callus. The β-TCP scaffold can specifically enhance the expression of osteoclast differentiation and extracellular space pathway genes to promote the differentiation of osteoclasts and promote the process of bone remodeling. In these processes, specific biomaterial properties can be used to guide cell behavior and regulate molecular network in the symbiotic microenvironment to reduce the barriers of regeneration and repair.
Collapse
Affiliation(s)
- Ce Ji
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Minglong Qiu
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Huitong Ruan
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Cuidi Li
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Liang Cheng
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Juan Wang
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Changwei Li
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Jin Qi
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Wenguo Cui
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Lianfu Deng
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| |
Collapse
|
29
|
Recombinant Proteins-Based Strategies in Bone Tissue Engineering. Biomolecules 2021; 12:biom12010003. [PMID: 35053152 PMCID: PMC8773742 DOI: 10.3390/biom12010003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/08/2021] [Accepted: 12/11/2021] [Indexed: 11/29/2022] Open
Abstract
The increase in fracture rates and/or problems associated with missing bones due to accidents or various pathologies generates socio-health problems with a very high impact. Tissue engineering aims to offer some kind of strategy to promote the repair of damaged tissue or its restoration as close as possible to the original tissue. Among the alternatives proposed by this specialty, the development of scaffolds obtained from recombinant proteins is of special importance. Furthermore, science and technology have advanced to obtain recombinant chimera’s proteins. This review aims to offer a synthetic description of the latest and most outstanding advances made with these types of scaffolds, particularly emphasizing the main recombinant proteins that can be used to construct scaffolds in their own right, i.e., not only to impregnate them, but also to make scaffolds from their complex structure, with the purpose of being considered in bone regenerative medicine in the near future.
Collapse
|
30
|
Sun T, Meng C, Ding Q, Yu K, Zhang X, Zhang W, Tian W, Zhang Q, Guo X, Wu B, Xiong Z. In situ bone regeneration with sequential delivery of aptamer and BMP2 from an ECM-based scaffold fabricated by cryogenic free-form extrusion. Bioact Mater 2021; 6:4163-4175. [PMID: 33997500 PMCID: PMC8099605 DOI: 10.1016/j.bioactmat.2021.04.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 12/11/2022] Open
Abstract
In situ tissue engineering is a powerful strategy for the treatment of bone defects. It could overcome the limitations of traditional bone tissue engineering, which typically involves extensive cell expansion steps, low cell survival rates upon transplantation, and a risk of immuno-rejection. Here, a porous scaffold polycaprolactone (PCL)/decellularized small intestine submucosa (SIS) was fabricated via cryogenic free-form extrusion, followed by surface modification with aptamer and PlGF-2123-144*-fused BMP2 (pBMP2). The two bioactive molecules were delivered sequentially. The aptamer Apt19s, which exhibited binding affinity to bone marrow-derived mesenchymal stem cells (BMSCs), was quickly released, facilitating the mobilization and recruitment of host BMSCs. BMP2 fused with a PlGF-2123-144 peptide, which showed "super-affinity" to the ECM matrix, was released in a slow and sustained manner, inducing BMSC osteogenic differentiation. In vitro results showed that the sequential release of PCL/SIS-pBMP2-Apt19s promoted cell migration, proliferation, alkaline phosphatase activity, and mRNA expression of osteogenesis-related genes. The in vivo results demonstrated that the sequential release system of PCL/SIS-pBMP2-Apt19s evidently increased bone formation in rat calvarial critical-sized defects compared to the sequential release system of PCL/SIS-BMP2-Apt19s. Thus, the novel delivery system shows potential as an ideal alternative for achieving cell-free scaffold-based bone regeneration in situ.
Collapse
Key Words
- 3D, three-dimensional
- Apt19s, aptamer 19s
- Aptamer
- BMD, bone mineral density
- BMP2
- BMP2, bone morphogenic protein 2
- BMSC, bone marrow-derived mesenchymal stem cell
- Bone regeneration in situ
- CLSM, confocal laser scanning microscopy
- CSD, critical-sized calvarial defect
- Cell recruitment
- Controlled delivery
- ECM, decellularized matrix
- FBS, fetal bovine serum
- FDA, US Food and Drug Administration
- FITC, fluorescein isothiocyanate
- FTIR, Fourier transform infrared
- H&E, hematoxylin and eosin
- HA, hydroxyapatite
- PCL, polycaprolactone
- PVDF, polyvinylidene difluoride
- Rh6G, rhodamine 6G
- SIS, small intestine submucosa
- pBMP2, PlGF-2123-144*-fused BMP2
- ssDNA, single-stranded DNA
Collapse
Affiliation(s)
- Tingfang Sun
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chunqing Meng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiuyue Ding
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Keda Yu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xianglin Zhang
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Wancheng Zhang
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Wenqing Tian
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Qi Zhang
- Wuhan Hi-tech Medical Tissue Research Center, Wuhan, 430206, China
| | - Xiaodong Guo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bin Wu
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zekang Xiong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
31
|
Luminescent hydrogels with tunable emission colors and excellent adhesion performance fabricated by lanthanide complexes induced crosslinking and physical interaction. POLYMER 2021. [DOI: 10.1016/j.polymer.2021.124319] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
32
|
Hao L, Tianyuan Z, Zhen Y, Fuyang C, Jiang W, Zineng Y, Zhengang D, Shuyun L, Chunxiang H, Zhiguo Y, Quanyi G. Biofabrication of cell-free dual drug-releasing biomimetic scaffolds for meniscal regeneration. Biofabrication 2021; 14. [PMID: 34610586 DOI: 10.1088/1758-5090/ac2cd7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 10/05/2021] [Indexed: 01/26/2023]
Abstract
Regenerating the meniscus remains challenging because of its avascular, aneural, and alymphatic nature. Three-dimensional (3D) printing technology provides a promising strategy to fabricate biomimetic meniscal scaffolds with an anisotropic architecture, a proper biomechanical microenvironment, and bioactive components. Herein, 3D printing technology is adopted by coencapsulating chemokines (platelet-derived growth factor-BB, PDGF-BB) and small chondroinductive molecules (kartogenin, KGN) within biomimetic polycaprolactone/hydrogel composite scaffolds. The incorporated PDGF-BB is expected to promote endogenous stem cell homing, and KGN in poly(lactic-co-glycolic) acid microspheres is employed to target the chondrogenesis of resident mesenchymal stem cells (MSCs). First, we chose basic bioinks composed of gelatin methacrylamide and hyaluronic acid methacrylate and then incorporated four concentrations (0%, 0.5%, 1.0%, and 2.0%) of meniscal extracellular matrix into the bioink to systematically study the superiority of these combinations and identify the optimally printable bioink. Next, we investigated the scaffold morphology and drug release profile. The effects of releasing the drugs in a sequentially controlled manner from the composite scaffolds on the fate of MSCs were also evaluated. The biofabricated scaffolds, with and without dual drug loading, were further studied in a rabbit model established with a critical-size medial meniscectomy. We found that meniscal scaffolds containing both drugs had combinational advantages in enhancing cell migration and synergistically promoted MSC chondrogenic differentiation. The dual drug-loaded scaffolds also significantly promotedin vivoneomeniscal regeneration three and six months after implantation in terms of histological and immunological phenotypes. The results presented herein reveal that this 3D-printed dual drug-releasing meniscal scaffold possesses the potential to act as an off-the-shelf product for the clinical treatment of meniscal injury and related joint degenerative diseases.
Collapse
Affiliation(s)
- Li Hao
- School of Medicine, Nankai University, Tianjin 300071, People's Republic of China.,Institute of Orthopedics, First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, People's Republic of China
| | - Zhao Tianyuan
- School of Medicine, Nankai University, Tianjin 300071, People's Republic of China.,Institute of Orthopedics, First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, People's Republic of China
| | - Yang Zhen
- School of Medicine, Nankai University, Tianjin 300071, People's Republic of China.,Institute of Orthopedics, First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, People's Republic of China.,Arthritis Clinic and Research Center, Peking University People's Hospital, Peking University, Beijing 100044, People's Republic of China
| | - Cao Fuyang
- Institute of Orthopedics, First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, People's Republic of China.,Department of Orthopedics, First Affiliated Hospital of Zhengzhou University, 1 Jian East Road, Eqi District, Zhengzhou 450052, People's Republic of China
| | - Wu Jiang
- Institute of Orthopedics, First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, People's Republic of China
| | - Yan Zineng
- Institute of Orthopedics, First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, People's Republic of China
| | - Ding Zhengang
- Institute of Orthopedics, First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, People's Republic of China
| | - Liu Shuyun
- School of Medicine, Nankai University, Tianjin 300071, People's Republic of China.,Institute of Orthopedics, First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, People's Republic of China
| | - Hao Chunxiang
- Institute of Anesthesia, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, People's Republic of China
| | - Yuan Zhiguo
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200030, People's Republic of China
| | - Guo Quanyi
- School of Medicine, Nankai University, Tianjin 300071, People's Republic of China.,Institute of Orthopedics, First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, People's Republic of China
| |
Collapse
|
33
|
Liu X, Chen W, Shao B, Zhang X, Wang Y, Zhang S, Wu W. Mussel patterned with 4D biodegrading elastomer durably recruits regenerative macrophages to promote regeneration of craniofacial bone. Biomaterials 2021; 276:120998. [PMID: 34237507 DOI: 10.1016/j.biomaterials.2021.120998] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 06/19/2021] [Accepted: 06/25/2021] [Indexed: 12/11/2022]
Abstract
Crosstalk between bone marrow mesenchymal stem cells (BMSCs) and macrophages plays vital role in bone healing. By investigating the mechanism of collagen membrane-guided bone regeneration, we found compact structure and rapid membrane degradation compromised the duration of M2 macrophages influx, which restricts the recruitment of BMSCs that is essential for bone healing. To tackle this issue, a biodegrading elastomeric compound consisting of poly(glycerol sebacate) (PGS) and polycaprolactone (PCL) was fabricated into hierarchically porous membrane. The rational design of 3D microstructure enabled sufficient polydopamine (PDA) coating. Without any addition of growth factors, the 3D-patterned PDA membrane enables early and durable influx of M2 macrophages, which in turn promotes BMSCs recruitment and osteogenic differentiation. Furthermore, 4D-morphing of the membrane fully regenerates the dome shaped calvarial bone as well as arc-shape bone in peri-implant alveolar defect without filling xenogenous substitute. This study revealed the superiority of 3D printed microstructures in immunomodulatory materials. The availability of 4D-morphing for PGS/PCL construct expanded their advantages in reconstructing craniofacial bone.
Collapse
Affiliation(s)
- Xuzheng Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology Department of Oral & Maxillofacial Surgery School of Stomatology the Fourth Military Medical University, Xi'an, Shaanxi, PR China; Department of Oral Implant Center, People's Hospital of Inner Mongolia Autonomous Region, Hohhot, 010110, PR China
| | - Wanli Chen
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology Department of Oral & Maxillofacial Surgery School of Stomatology the Fourth Military Medical University, Xi'an, Shaanxi, PR China
| | - Bo Shao
- Department of Oral Implant Center, People's Hospital of Inner Mongolia Autonomous Region, Hohhot, 010110, PR China
| | - Xinchi Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology Department of Oral & Maxillofacial Surgery School of Stomatology the Fourth Military Medical University, Xi'an, Shaanxi, PR China
| | - Yinggang Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology Department of Oral & Maxillofacial Surgery School of Stomatology the Fourth Military Medical University, Xi'an, Shaanxi, PR China
| | - Siqian Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology Department of Oral & Maxillofacial Surgery School of Stomatology the Fourth Military Medical University, Xi'an, Shaanxi, PR China
| | - Wei Wu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology Department of Oral & Maxillofacial Surgery School of Stomatology the Fourth Military Medical University, Xi'an, Shaanxi, PR China.
| |
Collapse
|
34
|
Subbiah R, Ruehle MA, Klosterhoff BS, Lin AS, Hettiaratchi MH, Willett NJ, Bertassoni LE, García AJ, Guldberg RE. Triple growth factor delivery promotes functional bone regeneration following composite musculoskeletal trauma. Acta Biomater 2021; 127:180-192. [PMID: 33823326 DOI: 10.1016/j.actbio.2021.03.066] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/28/2021] [Accepted: 03/31/2021] [Indexed: 12/20/2022]
Abstract
Successful bone healing in severe trauma depends on early revascularization to restore oxygen, nutrient, growth factor, and progenitor cell supply to the injury. Therapeutic angiogenesis strategies have therefore been investigated to promote revascularization following severe bone injuries; however, results have been inconsistent. This is the first study investigating the effects of dual angiogenic growth factors (VEGF and PDGF) with low-dose bone morphogenetic protein-2 (BMP-2; 2.5 µg) on bone healing in a clinically challenging composite bone-muscle injury model. Our hydrogel-based delivery systems demonstrated a more than 90% protein entrapment efficiency and a controlled simultaneous release of three growth factors over 28 days. Co-stimulation of microvascular fragment constructs with VEGF and PDGF promoted vascular network formation in vitro compared to VEGF or PDGF alone. In an in vivo model of segmental bone and volumetric muscle loss injury, combined VEGF (5 µg) and PDGF (7.5 µg or 15 µg) delivery with a low dose of BMP-2 significantly enhanced regeneration of vascularized bone compared to BMP-2 treatment alone. Notably, the regenerated bone mechanics reached ~60% of intact bone, a value that was previously only achieved by delivery of high-dose BMP-2 (10 µg) in this injury model. Overall, sustained delivery of VEGF, PDFG, and BMP-2 is a promising strategy to promote functional vascularized bone tissue regeneration following severe composite musculoskeletal injury. Although this study is conducted in a clinically relevant composite injury model in rats using a simultaneous release strategy, future studies are necessary to test the regenerative potential of spatiotemporally controlled delivery of triple growth factors on bone healing using large animal models. STATEMENT OF SIGNIFICANCE: Volumetric muscle loss combined with delayed union or non-union bone defect causes deleterious effects on bone regeneration even with the supplementation of bone morphogenetic protein-2 (BMP-2). In this study, the controlled delivery of dual angiogenic growth factors (vascular endothelial growth factor [VEGF] + Platelet-derived growth factor [PDGF]) increases vascular growth in vitro. Co-delivering VEGF+PDGF significantly increase the bone formation efficacy of low-dose BMP-2 and improves the mechanics of regenerated bone in a challenging composite bone-muscle injury model.
Collapse
|
35
|
Han X, Sun M, Chen B, Saiding Q, Zhang J, Song H, Deng L, Wang P, Gong W, Cui W. Lotus seedpod-inspired internal vascularized 3D printed scaffold for bone tissue repair. Bioact Mater 2021; 6:1639-1652. [PMID: 33313444 PMCID: PMC7701916 DOI: 10.1016/j.bioactmat.2020.11.019] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/27/2020] [Accepted: 11/12/2020] [Indexed: 12/16/2022] Open
Abstract
In the field of bone defect repair, 3D printed scaffolds have the characteristics of personalized customization and accurate internal structure. However, how to construct a well-structured vascular network quickly and effectively inside the scaffold is essential for bone repair after transplantation. Herein, inspired by the unique biological structure of "lotus seedpod", hydrogel microspheres encapsulating deferoxamine (DFO) liposomes were prepared through microfluidic technology as "lotus seeds", and skillfully combined with a three-dimensional (3D) printed bioceramic scaffold with biomimetic "lotus" biological structure which can internally grow blood vessels. In this composite scaffold system, DFO was effectively released by 36% in the first 6 h, which was conducive to promote the growth of blood vessels inside the scaffold quickly. In the following 7 days, the release rate of DFO reached 69%, which was fundamental in the formation of blood vessels inside the scaffold as well as osteogenic differentiation of bone mesenchymal stem cells (BMSCs). It was confirmed that the composite scaffold could significantly promote the human umbilical vein endothelial cells (HUVECs) to form the vascular morphology within 6 h in vitro. In vivo, the composite scaffold increased the expression of vascularization and osteogenic related proteins Hif1-α, CD31, OPN, and OCN in the rat femoral defect model, significantly cutting down the time of bone repair. To sum up, this "lotus seedpod" inspired porous bioceramic 3D printed scaffold with internal vascularization functionality has broad application prospects in the future.
Collapse
Affiliation(s)
- Xiaoyu Han
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, Shandong, 250013, PR China
- 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 2nd Road, Shanghai, 200025, PR China
| | - Mingjie Sun
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, Shandong, 250013, PR China
| | - Bo Chen
- 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 2nd Road, Shanghai, 200025, PR China
| | - Qimanguli Saiding
- 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 2nd Road, Shanghai, 200025, PR China
| | - Junyue Zhang
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, Shandong, 250013, PR China
| | - Hongliang Song
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, Shandong, 250013, PR China
| | - Lianfu Deng
- 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 2nd Road, Shanghai, 200025, PR China
| | - Peng Wang
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, Shandong, 250013, PR China
| | - Weiming Gong
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, Shandong, 250013, PR China
| | - Wenguo Cui
- 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 2nd Road, Shanghai, 200025, PR China
| |
Collapse
|
36
|
Madduma‐Bandarage USK, Madihally SV. Synthetic hydrogels: Synthesis, novel trends, and applications. J Appl Polym Sci 2020. [DOI: 10.1002/app.50376] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
37
|
Kim S, Lee M. Rational design of hydrogels to enhance osteogenic potential. CHEMISTRY OF MATERIALS : A PUBLICATION OF THE AMERICAN CHEMICAL SOCIETY 2020; 32:9508-9530. [PMID: 33551566 PMCID: PMC7857485 DOI: 10.1021/acs.chemmater.0c03018] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Bone tissue engineering (BTE) encompasses the field of biomaterials, cells, and bioactive molecules to successfully guide the growth and repair of bone tissue. Current BTE strategies rely on delivering osteogenic molecules or cells via scaffolding materials. However, growth factor- and stem cell-based treatments have several limitations, such as source restriction, low stability, difficulties in predicting long-term efficacy, and high costs, among others. These issues have promoted the development of material-based therapy with properties of accessibility, high stability, tunable efficacy, and low-cost production. Hydrogels are widely used in BTE applications because of their unique hydrophilic nature and tunable physicochemical properties to mimic the native bone environment. However, current hydrogel materials are not ideal candidates due to minimal osteogenic capability on their own. Therefore, recent studies of BTE hydrogels attempt to counterbalance these issues by modifying their biophysical properties. In this article, we review recent progress in the design of hydrogels to instruct osteogenic potential, and present strategies developed to precisely control its bone healing properties.
Collapse
Affiliation(s)
- Soyon Kim
- Division of Advanced Prosthodontics, University of California, Los Angeles, USA
| | - Min Lee
- Division of Advanced Prosthodontics, University of California, Los Angeles, USA
- Department of Bioengineering, University of California, Los Angeles, USA
| |
Collapse
|
38
|
Thanasrisuebwong P, Kiattavorncharoen S, Surarit R, Phruksaniyom C, Ruangsawasdi N. Red and Yellow Injectable Platelet-Rich Fibrin Demonstrated Differential Effects on Periodontal Ligament Stem Cell Proliferation, Migration, and Osteogenic Differentiation. Int J Mol Sci 2020; 21:ijms21145153. [PMID: 32708242 PMCID: PMC7404021 DOI: 10.3390/ijms21145153] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/14/2020] [Accepted: 07/20/2020] [Indexed: 02/07/2023] Open
Abstract
The biological benefits of using two fractions derived from injectable platelet-rich fibrin (i-PRF) in bone regeneration remain unclear. Thus, the current study examined two fractionation protocols producing yellow i-PRF and red i-PRF on periodontal ligament stem cells (PDLSCs). The i-PRF samples from five donors were harvested from two different levels, with and without a buffy coat layer, to obtain red and yellow i-PRF, respectively. The PDLSCs were isolated and characterized before their experimental use. The culture medium in each assay was loaded with 20% of the conditioned medium containing the factors released from the red and yellow i-PRF. Cell proliferation and cell migration were determined with an MTT and trans-well assay, respectively. Osteogenic differentiation was investigated using alkaline phosphatase and Alizarin red staining. The efficiency of both i-PRFs was statistically compared. We found that the factors released from the red i-PRF had a greater effect on cell proliferation and cell migration. Moreover, the factors released from the yellow i-PRF stimulated PDLSC osteogenic differentiation earlier compared with the red i-PRF. These data suggest that the red i-PRF might be suitable for using in bone regeneration because it induced the mobilization and growth of bone regenerative cells without inducing premature mineralization.
Collapse
Affiliation(s)
- Prakan Thanasrisuebwong
- Dental Implant Center, Dental Hospital, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand;
| | - Sirichai Kiattavorncharoen
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand;
| | - Rudee Surarit
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand;
| | - Chareerut Phruksaniyom
- Department of Pharmacology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand;
| | - Nisarat Ruangsawasdi
- Department of Pharmacology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand;
- Correspondence:
| |
Collapse
|
39
|
Hua C, Liu J, Hua X, Wang X. Synergistic Fabrication of Dose-Response Chitosan/Dextran/β-Glycerophosphate Injectable Hydrogel as Cell Delivery Carrier for Cardiac Healing After Acute Myocardial Infarction. Dose Response 2020; 18:1559325820941323. [PMID: 32922226 PMCID: PMC7457666 DOI: 10.1177/1559325820941323] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/26/2020] [Accepted: 06/01/2020] [Indexed: 12/14/2022] Open
Abstract
The human mesenchymal stem cells (hMSCs) therapy offering an encouraging the new methods to establish the conveying on the chitosan (C)/dextran (D)/β-glycerophosphate (β-GP) loaded with hMSCs to enhance the acute myocardial infarctions. The synthesized hMSCs-CD@β-GP system displayed the ratio of determination modules, size of the pore, absorbency, and the swellings ratio in the assortment of the 65 ka, 149 ± 39.8 µm, 92.2%, 42 ± 1.38, and 29 ± 1.9, respectively. The fabricated hMSCs-CD@β-GP was highly stable and physicochemical investigated and confirmed the suitability of the materials for cardiac regeneration applications. The in vitro examinations of the injectable hydrogels with hMSCs-CD@β-GP have recognized that the improved survival rate of the cells, increased the pro-inflammatory expressions factors, pro-angiogenic factors analysis confirmed the promising results of the ejection of fractions, fibrosis area, vessel density with decreased infractions size, with suggesting that the remarkable improvement of the heart regenerative function after myocardial infarctions. The new synergistic approach of the injectable hydrogels with hMSCs could able appropriate for the effective treatment of cardiac therapies after acute myocardial infarctions.
Collapse
Affiliation(s)
- Chongjun Hua
- Department of Cardiology, Jinhua Central Hospital, Jinhua, China
| | - Jing Liu
- Department of Cardiology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Xiuhong Hua
- Department of Pharmacy, Jinhua Fifth Hospital, Jinhua, China
| | - Xinyu Wang
- Department of Ultrasonography, Xiamen Cardiovascular Hospital Xiamen University, Xiamen, China
| |
Collapse
|