1
|
Zhao Z, Wang P, Li Z, Wei X, Li S, Lu X, Dai S, Huang B, Man Z, Li W. Targeted lipid nanoparticles distributed in hydrogel treat osteoarthritis by modulating cholesterol metabolism and promoting endogenous cartilage regeneration. J Nanobiotechnology 2024; 22:786. [PMID: 39707367 DOI: 10.1186/s12951-024-02965-9] [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: 08/18/2024] [Accepted: 10/31/2024] [Indexed: 12/23/2024] Open
Abstract
Osteoarthritis (OA) is the most common disease in aging joints and has characteristics of cartilage destruction and inflammation. It is currently considered a metabolic disease, and the CH25H-CYP7B1-RORα axis of cholesterol metabolism in chondrocytes plays a crucial catabolic regulatory role in its pathogenesis. Targeting of this axis in chondrocytes may provide a therapeutic approach for OA treatment. Here, in this study, we propose to use a combination of stem cell-recruiting hydrogels and lipid nanoparticles (LNPs) that modulate cholesterol metabolism to jointly promote a regenerative microenvironment. Specifically, we first developed an injectable, bioactive hydrogel composed of self-assembling peptide nanofibers that recruits endogenous synovial stem cells (SMSCs) and promotes their chondrogenic differentiation. At the same time, LNPs that regulate cholesterol metabolism are incorporated into the hydrogel and slowly released, thereby improving the inflammatory environment of OA. Enhancements were noted in the inflammatory conditions associated with OA, alongside the successful attraction of mesenchymal stem cells (MSCs) from the synovial membrane. These cells were then observed to differentiate into chondrocytes, contributing to effective cartilage restoration and chondrocyte regeneration, thereby offering a promising approach for OA treatment. In summary, this approach provides a feasible siRNA-based therapeutic option, offering a potential nonsurgical solution for treatment of OA.
Collapse
Affiliation(s)
- Zhibo Zhao
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, People's Republic of China
| | - Peng Wang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, People's Republic of China
| | - Ziyang Li
- Department of Sports Medicine & Orthopedic Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, People's Republic of China
| | - Xingchen Wei
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, People's Republic of China
| | - Shishuo Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People's Republic of China
| | - Xiaoqing Lu
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People's Republic of China
| | - Shimin Dai
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People's Republic of China
| | - Benzhao Huang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People's Republic of China
| | - Zhentao Man
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, People's Republic of China.
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People's Republic of China.
- College of Sports Medicine and Rehabilitation, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250021, People's Republic of China.
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250062, People's Republic of China.
| | - Wei Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, People's Republic of China.
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People's Republic of China.
- College of Sports Medicine and Rehabilitation, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250021, People's Republic of China.
| |
Collapse
|
2
|
Yang L, Li W, Zhao Y, Wang Y, Shang L. Stem cell recruitment polypeptide hydrogel microcarriers with exosome delivery for osteoarthritis treatment. J Nanobiotechnology 2024; 22:512. [PMID: 39192268 DOI: 10.1186/s12951-024-02765-1] [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: 03/10/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
With the accelerated aging tendency, osteoarthritis (OA) has become an intractable global public health challenge. Stem cells and their derivative exosome (Exo) have shown great potential in OA treatment. Research in this area tends to develop functional microcarriers for stem cell and Exo delivery to improve the therapeutic effect. Herein, we develop a novel system of Exo-encapsulated stem cell-recruitment hydrogel microcarriers from liquid nitrogen-assisted microfluidic electrospray for OA treatment. Benefited from the advanced droplet generation capability of microfluidics and mild cryogelation procedure, the resultant particles show uniform size dispersion and excellent biocompatibility. Moreover, acryloylated stem cell recruitment peptides SKPPGTSS are directly crosslinked within the particles by ultraviolet irradiation, thus simplifying the peptide coupling process and preventing its premature release. The SKPPGTSS-modified particles can recruit endogenous stem cells to promote cartilage repair and the released Exo from the particles further enhances the cartilage repair performance through synergistic effects. These features suggest that the proposed hydrogel microcarrier delivery system is a promising candidate for OA treatment.
Collapse
Affiliation(s)
- Lei Yang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Wenzhao Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China.
| | - Yongxiang Wang
- Department of Orthopedics, Northern Jiangsu People's Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou, 225001, China.
| | - Luoran Shang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and The Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
3
|
Lu N, Li Z, Su D, Chen J, Zhao J, Gao Y, Liu Q, Liu G, Luo X, Luo R, Deng X, Zhu H, Luo Z. Design of novel chiral self-assembling peptides to explore the efficiency and mechanism of mRNA-FIPV vaccine delivery vehicles. Int J Pharm 2024; 660:124344. [PMID: 38885779 DOI: 10.1016/j.ijpharm.2024.124344] [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: 04/03/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
The enhancement of conventional liposome and lipid nanoparticle (LNP) methodologies in the formulation and deployment of messenger RNA (mRNA) vaccines necessitates further refinement to augment both their effectiveness and biosafety profiles. Additionally, researching these innovative delivery carrier materials represents both a prominent focus and a significant challenge in the current scientific landscape. Here we designed new chiral self-assembling peptides as the delivery carrier for RNA vaccines to study the underlying mechanisms in the feline infectious peritonitis virus (FIPV) model system. Firstly, we successfully transcribed mature enhanced green fluorescent protein (EGFP) mRNA and feline infectious peritonitis virus nucleocapsid (FIPV N) mRNA in vitro from optimized vectors. Subsequently, we developed chiral self-assembling peptide-1 (CSP-1) and chiral self-assembling peptide-2 (CSP-2) peptides, taking into account the physical and chemical characteristics of nucleic acid molecules as well as the principles of self-assembling peptides, with the aim of improving the delivery efficiency of mRNA molecule complexes. We determined the optimal coating ratio between CSP and mRNA by electrophoretic mobility shift assay. We found that the peptides and mRNA complexes can protect the mRNA from RNase A enzyme and efficiently deliver mRNA into cells for target antigen proteins expression. Animal experiments confirmed that CSP-1/mRNA complex can effectively trigger immune response mechanisms involving IFN-γ and T cell activation. It can also stimulate CD4+ and CD8+ T cell proliferation and induce serum antibody titers up to 10,000 times higher. And no pathological changes were observed by immunohistochemistry in liver, spleen, and kidney, indicating that CSP-1 may be a safe and promising delivery system for mRNA vaccines. Methodologically, this research represents a novel endeavor in the utilization of chiral self-assembling peptides within the realm of mRNA vaccines. This approach not only introduces fresh prospects for employing such nanomaterials in various mRNA vaccines but also expands the potential for developing small molecules, proteins, and antibodies. Furthermore, it paves the way for new clinical applications of existing pharmaceuticals.
Collapse
Affiliation(s)
- Na Lu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Zhaoxu Li
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China; Department of Materials Science and Engineering, University of California, Irvine, CA, United States
| | - Di Su
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Jialei Chen
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Jiawei Zhao
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Yu Gao
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Qichen Liu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Guicen Liu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Xinyi Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China; Department of Physiology and Biophysics, University of California, Irvine, CA, United States
| | - Ruyue Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China; Department of Medicine, Northwestern University Feinberg School of Medicine,Chicago, IL,United States
| | - Xiaoyan Deng
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Huifang Zhu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Zhongli Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
4
|
Park HS, Choi GH, Jung TW, Lee T. Scaffold-based synergistic enhancement of stem cell effects for therapeutic angiogenesis in critical limb ischemia: an experimental animal study. Ann Surg Treat Res 2024; 107:50-57. [PMID: 38978685 PMCID: PMC11227915 DOI: 10.4174/astr.2024.107.1.50] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/07/2024] [Accepted: 05/16/2024] [Indexed: 07/10/2024] Open
Abstract
Purpose Stem cell-based therapies are considered an alternative approach for critical limb ischemia (CLI) patients with limited or exhausted options, yet their clinical use is limited by the lack of sustainability and unclear mechanism of action. In this study, a substance P-conjugated scaffold was injected with mesenchymal stem cells (MSCs) into an animal model of CLI to verify whether angiogenesis could be enhanced. Methods A self-assembling peptide (SAP) was conjugated with substance P, known to have the ability to recruit host stem cells into the site of action. This SAP was injected with MSCs into ischemic hindlimbs of rats, and the presence of MSCs was verified by immunohistochemical (IHC) staining of MSC-specific markers at days 7, 14, and 28. The degree of angiogenesis, cell apoptosis, and fibrosis was also quantified. Results Substance P-conjugated SAP was able to recruit intrinsic MSCs into the ischemic site of action. When injected in combination with MSCs, the presence of both injected and recruited MSCs was found in the ischemic tissues by double IHC staining. This in turn led to a higher degree of angiogenesis, less cell apoptosis, and less tissue fibrosis compared to the other groups at all time points. Conclusion The combination of substance P-conjugated SAP and MSCs was able to enhance angiogenesis and tissue repair, which was achieved by the additive effect from exogenously administered and intrinsically recruited MSCs. This scaffold-based intrinsic recruitment approach could be a viable option to enhance the therapeutic effects in patients with CLI.
Collapse
Affiliation(s)
- Hyung Sub Park
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Geum Hee Choi
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Tae Woo Jung
- Department of Pharmacology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Taeseung Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| |
Collapse
|
5
|
Zhou H, Zhu Y, Yang B, Huo Y, Yin Y, Jiang X, Ji W. Stimuli-responsive peptide hydrogels for biomedical applications. J Mater Chem B 2024; 12:1748-1774. [PMID: 38305498 DOI: 10.1039/d3tb02610h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Stimuli-responsive hydrogels can respond to external stimuli with a change in the network structure and thus have potential application in drug release, intelligent sensing, and scaffold construction. Peptides possess robust supramolecular self-assembly ability, enabling spontaneous formation of nanostructures through supramolecular interactions and subsequently hydrogels. Therefore, peptide-based stimuli-responsive hydrogels have been widely explored as smart soft materials for biomedical applications in the last decade. Herein, we present a review article on design strategies and research progress of peptide hydrogels as stimuli-responsive materials in the field of biomedicine. The latest design and development of peptide hydrogels with responsive behaviors to stimuli are first presented. The following part provides a systematic overview of the functions and applications of stimuli-responsive peptide hydrogels in tissue engineering, drug delivery, wound healing, antimicrobial treatment, 3D cell culture, biosensors, etc. Finally, the remaining challenges and future prospects of stimuli-responsive peptide hydrogels are proposed. It is believed that this review will contribute to the rational design and development of stimuli-responsive peptide hydrogels toward biomedical applications.
Collapse
Affiliation(s)
- Haoran Zhou
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, P. R. China.
| | - Yanhua Zhu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, P. R. China.
| | - Bingbing Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, P. R. China.
| | - Yehong Huo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, P. R. China.
| | - Yuanyuan Yin
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing 401147, P. R. China
| | - Xuemei Jiang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, P. R. China.
| | - Wei Ji
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, P. R. China.
| |
Collapse
|
6
|
Zhao J, Wu S, Zhang M, Hong X, Zhao M, Xu S, Ji J, Ren K, Fu G, Fu J. Adventitial delivery of miR-145 to treat intimal hyperplasia post vascular injuries through injectable and in-situ self-assembling peptide hydrogels. Acta Biomater 2024; 173:247-260. [PMID: 37939818 DOI: 10.1016/j.actbio.2023.10.039] [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: 06/26/2023] [Revised: 10/07/2023] [Accepted: 10/31/2023] [Indexed: 11/10/2023]
Abstract
Intimal hyperplasia is a common lesion that can be observed in diverse vascular diseases. Drug-eluting stents and drug-coated balloons, which can release anti-proliferative agents to inhibit smooth muscle cell (SMC) proliferation, are developed to prevent intimal hyperplasia. However, these intervention devices still cannot achieve satisfactory clinical outcomes. In contrast to endovascular drug delivery, vascular adventitial drug delivery is a new strategy. To develop a vascular adventitial drug delivery system to treat intimal hyperplasia post vascular injuries, we loaded miR-145-5p-agomir (miR-145) into an injectable and in-situ self-assembling RAD peptide hydrogel. In vitro data showed that the miR-145 could be well incorporated into the RAD peptide hydrogels and released in a slow and controlled manner. The released miR-145 could transfect SMCs successfully, and the transfected SMCs exhibited a reduced migration capacity and higher expressions of SMC contractile biomarkers as compared to the non-transfected SMCs. In vivo data showed that the retention of the miR-145 was greatly elongated by the RAD peptide hydrogels. In addition, the application of the miR-145-loaded RAD peptide hydrogels surrounding injured arteries decreased the proliferative SMCs, promoted the regeneration of endothelium, reduced the macrophage infiltration, inhibited the neointimal formation and prevented adverse ECM remodeling via downregulation of KLF4 expression. The RAD peptide hydrogels loaded with miR-145 can successfully inhibit intimal hyperplasia after vascular injuries and thus hold great potential as an innovative extravascular drug delivery approach to treat vascular diseases. STATEMENT OF SIGNIFICANCE: Intimal hyperplasia is a common lesion that can be observed in diverse vascular diseases. Drug-eluting stents and drug-coated balloons, which can release anti-proliferative agents to inhibit smooth muscle cell (SMC) proliferation, are developed to prevent intimal hyperplasia. However, these intervention devices still cannot achieve satisfactory clinical outcomes. In contrast to endovascular drug delivery, vascular adventitial drug delivery is a new strategy. Our work here demonstrates that the RAD peptide hydrogels loaded with miR-145-5p-agomir (miR-145) can successfully reverse intimal hyperplasia after vascular injuries and thus hold great potential as an innovative vascular adventitial drug delivery approach to treat vascular diseases. Our work proposes a possible paradigm shift from endovascular drug delivery to extravascular drug delivery for vascular disorder treatment.
Collapse
Affiliation(s)
- Jing Zhao
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China; MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Shaofei Wu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China
| | - Mingqi Zhang
- Guangxi University of Chinese Medicine, Nanning 530001, China
| | - Xulin Hong
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China
| | - Meng Zhao
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China
| | - Shihui Xu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China
| | - Jian Ji
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Kefeng Ren
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Guosheng Fu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China.
| | - Jiayin Fu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China.
| |
Collapse
|
7
|
Zhu G, Zhou Y, Xu Y, Wang L, Han M, Xi K, Tang J, Li Z, Kou Y, Zhou X, Feng Y, Gu Y, Chen L. Functionalized acellular periosteum guides stem cell homing to promote bone defect repair. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2023; 34:2000-2020. [PMID: 37071056 DOI: 10.1080/09205063.2023.2204779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/17/2023] [Indexed: 04/19/2023]
Abstract
The periosteum plays a key role in bone tissue regeneration, especially in the promotion and protection of new bones. However, among the bone repair materials, many biomimetic artificial periosteum lack the natural periosteal structure, stem cells, and immunoregulation required for bone regeneration. In this study, we used natural periosteum to produce acellular periosteum. To retain the appropriate cell survival structure and immunomodulatory proteins, we grafted the functional polypeptide SKP on the surface collagen of the periosteum via an amide bond, providing the acellular periosteum with the ability to recruit mesenchymal stem cells. Thus, we developed a biomimetic periosteum (DP-SKP) with the ability to promote stem cell homing and immunoregulation in vivo. Compared to the blank and simple decellularized periosteum groups, DP-SKP was more conducive to stem cell adhesion, growth, and osteogenic differentiation in vitro. Additionally, compared with the other two groups, DP-SKP significantly promoted mesenchymal stem cell homing to the periosteal transplantation site, improved the bone immune microenvironment, and accelerated new lamellar bone formation in the critical size defect of rabbit skulls in vivo. Therefore, this acellular periosteum with a mesenchymal stem cell homing effect is expected to be used as an extracellular artificial periosteum in clinical practice.
Collapse
Affiliation(s)
- Guoqing Zhu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Orthopedics, Suzhou Municipal Hospital, Suzhou, China
| | - Yidi Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yichang Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lingjun Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng Han
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Orthopedics, Xuzhou Central Hospital, Xuzhou, China
| | - Kun Xi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jincheng Tang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ziang Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yu Kou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xindie Zhou
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Yu Feng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yong Gu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Liang Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
8
|
Ligorio C, Mata A. Synthetic extracellular matrices with function-encoding peptides. NATURE REVIEWS BIOENGINEERING 2023; 1:1-19. [PMID: 37359773 PMCID: PMC10127181 DOI: 10.1038/s44222-023-00055-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/16/2023] [Indexed: 06/28/2023]
Abstract
The communication of cells with their surroundings is mostly encoded in the epitopes of structural and signalling proteins present in the extracellular matrix (ECM). These peptide epitopes can be incorporated in biomaterials to serve as function-encoding molecules to modulate cell-cell and cell-ECM interactions. In this Review, we discuss natural and synthetic peptide epitopes as molecular tools to bioengineer bioactive hydrogel materials. We present a library of functional peptide sequences that selectively communicate with cells and the ECM to coordinate biological processes, including epitopes that directly signal to cells, that bind ECM components that subsequently signal to cells, and that regulate ECM turnover. We highlight how these epitopes can be incorporated in different biomaterials as individual or multiple signals, working synergistically or additively. This molecular toolbox can be applied in the design of biomaterials aimed at regulating or controlling cellular and tissue function, repair and regeneration.
Collapse
Affiliation(s)
- Cosimo Ligorio
- Biodiscovery Institute, University of Nottingham, Nottingham, UK
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, UK
| | - Alvaro Mata
- Biodiscovery Institute, University of Nottingham, Nottingham, UK
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, UK
- School of Pharmacy, University of Nottingham, Nottingham, UK
| |
Collapse
|
9
|
Abdal Dayem A, Lee SB, Lim KM, Kim A, Shin HJ, Vellingiri B, Kim YB, Cho SG. Bioactive peptides for boosting stem cell culture platform: Methods and applications. Biomed Pharmacother 2023; 160:114376. [PMID: 36764131 DOI: 10.1016/j.biopha.2023.114376] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Peptides, short protein fragments, can emulate the functions of their full-length native counterparts. Peptides are considered potent recombinant protein alternatives due to their specificity, high stability, low production cost, and ability to be easily tailored and immobilized. Stem cell proliferation and differentiation processes are orchestrated by an intricate interaction between numerous growth factors and proteins and their target receptors and ligands. Various growth factors, functional proteins, and cellular matrix-derived peptides efficiently enhance stem cell adhesion, proliferation, and directed differentiation. For that, peptides can be immobilized on a culture plate or conjugated to scaffolds, such as hydrogels or synthetic matrices. In this review, we assess the applications of a variety of peptides in stem cell adhesion, culture, organoid assembly, proliferation, and differentiation, describing the shortcomings of recombinant proteins and their full-length counterparts. Furthermore, we discuss the challenges of peptide applications in stem cell culture and materials design, as well as provide a brief outlook on future directions to advance peptide applications in boosting stem cell quality and scalability for clinical applications in tissue regeneration.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Soo Bin Lee
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Kyung Min Lim
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Aram Kim
- Department of Urology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Hyun Jin Shin
- Department of Ophthalmology, Research Institute of Medical Science, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda 151401, Punjab, India
| | - Young Bong Kim
- Department of Biomedical Science & Engineering, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
10
|
Qin S, Zhu J, Zhang G, Sui Q, Niu Y, Ye W, Ma G, Liu H. Research progress of functional motifs based on growth factors in cartilage tissue engineering: A review. Front Bioeng Biotechnol 2023; 11:1127949. [PMID: 36824354 PMCID: PMC9941568 DOI: 10.3389/fbioe.2023.1127949] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 01/20/2023] [Indexed: 02/10/2023] Open
Abstract
Osteoarthritis is a chronic degenerative joint disease that exerts significant impacts on personal life quality, and cartilage tissue engineering is a practical treatment in clinical. Various growth factors are involved in cartilage regeneration and play important roles therein, which is the focus of current cartilage repair strategy. To compensate for the purification difficulty, high cost, poor metabolic stability, and circulating dilution of natural growth factors, the concept of functional motifs (also known as mimetic peptides) from original growth factor was introduced in recent studies. Here, we reviewed the selection mechanisms, biological functions, carrier scaffolds, and modification methods of growth factor-related functional motifs, and evaluated the repair performance in cartilage tissue engineering. Finally, the prospects of functional motifs in researches and clinical application were discussed.
Collapse
Affiliation(s)
- Shengao Qin
- School of Stomatology, Dalian Medical University, Dalian, China,Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian, China
| | - Jiaman Zhu
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China,Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Guangyong Zhang
- School of Stomatology, Dalian Medical University, Dalian, China,Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian, China
| | - Qijia Sui
- School of Stomatology, Dalian Medical University, Dalian, China,Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian, China
| | - Yimeng Niu
- School of Stomatology, Dalian Medical University, Dalian, China,Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian, China
| | - Weilong Ye
- School of Stomatology, Dalian Medical University, Dalian, China,Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China,*Correspondence: Weilong Ye, ; Guowu Ma, ; Huiying Liu,
| | - Guowu Ma
- School of Stomatology, Dalian Medical University, Dalian, China,Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian, China,*Correspondence: Weilong Ye, ; Guowu Ma, ; Huiying Liu,
| | - Huiying Liu
- School of Stomatology, Dalian Medical University, Dalian, China,Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian, China,*Correspondence: Weilong Ye, ; Guowu Ma, ; Huiying Liu,
| |
Collapse
|
11
|
Research progress of stem cell therapy for endometrial injury. Mater Today Bio 2022; 16:100389. [PMID: 36033375 PMCID: PMC9403503 DOI: 10.1016/j.mtbio.2022.100389] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 11/24/2022] Open
Abstract
Endometrial damage is an important factor leading to infertility and traditional conventional treatments have limited efficacy. As an emerging technology in recent years, stem cell therapy has provided new hope for the treatment of this disease. By comparing the advantages of stem cells from different sources, it is believed that menstrual blood endometrial stem cells have a good application prospect as a new source of stem cells. However, the clinical utility of stem cells is still limited by issues such as colonization rates, long-term efficacy, tumor formation, and storage and transportation. This paper summarizes the mechanism by which stem cells repair endometrial damage and clarifies the material basis of their effects from four aspects: replacement of damaged sites, paracrine effects, interaction with growth factors, and other new targets. According to the pathological characteristics and treatment requirements of intrauterine adhesion (IUA), the research work to solve the above problems from the aspects of functional bioscaffold preparation and multi-functional platform construction is also summarized. From the perspective of scaffold materials and component functions, this review will provide a reference for comprehensively optimizing the clinical application of stem cells.
Collapse
|
12
|
Guo W, Ma Y, Hu L, Feng Y, Liu Y, Yi X, Zhang W, Tang F. Modification Strategies for Ionic Complementary Self-Assembling Peptides: Taking RADA16-I as an Example. Polymers (Basel) 2022; 14:polym14235221. [PMID: 36501615 PMCID: PMC9739689 DOI: 10.3390/polym14235221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 12/04/2022] Open
Abstract
Ion-complementary self-assembling peptides have been studied in many fields for their distinct advantages, mainly due to their self-assembly properties. However, their shortcomings, such as insufficient specific activity and poor mechanical properties, also limited their application. For the better and wider application of these promising biomaterials, ion-complementary self-assembling peptides can be modified with their self-assembly properties not being destroyed to the greatest extent. The modification strategies were reviewed by taking RADA16-I as an example. For insufficient specific activity, RADA16-I can be structurally modified with active motifs derived from the active domain of the extracellular matrix or other related active factors. For weak mechanical properties, materials with strong mechanical properties or that can undergo chemical crosslinking were used to mix with RADA16-I to enhance the mechanical properties of RADA16-I. To improve the performance of RADA16-I as drug carriers, appropriate adjustment of the RADA16-I sequence and/or modification of the RADA16-I-related delivery system with polymer materials or specific molecules can be considered to achieve sustained and controlled release of specific drugs or active factors. The modification strategies reviewed in this paper may provide some references for further basic research and clinical application of ion-complementary self-assembling peptides and their derivatives.
Collapse
Affiliation(s)
- Weiwei Guo
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563006, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China
- The Key Laboratory of Clinical Pharmacy of Zuni City, Zunyi Medical University, Zunyi 563006, China
| | - Yinping Ma
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563006, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China
- The Key Laboratory of Clinical Pharmacy of Zuni City, Zunyi Medical University, Zunyi 563006, China
| | - Lei Hu
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563006, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China
- The Key Laboratory of Clinical Pharmacy of Zuni City, Zunyi Medical University, Zunyi 563006, China
| | - Yujie Feng
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563006, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China
- The Key Laboratory of Clinical Pharmacy of Zuni City, Zunyi Medical University, Zunyi 563006, China
| | - Yanmiao Liu
- The Key Laboratory of Clinical Pharmacy of Zuni City, Zunyi Medical University, Zunyi 563006, China
- School of Preclinical Medicine, Zunyi Medical University, Zunyi 563006, China
| | - Xuedong Yi
- Department of Pharmacy, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Wenzhi Zhang
- Department of Pharmacy, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Fushan Tang
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563006, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China
- The Key Laboratory of Clinical Pharmacy of Zuni City, Zunyi Medical University, Zunyi 563006, China
- Correspondence: or ; Tel.: +86-851-28642337
| |
Collapse
|
13
|
Shen X, Qu F, Pei Y, Lei S, Xia S, Liang J, Li S, Sun X, Liu L. Repairing sciatic nerve injury with self-assembling peptide nanofiber scaffold-containing chitosan conduit. Front Neurol 2022; 13:867711. [PMID: 36313506 PMCID: PMC9612716 DOI: 10.3389/fneur.2022.867711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Background An increasing number of nerve guide scaffolds have been used to replace the “gold-standard” autologous nerve graft for repairing peripheral nerve defects, but nerve regeneration is usually far from complete. Methods Here, we designed and prepared two functionalized self-assembling peptides (SAP) with the IKVAV and KLT sequences, which were derived from the combination of laminin and VEGF, respectively. Their mixtures were also obtained to combine the effects of neuroprotective and neurotrophic and proangiogenic factors. Results The beneficial effect of peptide gels on nerve regeneration was evaluated in vitro using Schwann cells (SCs). As a useful intraluminal filling, a three-dimensional (3D) functionalized self-assembling peptide (SAP) nanofiber hydrogel was formed in the hollow lumen of chitosan conduits under physiological conditions. In vivo, the combination of the two functionalized SAP gels containing a chitosan nerve conduit significantly accelerated nerve healing and enhanced morphological repair. Conclusion Based on the current findings, the combined application of two functionalized SAP gels with chitosan nerve conduit is a promising therapy for the engineering of peripheral nerve regeneration.
Collapse
Affiliation(s)
- Xuezhen Shen
- Department of Orthopedics, Beijing Luhe Hospital, Affiliated to Capital Medical University, Beijing, China
| | - Feng Qu
- Foot and Ankle Surgery Center, Beijing Tongren Hospital, Affiliated to Capital Medical University, Beijing, China
| | - Yilun Pei
- Department of Orthopedics, Beijing Luhe Hospital, Affiliated to Capital Medical University, Beijing, China
| | - Simeng Lei
- Department of Orthopedics, Beijing Luhe Hospital, Affiliated to Capital Medical University, Beijing, China
| | - Suhang Xia
- Department of Orthopedics, Beijing Luhe Hospital, Affiliated to Capital Medical University, Beijing, China
| | - Jing Liang
- Department of Pediatrics, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Shan Li
- Department of Pediatrics, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Xun Sun
- Department of Orthopedics, Tianjin Hospital, Tianjin University, Tianjin, China
- *Correspondence: Xun Sun
| | - Liang Liu
- Department of Orthopedics, Beijing Luhe Hospital, Affiliated to Capital Medical University, Beijing, China
- Liang Liu
| |
Collapse
|
14
|
Wang Y, Xu L, Zhao J, Liang J, Zhang Z, Li Q, Zhang J, Wan P, Wu Z. Reconstructing auto tissue engineering lamellar cornea with aspartic acid modified acellular porcine corneal stroma and preconditioned limbal stem cell for corneal regeneration. Biomaterials 2022; 289:121745. [DOI: 10.1016/j.biomaterials.2022.121745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 07/31/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022]
|
15
|
Ye W, Yang Z, Cao F, Li H, Zhao T, Zhang H, Zhang Z, Yang S, Zhu J, Liu Z, Zheng J, Liu H, Ma G, Guo Q, Wang X. Articular cartilage reconstruction with TGF-β1-simulating self-assembling peptide hydrogel-based composite scaffold. Acta Biomater 2022; 146:94-106. [PMID: 35552000 DOI: 10.1016/j.actbio.2022.05.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/14/2022] [Accepted: 05/05/2022] [Indexed: 12/15/2022]
Abstract
Transforming growth factor-β (TGF-β) is an important inducing factor for the differentiation of mesenchymal stem cells and the secretion of collagen II, but the inaccessibility and instability limit its application in clinical practice. In this study, the TGF-β1-simulating peptide LIANAK (CM) was connected with the self-assembling peptide Ac-(RADA)4-CONH2 (RAD) to obtain the functionalized self-assembling peptide Ac-(RADA)4-GG-LIANAK-CONH2 (RAD-CM). The results indicated that the CM-functionalized RAD hydrogel contributed to the enhanced expressions of chondrogenic genes and extracellular matrix deposition. The self-assembling peptides were then combined with decellularized cartilage extracellular matrix (DCM) to construct a composite scaffold for articular cartilage repair. The CM-functionalized composite scaffold RAD/RAD-CM/DCM (R/C/D) exhibited good bioactivity and structural stability and exhibited satisfactory performance in promoting neocartilage restoration and the reconstruction of the osteochondral unit. This study provides a promising strategy for in situ cartilage regeneration via the stable presentation of TGF-β1-simulating peptide. STATEMENT OF SIGNIFICANCE: Deficiency of effective chondrogenic inducers (especially, the TGF-β family) significantly limits the self-regeneration of cartilage in osteochondral defect cases. Oligopeptide LIANAK, named CM, could simulate TGF-β1's bioactivity with particular structure, but traditional chemical crosslinking to polymer scaffolds resulted in risks of safety and complication, which is unfavorable for clinical applications. Here, self-assembling peptide RAD was used to load CM, to obtain a TGF-β1 mimetic peptide hydrogel. Depending on the homology (amino acids) of RAD and CM, the synthesis of the whole peptide only needs simply extended sequences of CM following that of RAD by automated solid-phase peptide synthesis. The modified peptide effectively demonstrated osteochondrogenic bioactivity, ensured the convenience, safety, and mass production, which displayed great potential in tissue engineering research and translational medicine.
Collapse
Affiliation(s)
- Weilong Ye
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, No.1 Qinghuayuan Road, Beijing 100084, China; Department of Prosthodontics, School of Stomatology, Dalian Medical University, No.9 west section, Lvshunnan Road, Dalian 116044, China
| | - Zhen Yang
- Institute of Orthopedics, The 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, China
| | - Fuyang Cao
- Institute of Orthopedics, The 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, China; Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, 1 Jian East Road, Eqi District, Zhengzhou 450052, China
| | - Hao Li
- Institute of Orthopedics, The 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, China
| | - Tianyuan Zhao
- Institute of Orthopedics, The 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, China
| | - Huan Zhang
- Department of Prosthodontics, School of Stomatology, Dalian Medical University, No.9 west section, Lvshunnan Road, Dalian 116044, China
| | - Zhe Zhang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, No.1 Qinghuayuan Road, Beijing 100084, China
| | - Shuhui Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, No.1 Qinghuayuan Road, Beijing 100084, China
| | - Jinjin Zhu
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, No.1 Qinghuayuan Road, Beijing 100084, China
| | - Zhu Liu
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, No.1 Qinghuayuan Road, Beijing 100084, China; Department of Prosthodontics, School of Stomatology, Dalian Medical University, No.9 west section, Lvshunnan Road, Dalian 116044, China
| | - Jingchuan Zheng
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, No.1 Qinghuayuan Road, Beijing 100084, China; Department of Prosthodontics, School of Stomatology, Dalian Medical University, No.9 west section, Lvshunnan Road, Dalian 116044, China
| | - Huiying Liu
- Department of Prosthodontics, School of Stomatology, Dalian Medical University, No.9 west section, Lvshunnan Road, Dalian 116044, China
| | - Guowu Ma
- Department of Prosthodontics, School of Stomatology, Dalian Medical University, No.9 west section, Lvshunnan Road, Dalian 116044, China.
| | - Quanyi Guo
- Institute of Orthopedics, The 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, China.
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, No.1 Qinghuayuan Road, Beijing 100084, China.
| |
Collapse
|
16
|
Mu X, Liu H, Yang S, Li Y, Xiang L, Hu M, Wang X. Chitosan Tubes Inoculated with Dental Pulp Stem Cells and Stem Cell Factor Enhance Facial Nerve-Vascularized Regeneration in Rabbits. ACS OMEGA 2022; 7:18509-18520. [PMID: 35694480 PMCID: PMC9178771 DOI: 10.1021/acsomega.2c01176] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 05/16/2022] [Indexed: 06/15/2023]
Abstract
Facial nerve injury is a common clinical condition that leads to disfigurement and emotional distress in the affected individuals, and the recovery presents clinical challenges. Tissue engineering is the standard method to repair nerve defects. However, nerve regeneration is still not satisfactory because of poor neovascularization after implantation, especially for the long-segment nerve defects. In the current study, we aimed to investigate the potential of chitosan tubes inoculated with stem cell factor (SCF) and dental pulp stem cells (DPSCs) in facial nerve-vascularized regeneration. In the in vitro experiment, DPSCs were isolated, cultured, and then identified. The optimal concentration of SCF was screened by CCK8. Cytoskeleton and living-cell staining, migration, CCK8 test, and neural differentiation assays were performed, revealing that SCF promoted the biological activity of DPSCs. Surprisingly, SCF increased the neural differentiation of DPSCs. The migration and angiogenesis experiments were carried out to show that SCF promoted the angiogenesis and migration of human umbilical vein endothelial cells (HUVECs). In the facial nerve, 7 mm defects of New Zealand white rabbits, hematoxylin-eosin (HE), immunohistochemistry, toluidine blue staining, and transmission electron microscopy observation were performed at 12 weeks postsurgery to show more nerve fibers and better myelin sheath in the SCF + DPSC group. In addition, the whisker movements, Masson's staining, and western blot assays were performed, demonstrating functional repair and that the expression level of CD31 protein in the group SCF + DPSCs was relatively close to that in the group Autograft. In summary, chitosan tubes inoculated with SCF and DPSCs increased neurovascularization and provided an effective method for repairing facial nerve defects, indicating great promise for clinical application.
Collapse
Affiliation(s)
- Xiaodan Mu
- Department
of Stomotology, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Huawei Liu
- Department
of Stomotology, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Shuhui Yang
- Department
of Materials Science and Engineering, State Key Laboratory of New
Ceramics and Fine Processing, Tsinghua University, Beijing 100084, China
| | - Yongfeng Li
- Department
of Stomotology, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Lei Xiang
- Department
of Stomotology, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Min Hu
- Department
of Stomotology, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiumei Wang
- Department
of Materials Science and Engineering, State Key Laboratory of New
Ceramics and Fine Processing, Tsinghua University, Beijing 100084, China
| |
Collapse
|
17
|
Li J, Zhang Y, Zhou X, Wang S, Hao R, Han J, Li M, Zhao Y, Chen C, Xu H. Enzymatically functionalized RGD-gelatin scaffolds that recruit host mesenchymal stem cells in vivo and promote bone regeneration. J Colloid Interface Sci 2022; 612:377-391. [PMID: 34998197 DOI: 10.1016/j.jcis.2021.12.091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/20/2021] [Accepted: 12/14/2021] [Indexed: 01/27/2023]
Abstract
Critical-size bone defects are imposing a substantial biomedical burden. Despite being long regarded as a potential approach to mitigate this burden or an alternative to bone grafts, bone tissue engineering (BTE) has virtually not proceeded to widespread clinical practices. In the BTE field, it is highly required to find a facile method to prepare active scaffolds with tailored biological functions. Here, we immobilized cell adhesive RGD motifs onto gelatin sponge (GS) scaffolds through enzymatic linking. On the basis of the resulting RGD-functionalized GS (RGD/GS) scaffolds, we developed a new and convenient strategy for bone defect repair, in which the scaffolds were first used to recruit mesenchymal stem cells (MSCs) from skeletal muscle, immediately followed by their engraftment into bone defect. We demonstrated significantly enhanced host cells homing into RGD/GS scaffolds as a result of specific RGD-integrin interactions, and the recruited host cells showed a strong osteogenic differentiation potential. After ectopic implantation of cell-laden RGD/GS scaffolds into critical-size mouse bone defects, marked bone tissue regeneration occurred. The presented strategy not only provides an agile route for the preparation of bioactive scaffolds and the construction of osteoinductive bone-graft substitutes, but also avoids or minimizes the complicated and laborious cell isolation, in vitro expansion and cell seeding procedures used in the conventional BTE.
Collapse
Affiliation(s)
- Junling Li
- Department of Biological and Energy Chemical Engineering, College of Chemical Engineering, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China; Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key Lab for Biotech-Drugs of National Health Commission, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingdao Road, Ji'nan 250117, China
| | - Yan Zhang
- Department of Biological and Energy Chemical Engineering, College of Chemical Engineering, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Xing Zhou
- Qingdao West Coast New Area Marine Development Bureau, 59 Shuilingshan Road, Qingdao 266400, China
| | - Shili Wang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key Lab for Biotech-Drugs of National Health Commission, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingdao Road, Ji'nan 250117, China
| | - Ruirui Hao
- Department of Biological and Energy Chemical Engineering, College of Chemical Engineering, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Jinxiang Han
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key Lab for Biotech-Drugs of National Health Commission, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingdao Road, Ji'nan 250117, China
| | - Mian Li
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key Lab for Biotech-Drugs of National Health Commission, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingdao Road, Ji'nan 250117, China
| | - Yurong Zhao
- Department of Biological and Energy Chemical Engineering, College of Chemical Engineering, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Cuixia Chen
- Department of Biological and Energy Chemical Engineering, College of Chemical Engineering, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China.
| | - Hai Xu
- Department of Biological and Energy Chemical Engineering, College of Chemical Engineering, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China.
| |
Collapse
|
18
|
Hao Z, Li H, Wang Y, Hu Y, Chen T, Zhang S, Guo X, Cai L, Li J. Supramolecular Peptide Nanofiber Hydrogels for Bone Tissue Engineering: From Multihierarchical Fabrications to Comprehensive Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103820. [PMID: 35128831 PMCID: PMC9008438 DOI: 10.1002/advs.202103820] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/02/2022] [Indexed: 05/03/2023]
Abstract
Bone tissue engineering is becoming an ideal strategy to replace autologous bone grafts for surgical bone repair, but the multihierarchical complexity of natural bone is still difficult to emulate due to the lack of suitable biomaterials. Supramolecular peptide nanofiber hydrogels (SPNHs) are emerging biomaterials because of their inherent biocompatibility, satisfied biodegradability, high purity, facile functionalization, and tunable mechanical properties. This review initially focuses on the multihierarchical fabrications by SPNHs to emulate natural bony extracellular matrix. Structurally, supramolecular peptides based on distinctive building blocks can assemble into nanofiber hydrogels, which can be used as nanomorphology-mimetic scaffolds for tissue engineering. Biochemically, bioactive motifs and bioactive factors can be covalently tethered or physically absorbed to SPNHs to endow various functions depending on physiological and pharmacological requirements. Mechanically, four strategies are summarized to optimize the biophysical microenvironment of SPNHs for bone regeneration. Furthermore, comprehensive applications about SPNHs for bone tissue engineering are reviewed. The biomaterials can be directly used in the form of injectable hydrogels or composite nanoscaffolds, or they can be used to construct engineered bone grafts by bioprinting or bioreactors. Finally, continuing challenges and outlook are discussed.
Collapse
Affiliation(s)
- Zhuowen Hao
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Hanke Li
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Yi Wang
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Yingkun Hu
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Tianhong Chen
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Shuwei Zhang
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Xiaodong Guo
- Department of OrthopedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyJiefang Road 1277Wuhan430022China
| | - Lin Cai
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Jingfeng Li
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| |
Collapse
|
19
|
Zhu J, Yang S, Qi Y, Gong Z, Zhang H, Liang K, Shen P, Huang YY, Zhang Z, Ye W, Yue L, Fan S, Shen S, Mikos AG, Wang X, Fang X. Stem cell-homing hydrogel-based miR-29b-5p delivery promotes cartilage regeneration by suppressing senescence in an osteoarthritis rat model. SCIENCE ADVANCES 2022; 8:eabk0011. [PMID: 35353555 PMCID: PMC8967232 DOI: 10.1126/sciadv.abk0011] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Osteoarthritis (OA) is a common joint disease characterized by progressive loss of cartilage and reduction in lubricating synovial fluid, which lacks effective treatments currently. Here, we propose a hydrogel-based miRNA delivery strategy to rejuvenate impaired cartilage by creating a regenerative microenvironment to mitigate chondrocyte senescence that mainly contributes to cartilage breakdown during OA development. An aging-related miRNA, miR-29b-5p, was first found to be markedly down-regulated in OA cartilage, and their up-regulation suppressed the expression of matrix metalloproteinases and senescence-associated genes (P16INK4a/P21) via ten-eleven-translocation enzyme 1 (TET1). An injectable bioactive self-assembling peptide nanofiber hydrogel was applied to deliver agomir-29b-5p, which was functionalized by conjugating a stem cell-homing peptide SKPPGTSS for endogenous synovial stem cell recruitment simultaneously. Sustained miR-29b-5p delivery and recruitment of synovial stem cells and their subsequent differentiation into chondrocytes led to successful cartilage repair and chondrocyte rejuvenation. This strategy enables miRNA-based therapeutic modality to become a viable alternative for surgery in OA treatment.
Collapse
Affiliation(s)
- Jinjin Zhu
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang, Hangzhou 310016, China
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Shuhui Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Yadong Qi
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Zhe Gong
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang, Hangzhou 310016, China
| | - Haitao Zhang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang, Hangzhou 310016, China
| | - Kaiyu Liang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang, Hangzhou 310016, China
| | - Panyang Shen
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang, Hangzhou 310016, China
| | - Yin-Yuan Huang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Zhe Zhang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Weilong Ye
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Lei Yue
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Shunwu Fan
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang, Hangzhou 310016, China
| | - Shuying Shen
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang, Hangzhou 310016, China
| | - Antonios G. Mikos
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, TX 77005, USA
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
- Corresponding author. (X.F.); (X.W.)
| | - Xiangqian Fang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang, Hangzhou 310016, China
- Corresponding author. (X.F.); (X.W.)
| |
Collapse
|
20
|
Han L, Wang Z, Chen H, Li J, Zhang S, Zhang S, Shao S, Zhang Y, Shen C, Tao H. Sa12b-Modified Functional Self-Assembling Peptide Hydrogel Enhances the Biological Activity of Nucleus Pulposus Mesenchymal Stem Cells by Inhibiting Acid-Sensing Ion Channels. Front Cell Dev Biol 2022; 10:822501. [PMID: 35252187 PMCID: PMC8888415 DOI: 10.3389/fcell.2022.822501] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/26/2022] [Indexed: 01/08/2023] Open
Abstract
Various hydrogels have been studied for nucleus pulposus regeneration. However, they failed to overcome the changes in the acidic environment during intervertebral disc degeneration. Therefore, a new functionalized peptide RAD/SA1 was designed by conjugating Sa12b, an inhibitor of acid-sensing ion channels, onto the C-terminus of RADA16-I. Then, the material characteristics and biocompatibility of RAD/SA1, and the bioactivities and mechanisms of degenerated human nucleus pulposus mesenchymal stem cells (hNPMSCs) were evaluated. Atomic force microscopy (AFM) and scanning electron microscopy (SEM) confirmed that RAD/SA1 self-assembling into three-dimensional (3D) nanofiber hydrogel scaffolds under acidic conditions. Analysis of the hNPMSCs cultured in the 3D scaffolds revealed that both RADA16-I and RAD/SA1 exhibited reliable attachment and extremely low cytotoxicity, which were verified by SEM and cytotoxicity assays, respectively. The results also showed that RAD/SA1 increased the proliferation of hNPMSCs compared to that in culture plates and pure RADA16-I. Quantitative reverse transcription polymerase chain reaction, enzyme-linked immunosorbent assay, and western blotting demonstrated that the expression of collagen I was downregulated, while collagen II, aggrecan, and SOX-9 were upregulated. Furthermore, Ca2+ concentration measurement and western blotting showed that RAD/SA1 inhibited the expression of p-ERK through Ca2+-dependent p-ERK signaling pathways. Therefore, the functional self-assembling peptide nanofiber hydrogel designed with the short motif of Sa12b could be used as an excellent scaffold for nucleus pulposus tissue engineering. Moreover, RAD/SA1 exhibits great potential applications in the regeneration of mildly degenerated nucleus pulposus.
Collapse
Affiliation(s)
- Letian Han
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ziyu Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Haoyu Chen
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jie Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shengquan Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Sumei Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Shanzhong Shao
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yinshun Zhang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Cailiang Shen
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hui Tao
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
21
|
Bioinspired Sandcastle Worm-Derived Peptide-Based Hybrid Hydrogel for Promoting the Formation of Liver Spheroids. Gels 2022; 8:gels8030149. [PMID: 35323262 PMCID: PMC8950079 DOI: 10.3390/gels8030149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 12/10/2022] Open
Abstract
The generation of hepatic spheroids is beneficial for a variety of potential applications, including drug development, disease modeling, transplantation, and regenerative medicine. Natural hydrogels are obtained from tissues and have been widely used to promote the growth, differentiation, and retention of specific functionalities of hepatocytes. However, relying on natural hydrogels for the generation of hepatic spheroids, which have batch to batch variations, may in turn limit the previously mentioned potential applications. For this reason, we researched a way to establish a three-dimensional (3D) culture system that more closely mimics the interaction between hepatocytes and their surrounding microenvironments, thereby potentially offering a more promising and suitable system for drug development, disease modeling, transplantation, and regenerative medicine. Here, we developed self-assembling and bioactive hybrid hydrogels to support the generation and growth of hepatic spheroids. Our hybrid hydrogels (PC4/Cultrex) inspired by the sandcastle worm, an Arg-Gly-Asp (RGD) cell adhesion sequence, and bioactive molecules derived from Cultrex BME (Basement Membrane Extract). By performing optimizations to the design, the PC4/Cultrex hybrid hydrogels can enhance HepG2 cells to form spheroids and express their molecular signatures (e.g., Cyp3A4, Cyp7a1, A1at, Afp, Ck7, Ck1, and E-cad). Our study demonstrated that this hybrid hydrogel system offers potential advantages for hepatocytes in proliferating, differentiating, and self-organizing to form hepatic spheroids in a more controllable and reproducible manner. In addition, it is a versatile and cost-effective method for 3D tissue cultures in mass quantities. Importantly, we demonstrate that it is feasible to adapt a bioinspired approach to design biomaterials for 3D culture systems, which accelerates the design of novel peptide structures and broadens our research choices on peptide-based hydrogels.
Collapse
|
22
|
Kim NH, Choi H, Shahzad ZM, Ki H, Lee J, Chae H, Kim YH. Supramolecular assembly of protein building blocks: from folding to function. NANO CONVERGENCE 2022; 9:4. [PMID: 35024976 PMCID: PMC8755899 DOI: 10.1186/s40580-021-00294-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/03/2021] [Indexed: 06/14/2023]
Abstract
Several phenomena occurring throughout the life of living things start and end with proteins. Various proteins form one complex structure to control detailed reactions. In contrast, one protein forms various structures and implements other biological phenomena depending on the situation. The basic principle that forms these hierarchical structures is protein self-assembly. A single building block is sufficient to create homogeneous structures with complex shapes, such as rings, filaments, or containers. These assemblies are widely used in biology as they enable multivalent binding, ultra-sensitive regulation, and compartmentalization. Moreover, with advances in the computational design of protein folding and protein-protein interfaces, considerable progress has recently been made in the de novo design of protein assemblies. Our review presents a description of the components of supramolecular protein assembly and their application in understanding biological phenomena to therapeutics.
Collapse
Affiliation(s)
- Nam Hyeong Kim
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hojae Choi
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Zafar Muhammad Shahzad
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, 16419, Republic of Korea
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Heesoo Ki
- Department of Nano Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jaekyoung Lee
- Department of Nano Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Heeyeop Chae
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yong Ho Kim
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, 16419, Republic of Korea.
- Department of Nano Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, 16419, Republic of Korea.
| |
Collapse
|
23
|
An injectable self-assembling hydrogel based on RGD peptidomimetic β-sheets as multifunctional biomaterials. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 133:112633. [PMID: 35527136 DOI: 10.1016/j.msec.2021.112633] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/09/2021] [Accepted: 12/20/2021] [Indexed: 12/23/2022]
Abstract
Ability of the cells to adhere to an extracellular material is central to successful tissue genesis. Arg-Gly-Asp (RGD) sequences found in extracellular matrix proteins are well known for cell adhesion, however, enzymatic degradation and lack of specificity have limited their widespread use. Besides, a multifunctional material with inherent antimicrobial ability would help in invigorating the practical tissue engineering applications. Here, we report novel modified RGD (MR) and RGD mimic [R(K)] peptides (MOH and MNH2) which were synthesized post-in-silico screening, based on their interactions with integrin protein αVβ3 using HEX 8.0 docking server. These mimics, containing hydrophobic Phe-Phe (FF) moiety which has been specifically introduced to initiate the self-assembling process of β-sheet structures, were characterized thoroughly using various physicochemical and spectroscopic techniques. Under physiological conditions, these mimetics displayed thixotropic behavior rendering them highly suitable as injectable hydrogels having an added advantage of site-specific targeting abilities. Electron microscopy further revealed the formation of nanofibers upon self-assembly of these peptides. Besides, enhanced cell adhesiveness by these peptides compared to the commercial Poly l-lysine coated surfaces as well as the inherent antimicrobial potential against both sensitive and antibiotic-resistant pathogens (Methicillin-resistant Staphylococcus aureus and multi-drug resistant Salmonella enteritidis) substantiated the applicability of these unique injectable hydrogels wherein the porous fibrous framework offered a favorable environment for drug entrapment and 3D cell culture. Altogether, these properties render these novel RGD mimic peptides as promising multifunctional candidates for various tissue regenerative applications.
Collapse
|
24
|
Das S, Das D. Rational Design of Peptide-based Smart Hydrogels for Therapeutic Applications. Front Chem 2021; 9:770102. [PMID: 34869218 PMCID: PMC8635208 DOI: 10.3389/fchem.2021.770102] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/22/2021] [Indexed: 12/12/2022] Open
Abstract
Peptide-based hydrogels have captivated remarkable attention in recent times and serve as an excellent platform for biomedical applications owing to the impressive amalgamation of unique properties such as biocompatibility, biodegradability, easily tunable hydrophilicity/hydrophobicity, modular incorporation of stimuli sensitivity and other functionalities, adjustable mechanical stiffness/rigidity and close mimicry to biological molecules. Putting all these on the same plate offers smart soft materials that can be used for tissue engineering, drug delivery, 3D bioprinting, wound healing to name a few. A plethora of work has been accomplished and a significant progress has been realized using these peptide-based platforms. However, designing hydrogelators with the desired functionalities and their self-assembled nanostructures is still highly serendipitous in nature and thus a roadmap providing guidelines toward designing and preparing these soft-materials and applying them for a desired goal is a pressing need of the hour. This review aims to provide a concise outline for that purpose and the design principles of peptide-based hydrogels along with their potential for biomedical applications are discussed with the help of selected recent reports.
Collapse
Affiliation(s)
- Saurav Das
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati, India
| | - Debapratim Das
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati, India
| |
Collapse
|
25
|
Zhao X, Li Q, Guo Z, Li Z. Constructing a cell microenvironment with biomaterial scaffolds for stem cell therapy. Stem Cell Res Ther 2021; 12:583. [PMID: 34809719 PMCID: PMC8607654 DOI: 10.1186/s13287-021-02650-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/03/2021] [Indexed: 01/08/2023] Open
Abstract
Stem cell therapy is widely recognized as a promising strategy for exerting therapeutic effects after injury in degenerative diseases. However, limitations such as low cell retention and survival rates after transplantation exist in clinical applications. In recent years, emerging biomaterials that provide a supportable cellular microenvironment for transplanted cells have optimized the therapeutic efficacy of stem cells in injured tissues or organs. Advances in the engineered microenvironment are revolutionizing our understanding of stem cell-based therapies by co-transplanting with synthetic and tissue-derived biomaterials, which offer a scaffold for stem cells and propose an unprecedented opportunity to further employ significant influences in tissue repair and regeneration.
Collapse
Affiliation(s)
- Xiaotong Zhao
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, 453003, Henan, China.,Department of Cardiology, Zhengzhou Seventh People's Hospital, Zhengzhou, China
| | - Qiong Li
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, 453003, Henan, China
| | - Zhikun Guo
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, 453003, Henan, China. .,Department of Cardiology, Zhengzhou Seventh People's Hospital, Zhengzhou, China.
| | - Zongjin Li
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, 453003, Henan, China. .,Nankai University School of Medicine, 94 Weijin Road, Tianjin, 300071, China.
| |
Collapse
|
26
|
Yang S, Zhu J, Lu C, Chai Y, Cao Z, Lu J, Zhang Z, Zhao H, Huang YY, Yao S, Kong X, Zhang P, Wang X. Aligned fibrin/functionalized self-assembling peptide interpenetrating nanofiber hydrogel presenting multi-cues promotes peripheral nerve functional recovery. Bioact Mater 2021; 8:529-544. [PMID: 34541418 PMCID: PMC8435993 DOI: 10.1016/j.bioactmat.2021.05.056] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 05/07/2021] [Accepted: 05/28/2021] [Indexed: 12/19/2022] Open
Abstract
Nerve guidance conduits with hollow lumen fail to regenerate critical-sized peripheral nerve defects (15 mm in rats and 25 mm in humans), which can be improved by a beneficial intraluminal microenvironment. However, individual cues provided by intraluminal filling materials are inadequate to eliminate the functional gap between regenerated nerves and normal nerves. Herein, an aligned fibrin/functionalized self-assembling peptide (AFG/fSAP) interpenetrating nanofiber hydrogel that exerting synergistic topographical and biochemical cues for peripheral nerve regeneration is constructed via electrospinning and molecular self-assembly. The hydrogel possesses an aligned structure, high water content, appropriate mechanical properties and suitable biodegradation capabilities for nerve repair, which enhances the alignment and neurotrophin secretion of primary Schwann cells (SCs) in vitro, and successfully bridges a 15-mm sciatic nerve gap in rats in vivo. The rats transplanted with the AFG/fSAP hydrogel exhibit satisfactory morphological and functional recovery in myelinated nerve fibers and innervated muscles. The motor function recovery facilitated by the AFG/fSAP hydrogel is comparable with that of autografts. Moreover, the AFG/fSAP hydrogel upregulates the regeneration-associated gene expression and activates the PI3K/Akt and MAPK signaling pathways in the regenerated nerve. Altogether, the AFG/fSAP hydrogel represents a promising approach for peripheral nerve repair through an integration of structural guidance and biochemical stimulation. A novel aligned interpenetrating nanofiber hydrogel is first constructed for peripheral nerve regeneration. The aligned hydrogel presents synergistic topographical and biochemical cues for peripheral nerve regeneration. Nerve conduits filled with the aligned hydrogel can repair the long-distance sciatic nerve defects in 12 weeks. The function recovery facilitated by the aligned hydrogel is comparable with that of autografts. The aligned hydrogel upregulates regeneration-related genes and activates the PI3K/Akt and MAPK signaling pathways.
Collapse
Affiliation(s)
- Shuhui Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Jinjin Zhu
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China.,Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang, Hangzhou, 310016, PR China
| | - Changfeng Lu
- Key Laboratory of Trauma and Neural Regeneration, Peking University, Ministry of Education, Department of Trauma and Orthopedics, Peking University People's Hospital, Beijing, 100044, PR China
| | - Yi Chai
- School of Clinical Medicine, Tsinghua University, Beijing, 100084, PR China
| | - Zheng Cao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Jiaju Lu
- School of Materials Science and Engineering, Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
| | - Zhe Zhang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| | - He Zhao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Yin-Yuan Huang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Shenglian Yao
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, PR China
| | - Xiangdong Kong
- School of Materials Science and Engineering, Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
| | - Peixun Zhang
- Key Laboratory of Trauma and Neural Regeneration, Peking University, Ministry of Education, Department of Trauma and Orthopedics, Peking University People's Hospital, Beijing, 100044, PR China
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| |
Collapse
|
27
|
Applications of Functionalized Hydrogels in the Regeneration of the Intervertebral Disc. BIOMED RESEARCH INTERNATIONAL 2021; 2021:2818624. [PMID: 34458364 PMCID: PMC8397561 DOI: 10.1155/2021/2818624] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 07/26/2021] [Indexed: 02/08/2023]
Abstract
Intervertebral disc degeneration (IDD) is caused by genetics, aging, and environmental factors and is one of the leading causes of low back pain. The treatment of IDD presents many challenges. Hydrogels are biomaterials that possess properties similar to those of the natural extracellular matrix and have significant potential in the field of regenerative medicine. Hydrogels with various functional qualities have recently been used to repair and regenerate diseased intervertebral discs. Here, we review the mechanisms of intervertebral disc homeostasis and degeneration and then discuss the applications of hydrogel-mediated repair and intervertebral disc regeneration. The classification of artificial hydrogels and natural hydrogels is then briefly introduced, followed by an update on the development of functional hydrogels, which include noncellular therapeutic hydrogels, cellular therapeutic hydrogel scaffolds, responsive hydrogels, and multifunctional hydrogels. The challenges faced and future developments of the hydrogels used in IDD are discussed as they further promote their clinical translation.
Collapse
|
28
|
Zhou G, Tian A, Yi X, Fan L, Shao W, Wu H, Sun N. Study on a 3D-Bioprinted Tissue Model of Self-Assembled Nanopeptide Hydrogels Combined With Adipose-Derived Mesenchymal Stem Cells. Front Bioeng Biotechnol 2021; 9:663120. [PMID: 34414170 PMCID: PMC8369258 DOI: 10.3389/fbioe.2021.663120] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 07/07/2021] [Indexed: 12/14/2022] Open
Abstract
Objective: This study aimed to observe the cell growth status and multidirectional differentiation ability in a 3D-bioprinted tissue model of self-assembled nanopeptides and human adipose-derived mesenchymal stem cells (Ad-MSCs). Methods: Primary Ad-MSCs were isolated, cultured, and identified by flow cytometry. Tissue models were printed via 3D bioprinting technology using a “biological ink” consisting of a mixed solution of self-assembled nanopeptides and Ad-MSCs. Ad-MSCs were induced into osteogenic, adipogenic, and endothelial differentiation and compared with the control groups by staining. Results: The nanopeptide fiber was 10–30 nm in diameter and 200–500 nm in length under the atomic-force microscope. It had the characteristics of nano-scale materials. Flow cytometry showed that the isolated and cultured cells were positive for CD29 (98.51%), CD90 (97.87%), and CD166 (98.32%) but did not express CD31 (1.58%), CD34 (2.42%), CD45 (2.95%), or human leukocyte antigen (HLA)-DR (0.53%), consistent with the immunophenotype of Ad-MSCs. Then, a tissue model was printed using the biological ink, followed by induction of differentiation of Ad-MSCs within the tissue model. Alizarin red S staining showed the formation of calcium nodules in the osteogenesis induction experimental group, and oil red O stained lipid droplets in Ad-MSCs in the adipogenesis induction experimental group, whereas the two control groups were not stained. Conclusion: Ad-MSCs from primary cultures have the characteristics of stem cells. Self-assembled nanopeptide hydrogel is a good tissue engineering material that can serve as an extracellular matrix. Ad-MSCs in the 3D-printed tissue model using a biological ink consisting of a mixed solution of self-assembled nanopeptides and Ad-MSCs grew well and still had strong differentiation ability.
Collapse
Affiliation(s)
- Guanzhou Zhou
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China.,Department of General Surgery, Zibo Central Hospital, Zibo, China
| | - Ailing Tian
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Xin Yi
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Lufeng Fan
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Wenchong Shao
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Han Wu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Nianfeng Sun
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
29
|
Lin SJ, Chan YC, Su ZC, Yeh WL, Lai PL, Chu IM. Growth factor-loaded microspheres in mPEG-polypeptide hydrogel system for articular cartilage repair. J Biomed Mater Res A 2021; 109:2516-2526. [PMID: 34190399 DOI: 10.1002/jbm.a.37246] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 01/20/2023]
Abstract
We developed an injectable hydrogel system with a sustained release of TGF-β3 through growth factor-loaded microsphere to mimic the cartilage-like microenvironment. Poly(lactic-co-glycolic acid) (PLGA) microspheres incorporated in three dimensional (3D) scaffolds were chosen because of its regulatory approval, good biodegradability, and acting as carriers with sustained release behavior. We evaluated sustained release of TGF-β3 by PLGA microspheres encapsulated in methoxy poly(ethylene glycol)-poly(alanine) (mPA) hydrogels and the resulting enhanced chondrogenic effects. We reported here the effect of the proposed system for sustained release of growth factors on chondrogenesis in cartilage regeneration. PLGA microspheres were used in our thermosensitive mPA hydrogel system with bovine serum albumin as a stabilizing and protecting agent for the emulsion and TGF-β3 enabling sustained release. Gelation, structural properties, and in-vitro release of this composite, that is, microspheres in hydrogel, system were investigated. Using PLGA microspheres to carry growth factors could complement the mPA hydrogel's ability to provide an excellent 3D microenvironment for the promotion of chondrogenic phenotype as compared the systems using mPA hydrogel or microspheres alone. Our study demonstrated that this synthesized composite hydrogel system is capable of modulating the biosynthetic and differentiation activities of chondrocytes. The sustained release of TGF-β3 in this novel hydrogel system could improve biomedical applicability of mPEG-polypeptide scaffolds. The distinctive local growth factor delivery system successfully combined the use of both polymers to be a suitable candidate for prolonged articular cartilage regeneration.
Collapse
Affiliation(s)
- Shih-Jie Lin
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan.,Department of Orthopaedic Surgery, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Yun-Chen Chan
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Zih-Cheng Su
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Wen-Ling Yeh
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan.,Department of Orthopaedic Surgery, Lotung Poh-Ai Hospital, Yilan, Taiwan
| | - Po-Liang Lai
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - I-Ming Chu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
30
|
Gibler P, Gimble J, Hamel K, Rogers E, Henderson M, Wu X, Olesky S, Frazier T. Human Adipose-Derived Stromal/Stem Cell Culture and Analysis Methods for Adipose Tissue Modeling In Vitro: A Systematic Review. Cells 2021; 10:1378. [PMID: 34204869 PMCID: PMC8227575 DOI: 10.3390/cells10061378] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/11/2022] Open
Abstract
Human adipose-derived stromal/stem cells (hASC) are widely used for in vitro modeling of physiologically relevant human adipose tissue. These models are useful for the development of tissue constructs for soft tissue regeneration and 3-dimensional (3D) microphysiological systems (MPS) for drug discovery. In this systematic review, we report on the current state of hASC culture and assessment methods for adipose tissue engineering using 3D MPS. Our search efforts resulted in the identification of 184 independent records, of which 27 were determined to be most relevant to the goals of the present review. Our results demonstrate a lack of consensus on methods for hASC culture and assessment for the production of physiologically relevant in vitro models of human adipose tissue. Few studies have assessed the impact of different 3D culture conditions on hASC adipogenesis. Additionally, there has been a limited use of assays for characterizing the functionality of adipose tissue in vitro. Results from this study suggest the need for more standardized culture methods and further analysis on in vitro tissue functionality. These will be necessary to validate the utility of 3D MPS as an in vitro model to reduce, refine, and replace in vivo experiments in the drug discovery regulatory process.
Collapse
Affiliation(s)
- Peyton Gibler
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Jeffrey Gimble
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
- Department of Structural and Cell Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Katie Hamel
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Emma Rogers
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Michael Henderson
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Xiying Wu
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Spencer Olesky
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Trivia Frazier
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
- Department of Structural and Cell Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
31
|
Alshehri S, Susapto HH, Hauser CAE. Scaffolds from Self-Assembling Tetrapeptides Support 3D Spreading, Osteogenic Differentiation, and Angiogenesis of Mesenchymal Stem Cells. Biomacromolecules 2021; 22:2094-2106. [PMID: 33908763 PMCID: PMC8382244 DOI: 10.1021/acs.biomac.1c00205] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/15/2021] [Indexed: 01/01/2023]
Abstract
The apparent rise of bone disorders demands advanced treatment protocols involving tissue engineering. Here, we describe self-assembling tetrapeptide scaffolds for the growth and osteogenic differentiation of human mesenchymal stem cells (hMSCs). The rationally designed peptides are synthetic amphiphilic self-assembling peptides composed of four amino acids that are nontoxic. These tetrapeptides can quickly solidify to nanofibrous hydrogels that resemble the extracellular matrix and provide a three-dimensional (3D) environment for cells with suitable mechanical properties. Furthermore, we can easily tune the stiffness of these peptide hydrogels by just increasing the peptide concentration, thus providing a wide range of peptide hydrogels with different stiffnesses for 3D cell culture applications. Since successful bone regeneration requires both osteogenesis and vascularization, our scaffold was found to be able to promote angiogenesis of human umbilical vein endothelial cells (HUVECs) in vitro. The results presented suggest that ultrashort peptide hydrogels are promising candidates for applications in bone tissue engineering.
Collapse
Affiliation(s)
- Salwa Alshehri
- Laboratory
for Nanomedicine, Division of Biological and Environmental
Science and Engineering and Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Hepi H. Susapto
- Laboratory
for Nanomedicine, Division of Biological and Environmental
Science and Engineering and Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Charlotte A. E. Hauser
- Laboratory
for Nanomedicine, Division of Biological and Environmental
Science and Engineering and Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| |
Collapse
|
32
|
Distaffen HE, Jones CW, Abraham BL, Nilsson BL. Multivalent display of chemical signals on
self‐assembled
peptide scaffolds. Pept Sci (Hoboken) 2021. [DOI: 10.1002/pep2.24224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
33
|
Baumgartner L, Wuertz-Kozak K, Le Maitre CL, Wignall F, Richardson SM, Hoyland J, Ruiz Wills C, González Ballester MA, Neidlin M, Alexopoulos LG, Noailly J. Multiscale Regulation of the Intervertebral Disc: Achievements in Experimental, In Silico, and Regenerative Research. Int J Mol Sci 2021; 22:E703. [PMID: 33445782 PMCID: PMC7828304 DOI: 10.3390/ijms22020703] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/22/2020] [Accepted: 12/24/2020] [Indexed: 12/17/2022] Open
Abstract
Intervertebral disc (IVD) degeneration is a major risk factor of low back pain. It is defined by a progressive loss of the IVD structure and functionality, leading to severe impairments with restricted treatment options due to the highly demanding mechanical exposure of the IVD. Degenerative changes in the IVD usually increase with age but at an accelerated rate in some individuals. To understand the initiation and progression of this disease, it is crucial to identify key top-down and bottom-up regulations' processes, across the cell, tissue, and organ levels, in health and disease. Owing to unremitting investigation of experimental research, the comprehension of detailed cell signaling pathways and their effect on matrix turnover significantly rose. Likewise, in silico research substantially contributed to a holistic understanding of spatiotemporal effects and complex, multifactorial interactions within the IVD. Together with important achievements in the research of biomaterials, manifold promising approaches for regenerative treatment options were presented over the last years. This review provides an integrative analysis of the current knowledge about (1) the multiscale function and regulation of the IVD in health and disease, (2) the possible regenerative strategies, and (3) the in silico models that shall eventually support the development of advanced therapies.
Collapse
Affiliation(s)
- Laura Baumgartner
- BCN MedTech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, 08018 Barcelona, Spain; (L.B.); (C.R.W.); (M.A.G.B.)
| | - Karin Wuertz-Kozak
- Department of Biomedical Engineering, Rochester Institute of Technology (RIT), Rochester, NY 14623, USA;
- Schön Clinic Munich Harlaching, Spine Center, Academic Teaching Hospital and Spine Research Institute of the Paracelsus Medical University Salzburg (Austria), 81547 Munich, Germany
| | - Christine L. Le Maitre
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield S1 1WB, UK;
| | - Francis Wignall
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Oxford Road, Manchester M13 9PT, UK; (F.W.); (S.M.R.); (J.H.)
| | - Stephen M. Richardson
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Oxford Road, Manchester M13 9PT, UK; (F.W.); (S.M.R.); (J.H.)
| | - Judith Hoyland
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Oxford Road, Manchester M13 9PT, UK; (F.W.); (S.M.R.); (J.H.)
| | - Carlos Ruiz Wills
- BCN MedTech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, 08018 Barcelona, Spain; (L.B.); (C.R.W.); (M.A.G.B.)
| | - Miguel A. González Ballester
- BCN MedTech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, 08018 Barcelona, Spain; (L.B.); (C.R.W.); (M.A.G.B.)
- Catalan Institution for Research and Advanced Studies (ICREA), Pg. Lluis Companys 23, 08010 Barcelona, Spain
| | - Michael Neidlin
- Department of Mechanical Engineering, National Technical University of Athens, 15780 Athens, Greece; (M.N.); (L.G.A.)
| | - Leonidas G. Alexopoulos
- Department of Mechanical Engineering, National Technical University of Athens, 15780 Athens, Greece; (M.N.); (L.G.A.)
| | - Jérôme Noailly
- BCN MedTech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, 08018 Barcelona, Spain; (L.B.); (C.R.W.); (M.A.G.B.)
| |
Collapse
|
34
|
Castillo-Díaz LA, Ruiz-Pacheco JA, Elsawy MA, Reyes-Martínez JE, Enríquez-Rodríguez AI. Self-Assembling Peptides as an Emerging Platform for the Treatment of Metabolic Syndrome. Int J Nanomedicine 2020; 15:10349-10370. [PMID: 33376325 PMCID: PMC7762440 DOI: 10.2147/ijn.s278189] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/21/2020] [Indexed: 12/14/2022] Open
Abstract
Metabolic syndrome comprises a cluster of comorbidities that represent a major risk of developing chronic diseases, such as type II diabetes, cardiovascular diseases, and stroke. Alarmingly, metabolic syndrome reaches epidemic proportions worldwide. Today, lifestyle changes and multiple drug-based therapies represent the gold standard to address metabolic syndrome. However, such approaches face two major limitations: complicated drug therapeutic regimes, which in most cases could lead to patient incompliance, and limited drug efficacy. This has encouraged scientists to search for novel routes to deal with metabolic syndrome and related diseases. Within such approaches, self-assembled peptide formulations have emerged as a promising alternative for treating metabolic syndrome. In particular, self-assembled peptide hydrogels, either as acellular or cell-load three-dimensional scaffoldings have reached significant relevance in the biomedical field to prevent and restore euglycemia, as well as for controlling cardiovascular diseases and obesity. This has been possible thanks to the physicochemical tunability of peptides, which are developed from a chemical toolbox of versatile amino acids enabling flexibility of designing a wide range of self-assembled/co-assembled nanostructures forming biocompatible viscoelastic hydrogels. Peptide hydrogels can be combined with several biological entities, such as extracellular matrix proteins, drugs or cells, forming functional biologics with therapeutic ability for treatment of metabolic syndrome-comorbidities. Additionally, self-assembly peptides combine safety, tolerability, and effectivity attributes; by this presenting a promising platform for the development of novel pharmaceuticals capable of addressing unmet therapeutic needs for diabetes, cardiovascular disorders and obesity. In this review, recent advances in developing self-assembly peptide nanostructures tailored for improving treatment of metabolic syndrome and related diseases will be discussed from basic research to preclinical research studies. Challenges facing the development of approved medicinal products based on self-assembling peptide nanomaterials will be discussed in light of regulatory requirement for clinical authorization.
Collapse
Affiliation(s)
| | - Juan Alberto Ruiz-Pacheco
- West Biomedical Research Center, National Council of Science and Technology, Guadalajara, Jalisco, Mexico
| | - Mohamed Ahmed Elsawy
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester, Leicestershire, UK
| | | | | |
Collapse
|
35
|
He L, Xiao Q, Zhao Y, Li J, Reddy S, Shi X, Su X, Chiu K, Ramakrishna S. Engineering an Injectable Electroactive Nanohybrid Hydrogel for Boosting Peripheral Nerve Growth and Myelination in Combination with Electrical Stimulation. ACS APPLIED MATERIALS & INTERFACES 2020; 12:53150-53163. [PMID: 33179500 DOI: 10.1021/acsami.0c16885] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Electrical stimulation (ES) can be used to manipulate recovery after peripheral nerve injuries. Although biomaterial-based strategies have already been implemented to gain momentum for ES and engineer permissive microenvironments for neural regeneration, the development of biomaterials for specific stimuli-responsive modulation of neural cell properties remains a challenge. Herein, we homogeneously incorporate pristine carbon nanotubes into a functional self-assembling peptide to prepare a hybrid hydrogel with good injectability and conductivity. Two-dimensional (on the surface) and three-dimensional (within the hybrid hydrogel) culturing experiments demonstrate that ES promotes axon outgrowth and Schwann cell (SC) migration away from dorsal root ganglia spheres, further revealing that ES-enhanced interactions between SCs and axons result in improved myelination. Thus, our study not only advances the development of tailor-made materials but also provides useful insights into comprehensive approaches for promoting nerve growth and presents a practical strategy of repairing peripheral nerve injuries.
Collapse
Affiliation(s)
- Liumin He
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
- MOE Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Qiao Xiao
- MOE Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Yuyuan Zhao
- MOE Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Jun Li
- MOE Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Sathish Reddy
- MOE Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Xueshuang Shi
- MOE Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Xin Su
- MOE Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Kin Chiu
- Department of Ophthalmology, Faculty of Medicine, The University of Hong Kong, Hongkong, China
| | - Seeram Ramakrishna
- MOE Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou 510632, China
- Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore, Singapore 117576, Singapore
| |
Collapse
|
36
|
Ding X, Zhao H, Li Y, Lee AL, Li Z, Fu M, Li C, Yang YY, Yuan P. Synthetic peptide hydrogels as 3D scaffolds for tissue engineering. Adv Drug Deliv Rev 2020; 160:78-104. [PMID: 33091503 DOI: 10.1016/j.addr.2020.10.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/25/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022]
Abstract
The regeneration of tissues and organs poses an immense challenge due to the extreme complexity in the research work involved. Despite the tissue engineering approach being considered as a promising strategy for more than two decades, a key issue impeding its progress is the lack of ideal scaffold materials. Nature-inspired synthetic peptide hydrogels are inherently biocompatible, and its high resemblance to extracellular matrix makes peptide hydrogels suitable 3D scaffold materials. This review covers the important aspects of peptide hydrogels as 3D scaffolds, including mechanical properties, biodegradability and bioactivity, and the current approaches in creating matrices with optimized features. Many of these scaffolds contain peptide sequences that are widely reported for tissue repair and regeneration and these peptide sequences will also be discussed. Furthermore, 3D biofabrication strategies of synthetic peptide hydrogels and the recent advances of peptide hydrogels in tissue engineering will also be described to reflect the current trend in the field. In the final section, we will present the future outlook in the design and development of peptide-based hydrogels for translational tissue engineering applications.
Collapse
Affiliation(s)
- Xin Ding
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| | - Huimin Zhao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yuzhen Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Ashlynn Lingzhi Lee
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Zongshao Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Mengjing Fu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Chengnan Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yi Yan Yang
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore.
| | - Peiyan Yuan
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|
37
|
Yang Z, Li H, Yuan Z, Fu L, Jiang S, Gao C, Wang F, Zha K, Tian G, Sun Z, Huang B, Wei F, Cao F, Sui X, Peng J, Lu S, Guo W, Liu S, Guo Q. Endogenous cell recruitment strategy for articular cartilage regeneration. Acta Biomater 2020; 114:31-52. [PMID: 32652223 DOI: 10.1016/j.actbio.2020.07.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/02/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023]
Abstract
In the absence of timely and proper treatments, injuries to articular cartilage (AC) can lead to cartilage degeneration and ultimately result in osteoarthritis. Regenerative medicine and tissue engineering techniques are emerging as promising approaches for AC regeneration and repair. Although the use of cell-seeded scaffolds prior to implantation can regenerate and repair cartilage lesions to some extent, these approaches are still restricted by limited cell sources, excessive costs, risks of disease transmission and complex manufacturing practices. Recently developed acellular scaffold approaches that rely on the recruitment of endogenous cells to the injured sites avoid these drawbacks and offer great promise for in situ AC regeneration. Multiple endogenous stem/progenitor cells (ESPCs) are found in joint-resident niches and have the capability to migrate to sites of injury to participate in AC regeneration. However, the natural recruitment of ESPCs is insufficient, and the local microenvironment is hostile after injury. Hence, an endogenous cell recruitment strategy based on the combination of chemoattractants and acellular scaffolds to effectively and specifically recruit ESPCs and improve local microenvironment may provide new insights into in situ AC regeneration. This review provides a brief overview of: (1) the status of endogenous cell recruitment strategy; (2) the subpopulations, potential migration routes (PMRs) of joint-resident ESPCs and their immunomodulatory and reparative effects; (3) chemoattractants and their potential adverse effects; (4) scaffold-based drug delivery systems (SDDSs) that are utilized for in situ AC regeneration; and (5) the challenges and future perspectives of endogenous cell recruitment strategy for AC regeneration. STATEMENT OF SIGNIFICANCE: Although the endogenous cell recruitment strategy for articular cartilage (AC) regeneration has been investigated for several decades, much work remains to be performed in this field. Future studies should have the following aims: (1) reporting the up-to-date progress in the endogenous cell recruitment strategies; (2) determining the subpopulations of ESPCs, the cellular and molecular mechanisms underlying the migration of these cells and their anti-inflammatory, immunomodulatory and reparative effects; (3) elucidating the chemoattractants that enhance ESPC recruitment and their potential adverse effects; and (4) developing advanced SDDSs for chemoattractant dispatch. Herein, we present a systematic overview of the aforementioned issues to provide a better understanding of endogenous cell recruitment strategies for AC regeneration and repair.
Collapse
|
38
|
Elastin-Collagen Based Hydrogels as Model Scaffolds to Induce Three-Dimensional Adipocyte Culture from Adipose Derived Stem Cells. Bioengineering (Basel) 2020; 7:bioengineering7030110. [PMID: 32932577 PMCID: PMC7552710 DOI: 10.3390/bioengineering7030110] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023] Open
Abstract
This study aimed to probe the effect of formulation of scaffolds prepared using collagen and elastin-like polypeptide (ELP) and their resulting physico-chemical and mechanical properties on the adipogenic differentiation of human adipose derived stem cells (hASCs). Six different ELP-collagen scaffolds were prepared by varying the collagen concentration (2 and 6 mg/mL), ELP addition (6 mg/mL), or crosslinking of the scaffolds. FTIR spectroscopy indicated secondary bonding interactions between collagen and ELP, while scanning electron microscopy revealed a porous structure for all scaffolds. Increased collagen concentration, ELP addition, and presence of crosslinking decreased swelling ratio and increased elastic modulus and compressive strength of the scaffolds. The scaffold characteristics influenced cell morphology, wherein the hASCs seeded in the softer, non-crosslinked scaffolds displayed a spread morphology. We determined that stiffer and/or crosslinked elastin-collagen based scaffolds constricted the spreading of hASCs, leading to a spheroid morphology and yielded an enhanced adipogenic differentiation as indicated by Oil Red O staining. Overall, this study underscored the importance of spheroid morphology in adipogenic differentiation, which will allow researchers to create more physiologically-relevant three-dimensional, in vitro culture models.
Collapse
|
39
|
Xia K, Chen Z, Chen J, Xu H, Xu Y, Yang T, Zhang Q. RGD- and VEGF-Mimetic Peptide Epitope-Functionalized Self-Assembling Peptide Hydrogels Promote Dentin-Pulp Complex Regeneration. Int J Nanomedicine 2020; 15:6631-6647. [PMID: 32982223 PMCID: PMC7495350 DOI: 10.2147/ijn.s253576] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 07/15/2020] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Cell-based tissue engineering is a promising method for dentin-pulp complex (DPC) regeneration. The challenges associated with DPC regeneration include the generation of a suitable microenvironment that facilitates the complete odontogenic differentiation of dental pulp stem cells (DPSCs) and the rapid induction of angiogenesis. Thus, the survival and subsequent differentiation of DPSCs are limited. Extracellular matrix (ECM)-like biomimetic hydrogels composed of self-assembling peptides (SAPs) were developed to provide an appropriate microenvironment for DPSCs. For functional DPC regeneration, the most important considerations are to provide an environment that promotes the adequate attachment of DPSCs and rapid vascularization of the regenerating pulp. Morphogenic signals in the form of growth factors (GFs) have been incorporated into SAPs to promote productive DPSC behaviors. However, the use of GFs has several drawbacks. We envision using a scaffold with SAPs coupled with long-term factors to increase DPSC attachment and vascularization as a method to address this challenge. METHODS In this study, we developed synthetic material for an SAP-based scaffold with RGD- and vascular endothelial growth factor (VEGF)-mimetic peptide epitopes with the dual functions of dentin and pulp regeneration. DPSCs and human umbilical vein endothelial cells (HUVECs) were used to evaluate the biological effects of SAP-based scaffolds. Furthermore, the pulpotomized molar rat model was employed to test the reparative and regenerative effects of SAP-based scaffolds. RESULTS This scaffold simultaneously presented RGD- and VEGF-mimetic peptide epitopes and provided a 3D microenvironment for DPSCs. DPSCs grown on this composite scaffold exhibited significantly improved survival and angiogenic and odontogenic differentiation in the multifunctionalized group in vitro. Histological and functional evaluations of a partially pulpotomized rat model revealed that the multifunctionalized scaffold was superior to other options with respect to stimulating pulp recovery and dentin regeneration in vivo. CONCLUSION Based on our data obtained with the functionalized SAP scaffold, a 3D microenvironment that supports stem cell adhesion and angiogenesis was generated that has great potential for dental pulp tissue engineering and regeneration.
Collapse
Affiliation(s)
- Kun Xia
- Department of Endodontics, School and Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai200072, People’s Republic of China
- Department of Preventive Dentistry, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou325027, People’s Republic of China
| | - Zhuo Chen
- Department of Endodontics, The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou310006, People’s Republic of China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou310006, People’s Republic of China
| | - Jie Chen
- Department of Endodontics, School and Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai200072, People’s Republic of China
| | - Huaxing Xu
- Department of Endodontics, School and Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai200072, People’s Republic of China
| | - Yunfei Xu
- Department of Endodontics, School and Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai200072, People’s Republic of China
| | - Ting Yang
- Department of Endodontics, School and Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai200072, People’s Republic of China
| | - Qi Zhang
- Department of Endodontics, School and Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai200072, People’s Republic of China
| |
Collapse
|
40
|
Mu X, Shi L, Pan S, He L, Niu Y, Wang X. A Customized Self-Assembling Peptide Hydrogel-Wrapped Stem Cell Factor Targeting Pulp Regeneration Rich in Vascular-Like Structures. ACS OMEGA 2020; 5:16568-16574. [PMID: 32685822 PMCID: PMC7364552 DOI: 10.1021/acsomega.0c01266] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 06/15/2020] [Indexed: 05/27/2023]
Abstract
Pulp regeneration is to replace the inflamed/necrotic pulp tissue with regenerated pulp-like tissue to rejuvenate the teeth. Self-assembling peptide hydrogels RADA16-I (Ac-(RADA16-I)4-CONH2) can provide a three-dimensional environment for cells. The stem cell factor (SCF) plays a crucial role in homing stem cells. Combining these advantages, our study investigated the effects of SCF-RADA16-I on adhesion, proliferation, and migration of human dental pulp stem cells (DPSCs) and the angiogenesis of human umbilical vein endothelial cells (HUVECs). The β-sheet and grid structure were observed by circular dichroism (CD), scanning electron microscopy (SEM), and atomic force microscopy (AFM). Cytoskeleton staining, living cell staining, cell viability, cell migration, angiogenesis, and western blot assays were performed, and the results indicated that all the SCF groups were superior to the corresponding non-SCF groups in cell adhesion, proliferation, migration, and angiogenesis. RADA16-I provided a three-dimensional environment for DPSCs. Besides, the SCF promoted HUVECs to form more vascular-like structures and release more vascular endothelial growth factor A. In summary, the SCF-loaded RADA16-I scaffold improved adhesion, proliferation, and migration of DPSCs and the formation of more vascular-like structures of HUVECs. SCF-RADA16-I holds promise for guided pulp regeneration, and it can potentially be applied widely in tissue engineering and translational medicine in the future.
Collapse
Affiliation(s)
- Xiaodan Mu
- The
First Affiliated Hospital, Harbin Medical
University, 23 Post Street, Nangang District, Harbin, Heilongjiang 150001, China
- School
of Stomatology, Harbin Medical University, 143 Yiman Street, Nangang District, Harbin, Heilongjiang 150001, China
| | - Lei Shi
- The
First Affiliated Hospital, Harbin Medical
University, 23 Post Street, Nangang District, Harbin, Heilongjiang 150001, China
- School
of Stomatology, Harbin Medical University, 143 Yiman Street, Nangang District, Harbin, Heilongjiang 150001, China
| | - Shuang Pan
- The
First Affiliated Hospital, Harbin Medical
University, 23 Post Street, Nangang District, Harbin, Heilongjiang 150001, China
- School
of Stomatology, Harbin Medical University, 143 Yiman Street, Nangang District, Harbin, Heilongjiang 150001, China
| | - Lina He
- The
First Affiliated Hospital, Harbin Medical
University, 23 Post Street, Nangang District, Harbin, Heilongjiang 150001, China
- School
of Stomatology, Harbin Medical University, 143 Yiman Street, Nangang District, Harbin, Heilongjiang 150001, China
| | - Yumei Niu
- The
First Affiliated Hospital, Harbin Medical
University, 23 Post Street, Nangang District, Harbin, Heilongjiang 150001, China
- School
of Stomatology, Harbin Medical University, 143 Yiman Street, Nangang District, Harbin, Heilongjiang 150001, China
| | - Xiumei Wang
- Department
of Materials Science and Engineering, State Key Laboratory of New
Ceramics and Fine Processing, Tsinghua University, 30 Shuangqing Road, Haidian District, Beijing 100084, China
| |
Collapse
|
41
|
Yang S, Wang C, Zhu J, Lu C, Li H, Chen F, Lu J, Zhang Z, Yan X, Zhao H, Sun X, Zhao L, Liang J, Wang Y, Peng J, Wang X. Self-assembling peptide hydrogels functionalized with LN- and BDNF- mimicking epitopes synergistically enhance peripheral nerve regeneration. Theranostics 2020; 10:8227-8249. [PMID: 32724468 PMCID: PMC7381722 DOI: 10.7150/thno.44276] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 05/31/2020] [Indexed: 12/16/2022] Open
Abstract
The regenerative capacity of the peripheral nervous system is closely related to the role that Schwann cells (SCs) play in construction of the basement membrane containing multiple extracellular matrix proteins and secretion of neurotrophic factors, including laminin (LN) and brain-derived neurotrophic factor (BDNF). Here, we developed a self-assembling peptide (SAP) nanofiber hydrogel based on self-assembling backbone Ac-(RADA)4-NH2 (RAD) dual-functionalized with laminin-derived motif IKVAV (IKV) and a BDNF-mimetic peptide epitope RGIDKRHWNSQ (RGI) for peripheral nerve regeneration, with the hydrogel providing a three-dimensional (3D) microenvironment for SCs and neurites. Methods: Circular dichroism (CD), atomic force microscopy (AFM), and scanning electron microscopy (SEM) were used to characterize the secondary structures, microscopic structures, and morphologies of self-assembling nanofiber hydrogels. Then the SC adhesion, myelination and neurotrophin secretion were evaluated on the hydrogels. Finally, the SAP hydrogels were injected into hollow chitosan tubes to bridge a 10-mm-long sciatic nerve defect in rats, and in vivo gene expression at 1 week, axonal regeneration, target muscular re-innervation, and functional recovery at 12 weeks were assessed. Results: The bioactive peptide motifs were covalently linked to the C-terminal of the self-assembling peptide and the functionalized peptides could form well-defined nanofibrous hydrogels capable of providing a 3D microenvironment similar to native extracellular matrix. SCs displayed improved cell adhesion on hydrogels with both IKV and RGI, accompanied by increased cell spreading and elongation relative to other groups. RSCs cultured on hydrogels with IKV and RGI showed enhanced gene expression of NGF, BDNF, CNTF, PMP22 and NRP2, and decreased gene expression of NCAM compared with those cultured on other three groups after a 7-day incubation. Additionally, the secretion of NGF, BDNF, and CNTF of RSCs was significantly improved on dual-functionalized peptide hydrogels after 3 days. At 1 week after implantation, the expressions of neurotrophin and myelin-related genes in the nerve grafts in SAP and Autograft groups were higher than that in Hollow group, and the expression of S100 in groups containing both IKV and RGI was significantly higher than that in groups containing either IKV or RGI hydrogels, suggesting enhanced SC proliferation. The morphometric parameters of the regenerated nerves, their electrophysiological performance, the innervated muscle weight and remodeling of muscle fibers, and motor function showed that RAD/IKV/RGI and RAD/IKV-GG-RGI hydrogels could markedly improve axonal regeneration with enhanced re-myelination and motor functional recovery through the synergetic effect of IKV and RGI functional motifs. Conclusions: We found that the dual-functionalized SAP hydrogels promoted RSC adhesion, myelination, and neurotrophin secretion in vitro and successfully bridged a 10-mm gap representing a sciatic nerve defect in rats in vivo. The results demonstrated the synergistic effect of IKVAV and RGI on axonal regrowth and function recovery after peripheral nerve injury.
Collapse
Affiliation(s)
- Shuhui Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Chong Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226007, China
| | - Jinjin Zhu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang, Hangzhou 310016, China
| | - Changfeng Lu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226007, China
- Department of Orthopaedics and Trauma, Peking University People's Hospital, Beijing 100191, China
| | - Haitao Li
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226007, China
| | - Fuyu Chen
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226007, China
| | - Jiaju Lu
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Zhe Zhang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Xiaoqing Yan
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
- School of Clinical Medicine, Tsinghua University, Beijing 100084, China
| | - He Zhao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Xiaodan Sun
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Lingyun Zhao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Jing Liang
- Department of Pediatrics, Tianjin Hospital, Tianjin University, No. 406 Jiefang Nan Road, Tianjin 300211, China
| | - Yu Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226007, China
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226007, China
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| |
Collapse
|
42
|
Nanofibrous polyester-polypeptide block copolymer scaffolds with high porosity and controlled degradation promote cell adhesion, proliferation and differentiation. Eur Polym J 2020. [DOI: 10.1016/j.eurpolymj.2020.109647] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
43
|
Yin H, Strunz F, Yan Z, Lu J, Brochhausen C, Kiderlen S, Clausen-Schaumann H, Wang X, Gomes ME, Alt V, Docheva D. Three-dimensional self-assembling nanofiber matrix rejuvenates aged/degenerative human tendon stem/progenitor cells. Biomaterials 2020; 236:119802. [PMID: 32014804 DOI: 10.1016/j.biomaterials.2020.119802] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 01/14/2020] [Accepted: 01/18/2020] [Indexed: 12/29/2022]
Abstract
The poor healing capacity of tendons is known to worsen in the elderly. During tendon aging and degeneration, endogenous human tendon stem/progenitor cells (hTSPCs) experience profound pathological changes. Here, we explored a rejuvenation strategy for hTSPCs derived from aged/degenerated Achilles tendons (A-TSPCs) by providing three-dimensional (3D) nanofiber hydrogels and comparing them to young/healthy TSPCs (Y-TSPCs). RADA peptide hydrogel has a self-assembling ability, forms a nanofibrous 3D niche and can be further functionalized by adding RGD motifs. Cell survival, apoptosis, and proliferation assays demonstrated that RADA and RADA/RGD hydrogels support A-TSPCs in a comparable manner to Y-TSPCs. Moreover, they rejuvenated A-TSPCs to a phenotype similar to that of Y-TSPCs, as evidenced by restored cell morphology and cytoskeletal architecture. Transmission electron, confocal laser scanning and atomic force microscopies demonstrated comparable ultrastructure, surface roughness and elastic modulus of A- and Y-TSPC-loaded hydrogels. Lastly, quantitative PCR revealed similar expression profiles, as well a significant upregulation of genes related to tenogenesis and multipotency. Taken together, the RADA-based hydrogels exert a rejuvenating effect by recapitulating in vitro specific features of the natural microenvironment of human TSPCs, which strongly indicates their potential to direct cell behaviour and overcome the challenge of cell aging and degeneration in tendon repair.
Collapse
Affiliation(s)
- Heyong Yin
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany; Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Franziska Strunz
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany.
| | - Zexing Yan
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany.
| | - Jiaju Lu
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, China.
| | | | - Stefanie Kiderlen
- Center for Applied Tissue Engineering and Regenerative Medicine, Munich University of Applied Sciences, Munich, Germany; Center for NanoScience, Ludwig-Maximilians-University, Munich, Germany.
| | - Hauke Clausen-Schaumann
- Center for Applied Tissue Engineering and Regenerative Medicine, Munich University of Applied Sciences, Munich, Germany; Center for NanoScience, Ludwig-Maximilians-University, Munich, Germany.
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, China.
| | - Manuela E Gomes
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal.
| | - Volker Alt
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany.
| | - Denitsa Docheva
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany.
| |
Collapse
|
44
|
Wei S, Chen F, Geng Z, Cui R, Zhao Y, Liu C. Self-assembling RATEA16 peptide nanofiber designed for rapid hemostasis. J Mater Chem B 2020; 8:1897-1905. [DOI: 10.1039/c9tb02590a] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In this study, we synthesized a novel polypeptide material, RATEA16, by the solid phase method, and investigated the secondary structure, self-assembly performance, gelation ability, biocompatibility and hemostatic efficiencyin vitroandin vivo.
Collapse
Affiliation(s)
- Shuda Wei
- Engineering Research Center for Biomedical Materials of Ministry of Education
- East China University of Science and Technology
- Shanghai 200237
- P. R. China
| | - Fangping Chen
- Engineering Research Center for Biomedical Materials of Ministry of Education
- East China University of Science and Technology
- Shanghai 200237
- P. R. China
- The State Key Laboratory of Bioreactor Engineering
| | - Zhen Geng
- Engineering Research Center for Biomedical Materials of Ministry of Education
- East China University of Science and Technology
- Shanghai 200237
- P. R. China
| | - Ruihua Cui
- Engineering Research Center for Biomedical Materials of Ministry of Education
- East China University of Science and Technology
- Shanghai 200237
- P. R. China
| | - Yujiao Zhao
- Engineering Research Center for Biomedical Materials of Ministry of Education
- East China University of Science and Technology
- Shanghai 200237
- P. R. China
| | - Changsheng Liu
- Engineering Research Center for Biomedical Materials of Ministry of Education
- East China University of Science and Technology
- Shanghai 200237
- P. R. China
- The State Key Laboratory of Bioreactor Engineering
| |
Collapse
|
45
|
Faroni A, Workman VL, Saiani A, Reid AJ. Self-Assembling Peptide Hydrogel Matrices Improve the Neurotrophic Potential of Human Adipose-Derived Stem Cells. Adv Healthc Mater 2019; 8:e1900410. [PMID: 31348622 DOI: 10.1002/adhm.201900410] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/09/2019] [Indexed: 12/20/2022]
Abstract
Despite advances in microsurgical techniques, treatment options to restore prior function following peripheral nerve injury remain unavailable, and autologous nerve grafting remains the therapy of choice. Recent experimental work has focused on the development of artificial constructs incorporating smart biomaterials and stem cells, aspiring to match/improve the outcomes of nerve autografting. Chemically stimulated human adipose-derived stem cells (dhASC) can improve nerve regeneration outcomes; however, these properties are lost when chemical stimulation is withdrawn, and survival rate upon transplantation is low. It is hypothesized that interactions with synthetic hydrogel matrices could maintain and improve neurotrophic characteristics of dhASC. dhASC are cultured on PeptiGel-Alpha 1 and PeptiGel-Alpha 2 self-assembling peptide hydrogels, showing comparable viability to collagen I control gels. Culturing dhASC on Alpha 1 and Alpha 2 substrates allow the maintenance of neurotrophic features, such as the expression of growth factors and neuroglial markers. Both Alpha 1 and Alpha 2 substrates are suitable for the culture of peripheral sensory neurons, permitting sprouting of neuronal extensions without the need of biological extracellular matrices, and preserving neuronal function. PeptiGel substrates loaded with hdASC are proposed as promising candidates for the development of tissue engineering therapies for the repair of peripheral nerve injuries.
Collapse
Affiliation(s)
- Alessandro Faroni
- Blond McIndoe LaboratoriesDivision of Cell Matrix Biology and Regenerative MedicineSchool of Biological SciencesFaculty of Biology Medicine and HealthUniversity of ManchesterManchester Academic Health Science Centre Manchester M13 9PL UK
| | - Victoria L. Workman
- School of Materials & Manchester Institute of BiotechnologyFaculty of Science and EngineeringUniversity of Manchester Manchester M13 9PL UK
| | - Alberto Saiani
- School of Materials & Manchester Institute of BiotechnologyFaculty of Science and EngineeringUniversity of Manchester Manchester M13 9PL UK
| | - Adam J. Reid
- Blond McIndoe LaboratoriesDivision of Cell Matrix Biology and Regenerative MedicineSchool of Biological SciencesFaculty of Biology Medicine and HealthUniversity of ManchesterManchester Academic Health Science Centre Manchester M13 9PL UK
- Department of Plastic Surgery & BurnsWythenshawe HospitalManchester University NHS Foundation TrustManchester Academic Health Science Centre Manchester M23 9LT UK
| |
Collapse
|
46
|
He X, Ding Y, Xie W, Sun R, Hunt NC, Song J, Sun X, Peng C, Zeng Q, Tan Y, Liu Y. Rubidium-Containing Calcium Alginate Hydrogel for Antibacterial and Diabetic Skin Wound Healing Applications. ACS Biomater Sci Eng 2019; 5:4726-4738. [DOI: 10.1021/acsbiomaterials.9b00547] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | - Yufang Ding
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha 410013, P. R. China
| | | | | | - Nicola Claire Hunt
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, U.K
| | - Jian Song
- State Key Laboratory of Tribology, Tsinghua University, Beijing 100084, P. R. China
| | - Xinxin Sun
- A. James Clark School of Engineering, University of Maryland, College Park 20742, Maryland, United States
| | - Chen Peng
- Key Laboratory of Advanced Manufacturing Technology of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, P. R. China
| | - Qinghai Zeng
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha 410013, P. R. China
| | | | | |
Collapse
|
47
|
Abstract
Stem cell therapy is a promising alternative approach to the treatment of a number of incurable degenerative diseases. However, low cell retention and survival after transplantation limit the therapeutic efficacy of stem cells for clinical translational applications. The utilization of biomaterials has been progressively successful in controlling the fate of transplanted cells by imitating the cellular microenvironment for optimal tissue repair and regeneration. This review mainly focuses on the engineered microenvironments with synthetic biomaterials in modification of stem cell behaviors. Moreover, the possible advancements in translational therapy by using biomaterials with stem cells are prospected and the challenges of the current restriction in clinical applications are highlighted.
Collapse
|
48
|
Millar-Haskell CS, Dang AM, Gleghorn JP. Coupling synthetic biology and programmable materials to construct complex tissue ecosystems. MRS COMMUNICATIONS 2019; 9:421-432. [PMID: 31485382 PMCID: PMC6724541 DOI: 10.1557/mrc.2019.69] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/15/2019] [Indexed: 05/17/2023]
Abstract
Synthetic biology combines engineering and biology to produce artificial systems with programmable features. Specifically, engineered microenvironments have advanced immensely over the past few decades, owing in part to the merging of materials with biological mimetic structures. In this review, we adapt a traditional definition of community ecology to describe "cellular ecology", or the study of the distribution of cell populations and interactions within their microenvironment. We discuss two exemplar hydrogel platforms: (1) self-assembling peptide (SAP) hydrogels and (2) Poly(ethylene) glycol (PEG) hydrogels and describe future opportunities for merging smart material design and synthetic biology within the scope of multicellular platforms.
Collapse
Affiliation(s)
| | - Allyson M. Dang
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716
| | - Jason P. Gleghorn
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19716
| |
Collapse
|
49
|
Tu Y, Chen N, Li C, Liu H, Zhu R, Chen S, Xiao Q, Liu J, Ramakrishna S, He L. Advances in injectable self-healing biomedical hydrogels. Acta Biomater 2019; 90:1-20. [PMID: 30951899 DOI: 10.1016/j.actbio.2019.03.057] [Citation(s) in RCA: 198] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 03/13/2019] [Accepted: 03/29/2019] [Indexed: 01/17/2023]
Abstract
In recent years, implantable biomaterials have attracted significant interest owing to their potentials for use in the therapy of physical defects and traumas. Among the implantable biomaterials, hydrogels have received increasing attention for their tunable structures and good rheological behavior. However, the mechanical failures of traditional gel materials during normal operation remain a serious issue. To overcome this problem, hydrogel materials with self-healing and injectable abilities have been developed, with their potential for autonomous self-recovery and minimally invasive implantation. In this paper, the progress of injectable self-healing hydrogels is presented by combining developments in the fundamental knowledge of polymer designs and discussions on the practical biomedical applications of the materials. The mechanisms of different types of self-healing hydrogels are introduced first and their performances are then discussed, followed by a review of the self-healing hydrogels with injectability. The applications of the injectable self-healing hydrogels are discussed in the final section. STATEMENT OF SIGNIFICANCE: This paper provides an overview of the progress of a smart material, injectable self-healing hydrogel, during the past ten years and mainly focuses on its recent development. This paper presents developments in the fundamental knowledge in polymer designs and discussions on the practical biomedical application of the materials, which sheds more light on the advancement of injectable self-healing hydrogels. This paper should be of interest to the readers who are curious about the advances of injectable self-healing hydrogels.
Collapse
|
50
|
Hellmund KS, Koksch B. Self-Assembling Peptides as Extracellular Matrix Mimics to Influence Stem Cell's Fate. Front Chem 2019; 7:172. [PMID: 31001512 PMCID: PMC6455064 DOI: 10.3389/fchem.2019.00172] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/05/2019] [Indexed: 12/13/2022] Open
Abstract
Interest in biologically active materials that can be used as cell culture substrates for medicinal applications has increased dramatically over the last decade. The design and development of biomaterials mimicking the natural environment of different cell types, the so-called extracellular matrix (ECM), is the focus of research in this field. The ECM exists as an ensemble of several adhesion proteins with different functionalities that can be presented to the embedded cells. These functionalities regulate numerous cellular processes. Therefore, different approaches and strategies using peptide- and protein-based biopolymers have been investigated to support the proliferation, differentiation, and self-renewal of stem cells, in the context of regenerative medicine. This minireview summarizes recent developments in this area, with a focus on peptide-based biomaterials used as stem cell culture substrates.
Collapse
Affiliation(s)
| | - Beate Koksch
- Department of Biology, Chemistry, and Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|