1
|
Wang Y, Zhang L, Liu J, Yang Y, Bi Z, Ouyang J. Mechanical Force Regulates the Paracrine Function of ADSCs to Promote the Adipose-Regenerating Effects of AAM by Regulating Angiogenesis and the Inflammatory Response. Cell Prolif 2025:e70045. [PMID: 40288954 DOI: 10.1111/cpr.70045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/30/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
Conditioned medium (CM), obtained by mechanical regulation of the paracrine activity of ADSCs, was fused with acellular adipose matrix (AAM) and methyl cellulose (MC) to synthesize a composite hydrogel which was grafted onto nude mice. The composite hydrogel could promote soft tissue regeneration by regulating the level of vascular regeneration and inflammation.
Collapse
Affiliation(s)
- Yining Wang
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, National Key Discipline of Human Anatomy, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China
| | - Luyu Zhang
- Yue Bei People's Hospital Postdoctoral Innovation Practice Base, Southern Medical University, Guangzhou, P. R. China
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, P. R. China
| | - Jiaxuan Liu
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, National Key Discipline of Human Anatomy, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China
| | - Yuchao Yang
- Yue Bei People's Hospital Postdoctoral Innovation Practice Base, Southern Medical University, Guangzhou, P. R. China
| | - Zhenyu Bi
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, National Key Discipline of Human Anatomy, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China
| | - Jun Ouyang
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, National Key Discipline of Human Anatomy, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China
| |
Collapse
|
2
|
Kapat K, Gondane P, Kumbhakarn S, Takle S, Sable R. Challenges and Opportunities in Developing Tracheal Substitutes for the Recovery of Long-Segment Defects. Macromol Biosci 2024; 24:e2400054. [PMID: 39008817 DOI: 10.1002/mabi.202400054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/21/2024] [Indexed: 07/17/2024]
Abstract
Tracheal resection and reconstruction procedures are necessary when stenosis, tracheomalacia, tumors, vascular lesions, or tracheal injury cause a tracheal blockage. Replacement with a tracheal substitute is often recommended when the trauma exceeds 50% of the total length of the trachea in adults and 30% in children. Recently, tissue engineering and other advanced techniques have shown promise in fabricating biocompatible tracheal substitutes with physical, morphological, biomechanical, and biological characteristics similar to native trachea. Different polymers and biometals are explored. Even with limited success with tissue-engineered grafts in clinical settings, complete healing of tracheal defects remains a substantial challenge due to low mechanical strength and durability of the graft materials, inadequate re-epithelialization and vascularization, and restenosis. This review has covered a range of reconstructive and regenerative techniques, design criteria, the use of bioprostheses and synthetic grafts for the recovery of tracheal defects, as well as the traditional and cutting-edge methods of their fabrication, surface modification for increased immuno- or biocompatibility, and associated challenges.
Collapse
Affiliation(s)
- Kausik Kapat
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata, West Bengal, 700054, India
| | - Prashil Gondane
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata, West Bengal, 700054, India
| | - Sakshi Kumbhakarn
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata, West Bengal, 700054, India
| | - Shruti Takle
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata, West Bengal, 700054, India
| | - Rahul Sable
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata, West Bengal, 700054, India
| |
Collapse
|
3
|
Yang X, Jin L, Xu M, Liu X, Tan Z, Liu L. Adipose tissue reconstruction facilitated with low immunogenicity decellularized adipose tissue scaffolds. Biomed Mater 2024; 19:035023. [PMID: 38518362 DOI: 10.1088/1748-605x/ad3705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 03/22/2024] [Indexed: 03/24/2024]
Abstract
There is currently an urgent need to develop engineered scaffolds to support new adipose tissue formation and facilitate long-term maintenance of function and defect repair to further generate prospective bioactive filler materials capable of fulfilling surgical needs. Herein, adipose regeneration methods were optimized and decellularized adipose tissue (DAT) scaffolds with good biocompatibility were fabricated. Adipose-like tissues were reconstructed using the DAT and 3T3-L1 preadipocytes, which have certain differentiation potential, and the regenerative effects of the engineered adipose tissuesin vitroandin vivowere explored. The method improved the efficiency of adipose removal from tissues, and significantly shortened the time for degreasing. Thus, the DAT not only provided a suitable space for cell growth but also promoted the proliferation, migration, and differentiation of preadipocytes within it. Following implantation of the constructed adipose tissuesin vivo, the DAT showed gradual degradation and integration with surrounding tissues, accompanied by the generation of new adipose tissue analogs. Overall, the combination of adipose-derived extracellular matrix and preadipocytes for adipose tissue reconstruction will be of benefit in the artificial construction of biomimetic implant structures for adipose tissue reconstruction, providing a practical guideline for the initial integration of adipose tissue engineering into clinical medicine.
Collapse
Affiliation(s)
- Xun Yang
- Department of Traumatic Orthopedics, Shenzhen Second People's Hospital, The First Affiliated Hospital, Shenzhen University, Shenzhen 518028, People's Republic of China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, People's Republic of China
| | - Lijuan Jin
- Institute of Shenzhen, Hunan University, Shenzhen 518000, People's Republic of China
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou 511300, People's Republic of China
| | - Miaomiao Xu
- College of Biology, Hunan University, Changsha 410082, People's Republic of China
| | - Xiao Liu
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou 511300, People's Republic of China
| | - Zhikai Tan
- Institute of Shenzhen, Hunan University, Shenzhen 518000, People's Republic of China
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou 511300, People's Republic of China
- College of Biology, Hunan University, Changsha 410082, People's Republic of China
| | - Lijun Liu
- Department of Traumatic Orthopedics, Shenzhen Second People's Hospital, The First Affiliated Hospital, Shenzhen University, Shenzhen 518028, People's Republic of China
| |
Collapse
|
4
|
Wang Y, Chai YQ, Cai J, Huang SS, Wang YF, Yuan SS, Wang JL, Shi KQ, Deng JJ. Human Adipose Tissue Lysate-Based Hydrogel for Lasting Immunomodulation to Effectively Improve Spinal Cord Injury Repair. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2304318. [PMID: 38018305 DOI: 10.1002/smll.202304318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/22/2023] [Indexed: 11/30/2023]
Abstract
The long-term inflammatory microenvironment is one of the main obstacles to inhibit acute spinal cord injury (SCI) repair. The natural adipose tissue-derived extracellular matrix hydrogel shows effective anti-inflammatory regulation because of its unique protein components. However, the rapid degradation rate and removal of functional proteins during the decellularization process impair the lasting anti-inflammation function of the adipose tissue-derived hydrogel. To address this problem, adipose tissue lysate provides an effective way for SCI repair due to its abundance of anti-inflammatory and nerve regeneration-related proteins. Thereby, human adipose tissue lysate-based hydrogel (HATLH) with an appropriate degradation rate is developed, which aims to in situ long-term recruit and induce anti-inflammatory M2 macrophages through sustainedly released proteins. HATLH can recruit and polarize M2 macrophages while inhibiting pro-inflammatory M1 macrophages regardless of human or mouse-originated. The axonal growth of neuronal cells also can be effectively improved by HATLH and HATLH-induced M2 macrophages. In vivo experiments reveal that HATLH promotes endogenous M2 macrophages infiltration in large numbers (3.5 × 105/100 µL hydrogel) and maintains a long duration for over a month. In a mouse SCI model, HATLH significantly inhibits local inflammatory response, improves neuron and oligodendrocyte differentiation, enhances axonal growth and remyelination, as well as accelerates neurological function restoration.
Collapse
Affiliation(s)
- Yu Wang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang Province, Wenzhou, Zhejiang, 325000, China
- Zhejiang Engineering Research Center for Hospital Emergency and Process Digitization, Wenzhou, Zhejiang, 325000, China
| | - Ying-Qian Chai
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Jie Cai
- Department of Orthopedics, Xiaoshan Hospital Affiliated to Wenzhou Medical University, Hangzhou, Zhejiang, 310000, China
| | - Shan-Shan Huang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Ye-Feng Wang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Shan-Shan Yuan
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Ji-Long Wang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Ke-Qing Shi
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Jun-Jie Deng
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| |
Collapse
|
5
|
Cui L, Zhao Y, Zhong Y, Zhang L, Zhang X, Guo Z, Wang F, Chen X, Tong H, Fan J. Combining decellularized adipose tissue with decellularized adventitia extravascular matrix or small intestinal submucosa matrix for the construction of vascularized tissue-engineered adipose. Acta Biomater 2023; 170:567-579. [PMID: 37683968 DOI: 10.1016/j.actbio.2023.08.060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 08/24/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023]
Abstract
Adipose tissue is an endocrine organ. It serves many important functions, such as energy storage, hormones secretion, and providing insulation, cushioning and aesthetics to the body etc. Adipose tissue engineering offers a promising treatment for soft tissue defects. Early adipose tissue production and long-term survival are closely associated with angiogenesis. Decellularized matrix has a natural ECM (extracellular matrix) component, good biocompatibility, and low immunogenicity. Therefore, in this study, the injectable composite hydrogels were developed to construct vascularized tissue-engineered adipose by using the pro-angiogenic effects of aortic adventitia extravascular matrix (Adv) or small intestinal submucosa (SIS), and the pro-adipogenic effects of decellularized adipose tissue (DAT). The composite hydrogels were cross-linked by genipin. The adipogenic and angiogenic abilities of composite hydrogels were investigated in vitro, and in a rat dorsal subcutaneous implant model. The results showed that DAT and SIS or Adv 1:1 composite hydrogel promoted the migration and tube formation of endothelial cells. Furthermore, DAT and SIS or Adv 1:1 composite hydrogel enhanced adipogenic differentiation of adipose-derived mesenchymal stem cells (ASCs) through activation of PPARγ and C/EBPα. The in vivo studies further demonstrated that DAT with SIS or Adv in a 1:1 ratio also significantly promoted adipogenesis and angiogenesis. In addition, DAT with SIS or Adv in a 1:1 ratio hydrogel recruited macrophage population with enhanced M2-type macrophage polarization, suggesting a positive effect of inflammatory response on angiogenesis. In conclusion, these data suggest that the composite hydrogels of DAT with SIS or Adv in 1:1 ratio have apparent pro-adiogenic and angiogenic abilities, thus providing a promising cell-free tissue engineering biomaterial with broad clinical applications. STATEMENT OF SIGNIFICANCE: Decellularized adipose tissue (DAT) has emerged as an important biomaterial in adipose tissue regeneration. Early adipose tissue production and long-term survival is tightly related to the angiogenesis. The revascularization of the DAT is a key issue that needs to be solved in adipose regeneration. In this study, the injectable composite hydrogels were developed by using DAT with Adv (aortic adventitia extravascular matrix) or SIS (small intestinal submucosa) in different ratio. We demonstrated that the combination of DAT with SIS or Adv in 1:1 ratio effectively improved the proliferation of adipose stem cells and endothelial cells, and promoted greater adipose regeneration and tissue vascularization as compared to the DAT scaffold. This study provides the potential biomaterial for clinical soft tissue regeneration.
Collapse
Affiliation(s)
- Lu Cui
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, PR China
| | - Yujia Zhao
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, PR China
| | - Yuxuan Zhong
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, PR China
| | - Lanlan Zhang
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, PR China
| | - Xinnan Zhang
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, PR China
| | - Zhenglong Guo
- Second Clinical Medical College, Shengjing Hospital, China Medical University, No.36 Sanhao Road, Heping District, Shenyang, Liaoning Province 110004, PR China
| | - Fanglin Wang
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, PR China
| | - Xin Chen
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, PR China
| | - Hao Tong
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, PR China
| | - Jun Fan
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, PR China.
| |
Collapse
|
6
|
Nam H, Kim T, Moon S, Ji Y, Lee JB. Self-assembly of a multimeric genomic hydrogel via multi-primed chain reaction of dual single-stranded circular plasmids for cell-free protein production. iScience 2023; 26:107089. [PMID: 37416467 PMCID: PMC10319821 DOI: 10.1016/j.isci.2023.107089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/20/2023] [Accepted: 06/07/2023] [Indexed: 07/08/2023] Open
Abstract
Recent technical advances in cell-free protein synthesis (CFPS) offer several advantages over cell-based expression systems, including the application of cellular machinery, such as transcription and translation, in the test tube. Inspired by the advantages of CFPS, we have fabricated a multimeric genomic DNA hydrogel (mGD-gel) via rolling circle chain amplification (RCCA) using dual single-stranded circular plasmids with multiple primers. The mGD-gel exhibited significantly enhanced protein yield. In addition, mGD-gel can be reused at least five times, and the shape of the mGD-gel can be easily manipulated without losing the feasibility of protein expression. The mGD-gel platform based on the self-assembly of multimeric genomic DNA strands (mGD strands) has the potential to be used in CFPS systems for a variety of biotechnological applications.
Collapse
Affiliation(s)
- Hyangsu Nam
- Department of Chemical Engineering, University of Seoul, 163 Seoulsiripdaero, Dongdaemun-gu, Seoul 02504, Republic of Korea
| | - Taehyeon Kim
- Department of Chemical Engineering, University of Seoul, 163 Seoulsiripdaero, Dongdaemun-gu, Seoul 02504, Republic of Korea
| | - Sunghyun Moon
- Department of Chemical Engineering, University of Seoul, 163 Seoulsiripdaero, Dongdaemun-gu, Seoul 02504, Republic of Korea
| | - Yoonbin Ji
- Department of Chemical Engineering, University of Seoul, 163 Seoulsiripdaero, Dongdaemun-gu, Seoul 02504, Republic of Korea
| | - Jong Bum Lee
- Department of Chemical Engineering, University of Seoul, 163 Seoulsiripdaero, Dongdaemun-gu, Seoul 02504, Republic of Korea
| |
Collapse
|
7
|
Jiang B, Shi A, Xu Y, Zhang Y, Chen Y, Jiang X, Liu H, Zhang L. SDF-1α and CTGF functionalized Gelatin methacryloyl (GelMA) hydrogels enhance fibroblast activation to promote wound healing. MATERIALS TODAY COMMUNICATIONS 2023; 34:105152. [DOI: 10.1016/j.mtcomm.2022.105152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
8
|
Long J, Qin Z, Chen G, Song B, Zhang Z. Decellularized extracellular matrix (d-ECM): the key role of the inflammatory process in pre-regeneration after implantation. Biomater Sci 2023; 11:1215-1235. [PMID: 36625281 DOI: 10.1039/d2bm01204a] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Clinical medicine is encountering the challenge of repairing soft-tissue defects. Currently, natural and synthetic materials have been developed as natural scaffolds. Among them, the decellularized extracellular matrix (d-ECM) can achieve tissue remodeling following injury and, thus, replace defects due to its advantages of the extensiveness of the source and excellent biological and mechanical properties. However, by analyzing the existing decellularization techniques, we found that different preparation methods directly affect the residual components of the d-ECM, and further have different effects on inflammation and regeneration of soft tissues. Therefore, we analyzed the role of different residual components of the d-ECM after decellularization. Then, we explored the inflammatory process and immune cells in an attempt to understand the mechanisms and causes of tissue degeneration and regeneration after transplantation. In this paper, we summarize the current studies related to updated protocols for the preparation of the d-ECM, biogenic and exogenous residual substances, inflammation, and immune cells influencing the fate of the d-ECM.
Collapse
Affiliation(s)
- Jie Long
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Zijin Qin
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Guo Chen
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Baoqiang Song
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Ziang Zhang
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
9
|
Guo Y, Sun L, Wang Y, Wang Q, Jing D, Liu S. Nanomaterials based on thermosensitive polymer in biomedical field. Front Chem 2022; 10:946183. [PMID: 36212064 PMCID: PMC9532752 DOI: 10.3389/fchem.2022.946183] [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: 05/17/2022] [Accepted: 08/30/2022] [Indexed: 11/27/2022] Open
Abstract
The progress of nanotechnology enables us to make use of the special properties of materials on the nanoscale and open up many new fields of biomedical research. Among them, thermosensitive nanomaterials stand out in many biomedical fields because of their “intelligent” behavior in response to temperature changes. However, this article mainly reviews the research progress of thermosensitive nanomaterials, which are popular in biomedical applications in recent years. Here, we simply classify the thermally responsive nanomaterials according to the types of polymers, focusing on the mechanisms of action and their advantages and potential. Finally, we deeply investigate the applications of thermosensitive nanomaterials in drug delivery, tissue engineering, sensing analysis, cell culture, 3D printing, and other fields and probe the current challenges and future development prospects of thermosensitive nanomaterials.
Collapse
Affiliation(s)
- Yingshu Guo
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
- *Correspondence: Yingshu Guo,
| | - Li Sun
- School of Chemistry and Chemical Engineering, Linyi University, Linyi, China
| | - Yajing Wang
- School of Chemistry and Chemical Engineering, Linyi University, Linyi, China
| | - Qianqian Wang
- School of Chemistry and Chemical Engineering, Linyi University, Linyi, China
| | - Dan Jing
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Shiwei Liu
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| |
Collapse
|
10
|
Sutrisno L, Chen H, Yoshitomi T, Kawazoe N, Yang Y, Chen G. Preparation of composite scaffolds composed of gelatin and Au nanostar-deposited black phosphorus nanosheets for the photothermal ablation of cancer cells and adipogenic differentiation of stem cells. BIOMATERIALS ADVANCES 2022; 138:212938. [PMID: 35913234 DOI: 10.1016/j.bioadv.2022.212938] [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: 03/04/2022] [Revised: 05/11/2022] [Accepted: 05/21/2022] [Indexed: 06/15/2023]
Abstract
Photothermal nanoparticles are important in photothermal therapy. Combining different nanoparticles can achieve a high photothermal capacity. In this study, composite nanoparticles composed of black phosphorus nanosheets (BPNSs) and gold nanostars (BP-AuNSs) were synthesized by using BPNSs as the reductant. AuNSs were deposited on the BPNSs. The BP-AuNSs were further hybridized with porous gelatin scaffolds to prepare gelatin-BP-AuNS composite scaffolds. The gelatin-BP-AuNS composite scaffolds promoted cell migration and distribution. The synergistic effects of the BPNSs and AuNSs endowed the gelatin-BP-AuNS composite scaffolds with excellent photothermal properties. The gelatin-BP-AuNS composite scaffolds eliminated cancer cells after near infrared laser exposure and supported the adipogenic differentiation of human mesenchymal stem cells. Thus, this gelatin-BP-AuNS composite scaffold holds promise for breast cancer therapy.
Collapse
Affiliation(s)
- Linawati Sutrisno
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Huajian Chen
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Toru Yoshitomi
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Naoki Kawazoe
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Yingnan Yang
- Graduate School of Life and Environmental Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8572, Japan
| | - Guoping Chen
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan.
| |
Collapse
|
11
|
Béduer A, Genta M, Kunz N, Verheyen C, Martins M, Brefie-Guth J, Braschler T. Design of an elastic porous injectable biomaterial for tissue regeneration and volume retention. Acta Biomater 2022; 142:73-84. [PMID: 35101581 DOI: 10.1016/j.actbio.2022.01.050] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 01/01/2022] [Accepted: 01/24/2022] [Indexed: 11/01/2022]
Abstract
Soft tissue reconstruction currently relies on two main approaches, one involving the implantation of external biomaterials and the second one exploiting surgical autologous tissue displacement. While both methods have different advantages and disadvantages, successful long-term solutions for soft tissue repair are still limited. Specifically, volume retention over time and local tissue regeneration are the main challenges in the field. In this study the performance of a recently developed elastic porous injectable (EPI) biomaterial based on crosslinked carboxymethylcellulose is analyzed. Nearly quantitative volumetric stability, with over 90% volume retention at 6 months, is observed, and the pore space of the material is effectively colonized with autologous fibrovascular tissue. A comparative analysis with hyaluronic acid and collagen-based clinical reference materials is also performed. Mechanical stability, evidenced by a low-strain elastic storage modulus (G') approaching 1kPa and a yield strain of several tens of percent, is required for volume retention in-vivo. Macroporosity, along with in-vivo persistence of at least several months, is instead needed for successful host tissue colonization. This study demonstrates the importance of understanding material design criteria and defines the biomaterial requirements for volume retention and tissue colonization in soft tissue regeneration. STATEMENT OF SIGNIFICANCE: We present the design of an elastic, porous, injectable (EPI) scaffold suspension capable of inducing a precisely defined, stable volume of autologous connective tissue in situ. It combines volume stability and vascularized tissue induction capacity known from bulk scaffolds with the ease of injection in shear yielding materials. By comparative study with a series of clinically established biomaterials including a wound healing matrix and dermal fillers, we establish design rules regarding rheological and compressive mechanical properties as well as degradation characteristics that rationally underpin the volume stability and tissue induction in a high-performance biomaterial. These design rules should allow to streamline the development of new colonizable injectables.
Collapse
|
12
|
Chen J, Wu J, Raffa P, Picchioni F, Koning CE. Superabsorbent Polymers: From long-established, microplastics generating systems, to sustainable, biodegradable and future proof alternatives. Prog Polym Sci 2022. [DOI: 10.1016/j.progpolymsci.2021.101475] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
13
|
Sutrisno L, Chen H, Yoshitomi T, Kawazoe N, Yang Y, Chen G. PLGA-collagen-BPNS Bifunctional composite mesh for photothermal therapy of melanoma and skin tissue engineering. J Mater Chem B 2022; 10:204-213. [PMID: 34935026 DOI: 10.1039/d1tb02366g] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The treatment of melanoma requires not only the elimination of skin cancer cells but also skin regeneration to heal defects. To achieve this goal, a bifunctional composite scaffold of poly(DL-lactic-co-glycolic acid) (PLGA), collagen and black phosphorus nanosheets (BPNSs) was prepared by hybridizing a BPNS-embedded collagen sponge with a PLGA knitted mesh. The composite mesh increased the temperature under near-infrared laser irradiation. The incorporation of BPNSs provided the PLGA-collagen-BPNS composite mesh with excellent photothermal properties for the photothermal ablation of melanoma cells both in vitro and in vivo. The PLGA-collagen-BPNS composite mesh had high mechanical strength for easy handling. The PLGA-collagen-BPNS composite mesh facilitated the proliferation of fibroblasts, promoted the expression of angiogenesis-related genes and the genes of components of the extracellular matrix for skin tissue regeneration. The high mechanical strength, photothermal ablation capability and skin tissue regeneration effects demonstrate that the bifunctional PLGA-collagen-BPNS composite mesh is a versatile and effective platform for the treatment of melanoma and the regeneration of skin defects.
Collapse
Affiliation(s)
- Linawati Sutrisno
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan.
- Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Huajian Chen
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan.
- Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Toru Yoshitomi
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan.
| | - Naoki Kawazoe
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan.
| | - Yingnan Yang
- Graduate School of Life and Environmental Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572, Japan
| | - Guoping Chen
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan.
- Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| |
Collapse
|
14
|
Liu K, He Y, Yao Y, Zhang Y, Cai Z, Ru J, Zhang X, Jin X, Xu M, Li Y, Ma Q, Gao J, Lu F. Methoxy polyethylene glycol modification promotes adipogenesis by inducing the production of regulatory T cells in xenogeneic acellular adipose matrix. Mater Today Bio 2021; 12:100161. [PMID: 34870140 PMCID: PMC8626673 DOI: 10.1016/j.mtbio.2021.100161] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 12/22/2022] Open
Abstract
Acellular adipose matrix (AAM) has emerged as an important biomaterial for adipose tissue regeneration. Current decellularization methods damage the bioactive components of the extracellular matrix (ECM), and the residual immunogenic antigens may induce adverse immune responses. Here, we adopted a modified decellularization method which can protect more bioactive components with less immune reaction by methoxy polyethylene glycol (mPEG). Then, we determined the adipogenic mechanisms of mPEG-modified AAM after xenogeneic transplantation. AAM transplantation caused significantly lesser adipogenesis in the wild-type group than in the immune-deficient group. The mPEG-modified AAM showed significantly lower immunogenicity and higher adipogenesis than the AAM alone after xenogeneic transplantation. Furthermore, mPEG modification increased regulatory T (Treg) cell numbers in the AAM grafts, which in turn enhanced the M2/M1 macrophage ratio by secreting IL-10, IL-13, and TGF-β1. These findings suggest that mPEG modification effectively reduces the immunogenicity of xenogeneic AAM and promotes adipogenesis in the AAM grafts. Hence, mPEG-modified AAM can serve as an ideal biomaterial for xenogeneic adipose tissue engineering.
Collapse
Affiliation(s)
- Kaiyang Liu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yunfan He
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yao Yao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yuchen Zhang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Zihan Cai
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Jiangjiang Ru
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xiangdong Zhang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xiaoxuan Jin
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Mimi Xu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yibao Li
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Qizhuan Ma
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Jianhua Gao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
15
|
Li S, Liu Y, McCann J, Ravnic DJ, Gimble JM, Hayes DJ. Hybrid adipose graft materials synthesized from chemically modified adipose extracellular matrix. J Biomed Mater Res A 2021; 110:156-163. [PMID: 34263999 DOI: 10.1002/jbm.a.37273] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/24/2021] [Accepted: 07/07/2021] [Indexed: 12/17/2022]
Abstract
Decellularized extracellular matrix (ECM) from tissues is a promising biomaterial that can provide a complex 3D microenvironment capable of modulating cell response and tissue regeneration. In this study, we have integrated the decellularized thiolated adipose-derived ECM, at different concentrations, with polyethylene glycol (PEG) using Michael addition between thiol and acrylate moieties. The potential for this material to support adipogenic differentiation of human adipose-derived stem cells was evaluated by encapsulating cells in hydrogels with increasing concentrations of chemically modified ECM (mECM). Our results demonstrated a positive correlation between the ECM content in the hydrogels and cell proliferation, adipogenic marker expression, and lipid formation and accumulation. Furthermore, we have shown host cell infiltration and enhanced adipogenesis in vivo after implantation. These findings support the graft as a potential alternative for adipose tissue regeneration.
Collapse
Affiliation(s)
- Shue Li
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Yiming Liu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Jacob McCann
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Dino J Ravnic
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Jeffrey M Gimble
- Obatala Sciences, Inc., Advanced Materials Research Institute, University of New Orleans, New Orleans, Louisiana, USA
| | - Daniel J Hayes
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA.,Material Research Institute, The Pennsylvania State University, University Park, Pennsylvania, USA.,The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
16
|
Hu H, Liu W, Sun C, Wang Q, Yang W, Zhang Z, Xia Z, Shao Z, Wang B. Endogenous Repair and Regeneration of Injured Articular Cartilage: A Challenging but Promising Therapeutic Strategy. Aging Dis 2021; 12:886-901. [PMID: 34094649 PMCID: PMC8139200 DOI: 10.14336/ad.2020.0902] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/02/2020] [Indexed: 12/12/2022] Open
Abstract
Articular cartilage (AC) has a very limited intrinsic repair capacity after injury or disease. Although exogenous cell-based regenerative approaches have obtained acceptable outcomes, they are usually associated with complicated procedures, donor-site morbidities and cell differentiation during ex vivo expansion. In recent years, endogenous regenerative strategy by recruiting resident mesenchymal stem/progenitor cells (MSPCs) into the injured sites, as a promising alternative, has gained considerable attention. It takes full advantage of body's own regenerative potential to repair and regenerate injured tissue while avoiding exogenous regenerative approach-associated limitations. Like most tissues, there are also multiple stem-cell niches in AC and its surrounding tissues. These MSPCs have the potential to migrate into injured sites to produce replacement cells under appropriate stimuli. Traditional microfracture procedure employs the concept of MSPCs recruitment usually fails to regenerate normal hyaline cartilage. The reasons for this failure might be attributed to an inadequate number of recruiting cells and adverse local tissue microenvironment after cartilage injury. A strategy that effectively improves local matrix microenvironment and recruits resident MSPCs may enhance the success of endogenous AC regeneration (EACR). In this review, we focused on the reasons why AC cannot regenerate itself in spite of potential self-repair capacity and summarized the latest developments of the three key components in the field of EACR. In addition, we discussed the challenges facing in the present EACR strategy. This review will provide an increasing understanding of EACR and attract more researchers to participate in this promising research arena.
Collapse
Affiliation(s)
- Hongzhi Hu
- 1Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Weijian Liu
- 1Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Caixia Sun
- 2Department of Gynecology, General Hospital of the Yangtze River Shipping, Wuhan 430022, China
| | - Qiuyuan Wang
- 3Department of Nephrology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441100, China
| | - Wenbo Yang
- 1Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - ZhiCai Zhang
- 1Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhidao Xia
- 4Centre for Nanohealth, ILS2, Swansea university Medical school, Swansea, SA2 8PP, UK
| | - Zengwu Shao
- 1Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Baichuan Wang
- 1Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,4Centre for Nanohealth, ILS2, Swansea university Medical school, Swansea, SA2 8PP, UK
| |
Collapse
|
17
|
Sutrisno L, Chen H, Chen Y, Yoshitomi T, Kawazoe N, Yang Y, Chen G. Composite scaffolds of black phosphorus nanosheets and gelatin with controlled pore structures for photothermal cancer therapy and adipose tissue engineering. Biomaterials 2021; 275:120923. [PMID: 34098151 DOI: 10.1016/j.biomaterials.2021.120923] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/17/2021] [Accepted: 05/21/2021] [Indexed: 02/07/2023]
Abstract
Breast cancer treatment needs to eradicate cancer cells and restore breast defects after surgical intervention. Herein, bifunctional composite scaffolds of black phosphorus nanosheets (BPNSs) and gelatin were designed to kill breast cancer cells and induce adipose tissue reconstruction. The composite scaffolds were prepared by hybridizing photothermal BPNSs with porous gelatin matrices by adding pre-prepared ice particles to precisely adjust their pore structures. The composite scaffolds had large, well-interconnected spherical pores, which allowed cell migration and infiltration. Hybridization with BPNSs increased the compression strength of the scaffolds. The composite scaffolds possessed a high photothermal conversion capacity that was dependent on the amount of BPNSs. The composite scaffold with a high amount of BPNSs could completely kill breast cancer cells in vitro and in vivo under laser irradiation. Moreover, cell culture and animal experiment results showed that the composite scaffolds promoted lipid oil droplet formation and upregulated the expression of adipogenesis-related genes when hMSCs were cultured in the scaffolds. The composite scaffolds could offer a facile platform to exert anticancer effects against breast cancer cells and promote the reconstruction of adipose tissue.
Collapse
Affiliation(s)
- Linawati Sutrisno
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan; Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Huajian Chen
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan; Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Yazhou Chen
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan; Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Toru Yoshitomi
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
| | - Naoki Kawazoe
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
| | - Yingnan Yang
- Graduate School of Life and Environmental Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572, Japan
| | - Guoping Chen
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan; Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
| |
Collapse
|
18
|
Overcoming functional challenges in autologous and engineered fat grafting trends. Trends Biotechnol 2021; 40:77-92. [PMID: 34016480 DOI: 10.1016/j.tibtech.2021.04.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/06/2021] [Accepted: 04/06/2021] [Indexed: 12/11/2022]
Abstract
Autologous fat grafting offers significant promise for the repair of soft tissue deformities; however, high resorption rates indicate that engineered solutions are required to improve adipose tissue (AT) survival. Advances in material development and biofabrication have laid the foundation for the generation of functional AT constructs; however, a balance needs to be struck between clinically feasible delivery and improved structural integrity of the grafts. A new approach combining the objectives from both the clinical and research communities will assist in developing morphologically and genetically mature AT constructs, with controlled spatial arrangement and increased potential for neovascularization. In a rapidly progressing field, this review addresses research in both the preclinical and bioengineering domains and assesses their ability to resolve functional challenges.
Collapse
|
19
|
Improved chondrogenic performance with protective tracheal design of Chitosan membrane surrounding 3D-printed trachea. Sci Rep 2021; 11:9258. [PMID: 33927302 PMCID: PMC8085235 DOI: 10.1038/s41598-021-88830-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/09/2021] [Indexed: 12/04/2022] Open
Abstract
In recent tracheal tissue engineering, limitations in cartilage reconstruction, caused by immature delivery of chondrocyte-laden components, have been reported beyond the complete epithelialization and integration of the tracheal substitutes with the host tissue. In an attempt to overcome such limitations, this article introduces a protective design of tissue-engineered trachea (TraCHIM) composed of a chitosan-based nanofiber membrane (CHIM) and a 3D-printed biotracheal construct. The CHIM was created from chitosan and polycaprolactone (PCL) using an electrospinning process. Upon addition of chitosan to PCL, the diameter of electrospun fibers became thinner, allowing them to be stacked more closely, thereby improving its mechanical properties. Chitosan also enhances the hydrophilicity of the membranes, preventing them from slipping and delaminating over the cell-laden bioink of the biotracheal graft, as well as protecting the construct. Two weeks after implantation in Sprague–Dawley male rats, the group with the TraCHIM exhibited a higher number of chondrocytes, with enhanced chondrogenic performance, than the control group without the membrane. This study successfully demonstrates enhanced chondrogenic performance of TraCHIM in vivo. The protective design of TraCHIM opens a new avenue in engineered tissue research, which requires faster tissue formation from 3D biodegradable materials, to achieve complete replacement of diseased tissue.
Collapse
|
20
|
Yang J, Zhou C, Fu J, Yang Q, He T, Tan Q, Lv Q. In situ Adipogenesis in Biomaterials Without Cell Seeds: Current Status and Perspectives. Front Cell Dev Biol 2021; 9:647149. [PMID: 33763426 PMCID: PMC7982583 DOI: 10.3389/fcell.2021.647149] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/08/2021] [Indexed: 02/05/2023] Open
Abstract
For cosmetic and reconstructive purposes in the setting of small-volume adipose tissue damage due to aging, traumatic defects, oncological resections, and degenerative diseases, the current strategies for soft tissue replacement involve autologous fat grafts and tissue fillers with synthetic, bioactive, or tissue-engineered materials. However, they all have drawbacks such as volume shrinkage and foreign-body responses. Aiming to regenerate bioactive vascularized adipose tissue on biomaterial scaffolds, adipose tissue engineering (ATE) has emerged as a suitable substitute for soft tissue repair. The essential components of ATE include scaffolds as support, cells as raw materials for fat formation, and a tolerant local environment to allow regeneration to occur. The commonly loaded seeding cells are adipose-derived stem cells (ASCs), which are expected to induce stable and predictable adipose tissue formation. However, defects in stem cell enrichment, such as donor-site sacrifice, limit their wide application. As a promising alternative approach, cell-free bioactive scaffolds recruit endogenous cells for adipogenesis. In biomaterials without cell seeds, the key to sufficient adipogenesis relies on the recruitment of endogenous host cells and continuous induction of cell homing to scaffolds. Regeneration, rather than repair, is the fundamental dominance of an optimal mature product. To induce in situ adipogenesis, many researchers have focused on the mechanical and biochemical properties of scaffolds. In addition, efforts to regulate an angiogenic and adipogenic microenvironment in cell-free settings involve integrating growth factors or extracellular matrix (ECM) proteins onto bioactive scaffolds. Despite the theoretical feasibility and encouraging results in animal models, few of the reported cell-free biomaterials have been tested in humans, and failures of decellularized adipose tissues in adipogenesis have also been reported. In these cases, the most likely reason was the lack of supporting vasculature. This review summarizes the current status of biomaterials without cell seeds. Related mechanisms and influencing factors of in situ adipogenesis in cell-free biomaterials, dilemma in the development of biomaterials, and future perspectives are also addressed.
Collapse
Affiliation(s)
- Jiqiao Yang
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Tumor Targeted and Immune Therapy, Clinical Research Center for Breast Disease, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chen Zhou
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jingyang Fu
- West China School of Medicine/West China Hospital, Sichuan University, Chengdu, China
| | - Qianru Yang
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Tao He
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Qiuwen Tan
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qing Lv
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Yoo B, Kim S, Shin BH, Lee MH, Choy YB, Lee K, Heo CY, Koh WG. Preparation of alginate hydrogel with human-derived adipose tissue to improve fat graft survival and adipogenesis. J IND ENG CHEM 2021. [DOI: 10.1016/j.jiec.2020.12.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
22
|
Patil R, Rajput A, Dewani M, Mehta S, Ahamad N, Banerjee R. Hydrogel-Based Therapies for Cardiovascular Diseases. ENGINEERING MATERIALS FOR STEM CELL REGENERATION 2021:399-427. [DOI: 10.1007/978-981-16-4420-7_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
23
|
Bonetti L, De Nardo L, Farè S. Thermo-Responsive Methylcellulose Hydrogels: From Design to Applications as Smart Biomaterials. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:486-513. [DOI: 10.1089/ten.teb.2020.0202] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Lorenzo Bonetti
- Department of Chemistry, Materials and Chemical Engineering “G. Natta,” Politecnico di Milano, Milan, Italy
| | - Luigi De Nardo
- Department of Chemistry, Materials and Chemical Engineering “G. Natta,” Politecnico di Milano, Milan, Italy
- INSTM, National Interuniversity Consortium of Materials Science and Technology, Florence, Italy
| | - Silvia Farè
- Department of Chemistry, Materials and Chemical Engineering “G. Natta,” Politecnico di Milano, Milan, Italy
- INSTM, National Interuniversity Consortium of Materials Science and Technology, Florence, Italy
| |
Collapse
|
24
|
Xing H, Lee H, Luo L, Kyriakides TR. Extracellular matrix-derived biomaterials in engineering cell function. Biotechnol Adv 2020; 42:107421. [PMID: 31381963 PMCID: PMC6995418 DOI: 10.1016/j.biotechadv.2019.107421] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 07/12/2019] [Accepted: 07/31/2019] [Indexed: 12/11/2022]
Abstract
Extracellular matrix (ECM) derived components are emerging sources for the engineering of biomaterials that are capable of inducing desirable cell-specific responses. This review explores the use of biomaterials derived from naturally occurring ECM proteins and their derivatives in approaches that aim to regulate cell function. Biomaterials addressed are grouped into six categories: purified single ECM proteins, combinations of purified ECM proteins, cell-derived ECM, tissue-derived ECM, diseased and modified ECM, and ECM-polymer coupled biomaterials. Purified ECM proteins serve as a material coating for enhanced cell adhesion and biocompatibility. Cell-derived and tissue-derived ECM, generated by cell isolation and decellularization technologies, can capture the native state of the ECM environment and guide cell migration and alignment patterns as well as stem cell differentiation. We focus primarily on recent advances in the fields of soft tissue, cardiac, and dermal repair, and explore the utilization of ECM proteins as biomaterials to engineer cell responses.
Collapse
Affiliation(s)
- Hao Xing
- Department of Biomedical Engineering, Yale University, United States of America
| | - Hudson Lee
- Department of Molecular Biophysics and Biochemistry, Yale University, United States of America
| | - Lijing Luo
- Department of Pathology, Yale University, United States of America
| | - Themis R Kyriakides
- Department of Biomedical Engineering, Yale University, United States of America; Department of Pathology, Yale University, United States of America.
| |
Collapse
|
25
|
McLuckie M, Robotti F, Sanchez-Macedo N, Enderlin D, Frese L, Cheng PF, Levesque MP, Egaña JT, Poulikakos D, Ferrari A, Lindenblatt N. Lipoconstruct surface topography grating size influences vascularization onset in the dorsal skinfold chamber model. Acta Biomater 2020; 106:136-144. [PMID: 32044460 DOI: 10.1016/j.actbio.2020.01.050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/10/2020] [Accepted: 01/31/2020] [Indexed: 12/14/2022]
Abstract
After skin tissue injury or pathological removal, vascularization timing is paramount in graft survival. As full thickness skin grafts often fail to become perfused over larger surfaces, split-thickness grafts are preferred and can be used together with biomaterials, which themselves are non-angiogenic. One way of promoting vascular ingrowth is to "pre-vascularize" an engineered substitute by introducing endothelial cells (ECs). Since it has been previously demonstrated that surface structured biomaterials have an effect on wound healing, skin regeneration, and fibrosis reduction, we proposed that a microvascular-rich lipoconstruct with anisotropic topographical cues could be a clinically translatable vascularization approach. Murine lipofragments were formed with three polydimethylsiloxane molds (flat, 5 µm, and 50 µm parallel gratings) and implanted into the dorsal skinfold chamber of male C57BL/6 mice. Vascular ingrowth was observed through intravital microscopy over 21 days and further assessed by histology and protein identification. Our investigation revealed that topographical feature size influenced the commencement of neovascular ingrowth, with 5 µm gratings exhibiting early construct perfusion at 3 days post-operation, and 50 µm being delayed until day 5. We therefore postulate that surface structured lipoconstructs may serve as an easily obtained and produced construct suitable for providing soft tissue and ECs to tissue defects. STATEMENT OF SIGNIFICANCE: Skin graft failures due to inadequate or uneven perfusion frequently occur and can be even more complicated in deep, difficult to heal wounds, or bone coverage. In complex injuries, biomaterials are often used to cover bone structures with a standard split thickness graft; however, perfusion can take up to 3 weeks. Thus, any means to promote faster and uniform vascularization could significantly reduce healing time, as well as lower patient down-time. As pre-vascularized constructs have reported success in research, we created a cost-efficient, translatable method with no additional laboratory time as adipose tissue can be harvested and used immediately. We further used surface topography as an aspect to modulate construct perfusion, which has been reported for the first time here.
Collapse
|
26
|
Jung YJ, Kim HK, Cho Y, Choi JS, Woo CH, Lee KS, Sul JH, Lee CM, Han J, Park JH, Jo DG, Cho YW. Cell reprogramming using extracellular vesicles from differentiating stem cells into white/beige adipocytes. SCIENCE ADVANCES 2020; 6:eaay6721. [PMID: 32232152 PMCID: PMC7096171 DOI: 10.1126/sciadv.aay6721] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 01/03/2020] [Indexed: 05/18/2023]
Abstract
Stem cell-derived extracellular vesicles (EVs) offer alternative approaches to stem cell-based therapy for regenerative medicine. In this study, stem cell EVs derived during differentiation are developed to use as cell-free therapeutic systems by inducing tissue-specific differentiation. EVs are isolated from human adipose-derived stem cells (HASCs) during white and beige adipogenic differentiation (D-EV and BD-EV, respectively) via tangential flow filtration. D-EV and BD-EV can successfully differentiate HASCs into white and beige adipocytes, respectively. D-EV are transplanted with collagen/methylcellulose hydrogels on the backs of BALB/c mice, and they produce numerous lipid droplets in injected sites. Treatments of BD-EV attenuate diet-induced obesity through browning of adipose tissue in mice. Furthermore, high-fat diet-induced hepatic steatosis and glucose tolerance are improved by BD-EV treatment. miRNAs are responsible for the observed effects of BD-EV. These results reveal that secreted EVs during stem cell differentiation into white adipocytes or beige adipocytes can promote cell reprogramming.
Collapse
Affiliation(s)
- Youn Jae Jung
- Department of Materials Science and Chemical Engineering, Hanyang University ERICA, Ansan 15588, Republic of Korea
- ExoStemTech Inc., Ansan 15588, Republic of Korea
| | - Hark Kyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yoonsuk Cho
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ji Suk Choi
- Department of Materials Science and Chemical Engineering, Hanyang University ERICA, Ansan 15588, Republic of Korea
- ExoStemTech Inc., Ansan 15588, Republic of Korea
| | - Chang Hee Woo
- Department of Materials Science and Chemical Engineering, Hanyang University ERICA, Ansan 15588, Republic of Korea
- ExoStemTech Inc., Ansan 15588, Republic of Korea
| | - Kyoung Soo Lee
- Department of Materials Science and Chemical Engineering, Hanyang University ERICA, Ansan 15588, Republic of Korea
| | - Jae Hoon Sul
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Chan Mi Lee
- ExoStemTech Inc., Ansan 15588, Republic of Korea
| | - Jihoon Han
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jae Hyung Park
- ExoStemTech Inc., Ansan 15588, Republic of Korea
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Dong-Gyu Jo
- ExoStemTech Inc., Ansan 15588, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Corresponding author. (D.-G.J.); (Y.W.C.)
| | - Yong Woo Cho
- Department of Materials Science and Chemical Engineering, Hanyang University ERICA, Ansan 15588, Republic of Korea
- ExoStemTech Inc., Ansan 15588, Republic of Korea
- Corresponding author. (D.-G.J.); (Y.W.C.)
| |
Collapse
|
27
|
Giatsidis G, Succar J, Waters TD, Liu W, Rhodius P, Wang C, Nilsen TJ, Chnari E, Orgill DP. Tissue-Engineered Soft-Tissue Reconstruction Using Noninvasive Mechanical Preconditioning and a Shelf-Ready Allograft Adipose Matrix. Plast Reconstr Surg 2019; 144:884-895. [PMID: 31568297 DOI: 10.1097/prs.0000000000006085] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Adipose tissue defects leading to severe functional (disability) and morphologic (disfigurement) morbidity are often treated in plastic surgery with fat grafting, which can be limited by resorption, necrosis, and cyst formation. This study aimed to assess whether adipose scaffolds could provide an environment for in situ autologous fat grafting, and to study whether adipose cell migration and proliferation (adipogenesis) within scaffolds could be enhanced by preliminarily increasing the vascularity (preconditioning) of the surrounding tissue receiving the scaffolds. METHODS Using an established rodent model of subcutaneous tissue/scaffold grafting, the authors tested the potential of a human-derived, shelf-ready, injectable, decellularized allograft adipose matrix to reconstruct soft-tissue defects when used in combination with noninvasive mechanical (suction-induced) skin preconditioning. RESULTS Combined use of the allograft adipose matrix and noninvasive skin preconditioning significantly improved long-term volume retention (50 to 80 percent higher at a 12-week follow-up) and histologic quality of reconstructed tissues compared with standard of care (autologous adipose grafts). The components of the allograft adipose matrix supported adipogenesis and angiogenesis. Combining the allograft adipose matrix with living adipose grafts mitigated negative outcomes (lower long-term volume retention, higher presence of cystic-like areas). CONCLUSIONS This study suggests that the synergistic use of the allograft adipose matrix and noninvasive tissue preconditioning provides an effective solution for improving fat grafting. These strategies can easily be tested in clinical trials and could establish the basis for a novel therapeutic paradigm in reconstructive surgery.
Collapse
Affiliation(s)
- Giorgio Giatsidis
- From the Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School; Department of Molecular Medicine, University of Padova; Preventive Medicine, University of New Mexico; Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; and the Musculoskeletal Transplant Foundation
| | - Julien Succar
- From the Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School; Department of Molecular Medicine, University of Padova; Preventive Medicine, University of New Mexico; Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; and the Musculoskeletal Transplant Foundation
| | - Trevon D Waters
- From the Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School; Department of Molecular Medicine, University of Padova; Preventive Medicine, University of New Mexico; Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; and the Musculoskeletal Transplant Foundation
| | - Wenyue Liu
- From the Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School; Department of Molecular Medicine, University of Padova; Preventive Medicine, University of New Mexico; Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; and the Musculoskeletal Transplant Foundation
| | - Patrick Rhodius
- From the Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School; Department of Molecular Medicine, University of Padova; Preventive Medicine, University of New Mexico; Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; and the Musculoskeletal Transplant Foundation
| | - Chenglong Wang
- From the Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School; Department of Molecular Medicine, University of Padova; Preventive Medicine, University of New Mexico; Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; and the Musculoskeletal Transplant Foundation
| | - Todd J Nilsen
- From the Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School; Department of Molecular Medicine, University of Padova; Preventive Medicine, University of New Mexico; Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; and the Musculoskeletal Transplant Foundation
| | - Evangelia Chnari
- From the Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School; Department of Molecular Medicine, University of Padova; Preventive Medicine, University of New Mexico; Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; and the Musculoskeletal Transplant Foundation
| | - Dennis P Orgill
- From the Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School; Department of Molecular Medicine, University of Padova; Preventive Medicine, University of New Mexico; Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; and the Musculoskeletal Transplant Foundation
| |
Collapse
|
28
|
Zhou S, Chen S, Jiang Q, Pei M. Determinants of stem cell lineage differentiation toward chondrogenesis versus adipogenesis. Cell Mol Life Sci 2019; 76:1653-1680. [PMID: 30689010 PMCID: PMC6456412 DOI: 10.1007/s00018-019-03017-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 12/10/2018] [Accepted: 01/15/2019] [Indexed: 12/12/2022]
Abstract
Adult stem cells, also termed as somatic stem cells, are undifferentiated cells, detected among differentiated cells in a tissue or an organ. Adult stem cells can differentiate toward lineage specific cell types of the tissue or organ in which they reside. They also have the ability to differentiate into mature cells of mesenchymal tissues, such as cartilage, fat and bone. Despite the fact that the balance has been comprehensively scrutinized between adipogenesis and osteogenesis and between chondrogenesis and osteogenesis, few reviews discuss the relationship between chondrogenesis and adipogenesis. In this review, the developmental and transcriptional crosstalk of chondrogenic and adipogenic lineages are briefly explored, followed by elucidation of signaling pathways and external factors guiding lineage determination between chondrogenic and adipogenic differentiation. An in-depth understanding of overlap and discrepancy between these two mesenchymal tissues in lineage differentiation would benefit regeneration of high-quality cartilage tissues and adipose tissues for clinical applications.
Collapse
Affiliation(s)
- Sheng Zhou
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA
- Department of Sports Medicine and Adult Reconstructive Surgery, School of Medicine, Drum Tower Hospital, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Song Chen
- Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, 610083, Sichuan, People's Republic of China
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstructive Surgery, School of Medicine, Drum Tower Hospital, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA.
- Robert C. Byrd Health Sciences Center, WVU Cancer Institute, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
29
|
Aulthouse AL, Freeh E, Newstead S, Stockert AL. Part 1: A Novel Model for Three-Dimensional Culture of 3T3-L1 Preadipocytes Stimulates Spontaneous Cell Differentiation Independent of Chemical Induction Typically Required in Monolayer. Nutr Metab Insights 2019; 12:1178638819841399. [PMID: 31001061 PMCID: PMC6454649 DOI: 10.1177/1178638819841399] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 02/08/2019] [Indexed: 12/13/2022] Open
Abstract
Differences in monolayer and three-dimensional (3D) culture systems have been recognized for several years. Despite the recognized importance of 3D systems, low cost and convenience of monolayer culture are still readily used for metabolic and nutritional studies. Here, we present part 1 of a 2-part series that will highlight (1) a novel and cost-effective model for culturing 3T3-L1 preadipocytes in 3D agarose as well as (2) an initial study showing the successful use of this 3D model for experimental analysis of these cells treated with cinnamon extract while suspended in agarose. In part 1, we provide a full characterization of the model system for the 3T3-L1 cells that demonstrate the functionality and convenience of this system. Importantly, we note spontaneous differentiation to adipocytes while cultured under these methods, independent of chemical induction. We present a 2.5-week time course with rounded cells forming vacuoles as early as 24 hours and accumulation of lipid detectable by Oil Red O stain at 0.5 weeks. Serum selection, lipid volume determination, and cell size are characterized. We conclusively demonstrate adipogenesis based on a peroxisome proliferator-activated receptor γ (PPARγ) detection using immunohistochemistry (IHC) of sections from these 3D cultures. Methods, materials and recommendations are described as well as proposed benefits to the use of this culture system for 3T3-L1 cells.
Collapse
Affiliation(s)
- Amy L Aulthouse
- Department of Biological and Allied
Health Sciences, Ohio Northern University, Ada, OH, USA
| | - Ellen Freeh
- Department of Biological and Allied
Health Sciences, Ohio Northern University, Ada, OH, USA
| | - Sabrina Newstead
- Department of Biological and Allied
Health Sciences, Ohio Northern University, Ada, OH, USA
| | - Amy L Stockert
- Department of Pharmaceutical and
Biomedical Sciences, Ohio Northern University, Ada, OH, USA
| |
Collapse
|
30
|
O'Halloran NA, Dolan EB, Kerin MJ, Lowery AJ, Duffy GP. Hydrogels in adipose tissue engineering-Potential application in post-mastectomy breast regeneration. J Tissue Eng Regen Med 2018; 12:2234-2247. [PMID: 30334613 DOI: 10.1002/term.2753] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 10/02/2018] [Accepted: 10/15/2018] [Indexed: 12/11/2022]
Abstract
Current methods of breast reconstruction are associated with significant shortcomings, including capsular contracture, infection, rupture, the need for reoperation in implant-based reconstruction, and donor site morbidity in autologous reconstruction. These limitations result in severe physical and psychological issues for breast cancer patients. Recently, research has moved into the field of adipose tissue engineering to overcome these limitations. A wide range of regenerative strategies has been devised utilising various scaffold designs and biomaterials. A scaffold capable of providing appropriate biochemical and biomechanical cues for adipogenesis is required. Hydrogels have been widely studied for their suitability for adipose tissue regeneration and are advantageous secondary to their ability to accurately imitate the native extracellular matrix. The aim of this review was to analyse the use of hydrogel scaffolds in the field of adipose tissue engineering.
Collapse
Affiliation(s)
- Niamh A O'Halloran
- Discipline of Surgery, The Lambe Institute, National University of Ireland Galway, Galway, Ireland
| | - Eimear B Dolan
- School of Pharmacy, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,Discipline of Anatomy, School of Medicine, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Michael J Kerin
- Discipline of Surgery, The Lambe Institute, National University of Ireland Galway, Galway, Ireland
| | - Aoife J Lowery
- Discipline of Surgery, The Lambe Institute, National University of Ireland Galway, Galway, Ireland
| | - Garry P Duffy
- Discipline of Anatomy, School of Medicine, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
31
|
Argentati C, Morena F, Bazzucchi M, Armentano I, Emiliani C, Martino S. Adipose Stem Cell Translational Applications: From Bench-to-Bedside. Int J Mol Sci 2018; 19:E3475. [PMID: 30400641 PMCID: PMC6275042 DOI: 10.3390/ijms19113475] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/22/2018] [Accepted: 11/01/2018] [Indexed: 02/08/2023] Open
Abstract
During the last five years, there has been a significantly increasing interest in adult adipose stem cells (ASCs) as a suitable tool for translational medicine applications. The abundant and renewable source of ASCs and the relatively simple procedure for cell isolation are only some of the reasons for this success. Here, we document the advances in the biology and in the innovative biotechnological applications of ASCs. We discuss how the multipotential property boosts ASCs toward mesenchymal and non-mesenchymal differentiation cell lineages and how their character is maintained even if they are combined with gene delivery systems and/or biomaterials, both in vitro and in vivo.
Collapse
Affiliation(s)
- Chiara Argentati
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
| | - Martina Bazzucchi
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
| | - Ilaria Armentano
- Department of Ecological and Biological Sciences, Tuscia University Largo dell'Università, snc, 01100 Viterbo, Italy.
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
- CEMIN, Center of Excellence on Nanostructured Innovative Materials, Via del Giochetto, 06126 Perugia, Italy.
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
- CEMIN, Center of Excellence on Nanostructured Innovative Materials, Via del Giochetto, 06126 Perugia, Italy.
| |
Collapse
|
32
|
Li S, Poche JN, Liu Y, Scherr T, McCann J, Forghani A, Smoak M, Muir M, Berntsen L, Chen C, Ravnic DJ, Gimble J, Hayes DJ. Hybrid Synthetic-Biological Hydrogel System for Adipose Tissue Regeneration. Macromol Biosci 2018; 18:e1800122. [PMID: 30247815 DOI: 10.1002/mabi.201800122] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 08/12/2018] [Indexed: 01/28/2023]
Abstract
Hydrogels are promising scaffolds for adipose tissue regeneration. Currently, the incorporation of bioactive molecules in hydrogel system is used, which can increase the cell proliferation rate or improve adipogenic differentiation performance of stromal stem cells but often suffers from high expense or cytotoxicity because of light/thermal curing used for polymerization. In this study, decellularized adipose tissue is incorporated, at varying concentrations, with a thiol-acrylate fraction that is then polymerized to produce hydrogels via a Michael addition reaction. The results reveal that the major component of isolated adipose-derived extracellular matrix (ECM) is Collagen I. Mechanical properties of ECM polyethylene glycol (PEG) are not negatively affected by the incorporation of ECM. Additionally, human adipose-derived stem cells (hASCs) are encapsulated in ECM PEG hydrogel with ECM concentrations varying from 0% to 1%. The results indicate that hASCs maintained the highest viability and proliferation rate in 1% ECM PEG hydrogel with most lipids formation when cultured in adipogenic conditions. Furthermore, more adipose regeneration is observed in 1% ECM group with in vivo study by Day 14 compared to other ECM PEG hydrogels with lower ECM content. Taken together, these findings suggest the ECM PEG hydrogel is a promising substitute for adipose tissue regeneration applications.
Collapse
Affiliation(s)
- Shue Li
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Department of Biomedical Engineering, Pennsylvania State University, Millennium Science Complex, University Park, PA, 16802, USA
| | | | - Yiming Liu
- Department of Biomedical Engineering, Pennsylvania State University, Millennium Science Complex, University Park, PA, 16802, USA
| | - Thomas Scherr
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA
| | - Jacob McCann
- Department of Biomedical Engineering, Pennsylvania State University, Millennium Science Complex, University Park, PA, 16802, USA
| | - Anoosha Forghani
- Department of Biomedical Engineering, Pennsylvania State University, Millennium Science Complex, University Park, PA, 16802, USA
| | - Mollie Smoak
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Mitchell Muir
- Department of Biological Engineering, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Lisa Berntsen
- Department of Biomedical Engineering, Pennsylvania State University, Millennium Science Complex, University Park, PA, 16802, USA
| | - Cong Chen
- Department of Biomedical Engineering, Pennsylvania State University, Millennium Science Complex, University Park, PA, 16802, USA
| | - Dino J Ravnic
- Division of Plastic Surgery, Department of Surgery, PennState Health Milton S. Hershey Medical Center, Hershey, PA, 17033, USA
| | - Jeffrey Gimble
- Department of Medicine and Surgery, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Daniel J Hayes
- Department of Biomedical Engineering, Pennsylvania State University, Millennium Science Complex, University Park, PA, 16802, USA
| |
Collapse
|
33
|
Tan QW, Tang SL, Zhang Y, Yang JQ, Wang ZL, Xie HQ, Lv Q. Hydrogel from Acellular Porcine Adipose Tissue Accelerates Wound Healing by Inducing Intradermal Adipocyte Regeneration. J Invest Dermatol 2018; 139:455-463. [PMID: 30195900 DOI: 10.1016/j.jid.2018.08.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 08/03/2018] [Accepted: 08/09/2018] [Indexed: 02/09/2023]
Abstract
As an important component of the skin, intradermal adipocytes are closely associated with skin homeostasis and wound healing. Although studies have focused on the role of fibroblasts, keratinocytes, and inflammatory cells in wound healing, the role of adipocytes has not been fully investigated. Here, we verified whether the induction of adipocyte regeneration in a wound bed can effectively promote wound healing, finding that the hydrogel from acellular porcine adipose tissue in combination with adipose-derived stem cells can induce in situ adipogenesis in the wound microenvironment. The newly regenerated adipocytes enhanced fibroblast migration, accelerated wound closing, and enhanced wound epithelialization. More importantly, newly formed intact skin structure was observed after treating the wound with adipose-derived stem cell-loaded hydrogel from acellular porcine adipose tissue. These results show that hydrogel from acellular porcine adipose tissue might substantially improve re-epithelialization, angiogenesis, and skin-appendage regeneration, making it a promising therapeutic biomaterial for skin wound healing.
Collapse
Affiliation(s)
- Qiu-Wen Tan
- Department of Breast Surgery, Clinical Research Center for Breast, West China Hospital, Sichuan University, Sichuan, China; Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, China
| | - Shen-Li Tang
- Department of Breast Surgery, Clinical Research Center for Breast, West China Hospital, Sichuan University, Sichuan, China
| | - Yi Zhang
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, China
| | - Ji-Qiao Yang
- Department of Breast Surgery, Clinical Research Center for Breast, West China Hospital, Sichuan University, Sichuan, China
| | - Zhu-Le Wang
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, China
| | - Hui-Qi Xie
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, China.
| | - Qing Lv
- Department of Breast Surgery, Clinical Research Center for Breast, West China Hospital, Sichuan University, Sichuan, China.
| |
Collapse
|
34
|
Dong J, Yu M, Zhang Y, Yin Y, Tian W. Recent developments and clinical potential on decellularized adipose tissue. J Biomed Mater Res A 2018; 106:2563-2574. [PMID: 29664222 DOI: 10.1002/jbm.a.36435] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/20/2018] [Accepted: 04/10/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Jia Dong
- State Key Laboratory of Oral Disease; West China Hospital of Stomatology, Sichuan University; Chengdu China
- National Clinical Research Center for Oral Diseases; West China Hospital of Stomatology, Sichuan University; Chengdu China
- National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology, Sichuan University; Chengdu China
- Department of Oral and Maxillofacial Surgery; West China Hospital of Stomatology, Sichuan University; Chengdu China
| | - Mei Yu
- State Key Laboratory of Oral Disease; West China Hospital of Stomatology, Sichuan University; Chengdu China
- National Clinical Research Center for Oral Diseases; West China Hospital of Stomatology, Sichuan University; Chengdu China
- National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology, Sichuan University; Chengdu China
- Department of Oral and Maxillofacial Surgery; West China Hospital of Stomatology, Sichuan University; Chengdu China
| | - Yan Zhang
- State Key Laboratory of Oral Disease; West China Hospital of Stomatology, Sichuan University; Chengdu China
- National Clinical Research Center for Oral Diseases; West China Hospital of Stomatology, Sichuan University; Chengdu China
- National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology, Sichuan University; Chengdu China
- Department of Oral and Maxillofacial Surgery; West China Hospital of Stomatology, Sichuan University; Chengdu China
| | - Yin Yin
- State Key Laboratory of Oral Disease; West China Hospital of Stomatology, Sichuan University; Chengdu China
- National Clinical Research Center for Oral Diseases; West China Hospital of Stomatology, Sichuan University; Chengdu China
- National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology, Sichuan University; Chengdu China
- Department of Oral and Maxillofacial Surgery; West China Hospital of Stomatology, Sichuan University; Chengdu China
| | - Weidong Tian
- State Key Laboratory of Oral Disease; West China Hospital of Stomatology, Sichuan University; Chengdu China
- National Clinical Research Center for Oral Diseases; West China Hospital of Stomatology, Sichuan University; Chengdu China
- National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology, Sichuan University; Chengdu China
- Department of Oral and Maxillofacial Surgery; West China Hospital of Stomatology, Sichuan University; Chengdu China
| |
Collapse
|
35
|
Xia P, Zhang K, Gong Y, Li G, Yan S, Yin J. Injectable Stem Cell Laden Open Porous Microgels That Favor Adipogenesis: In Vitro and in Vivo Evaluation. ACS APPLIED MATERIALS & INTERFACES 2017; 9:34751-34761. [PMID: 28930432 DOI: 10.1021/acsami.7b13065] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Microgels, with large surface area per volume, show great advantages in adipose tissue engineering due to their injectability and similarity with natural extracellular matrix. However, to date, no studies have tried applying microgels to adipose tissue regeneration. Herein, based on double-bonded poly(l-glutamic acid)-g-2-hydroxyethyl methacrylate (PLGA-g-HEMA) and maleic anhydride-modified chitosan (MCS), an open porous microgel with high hydrophilicity and great injectability is successfully prepared (microgels diameters of 200-300 μm, pore diameter of 38 μm, and porosity of 88.3%). The storage modulus of 30 mg/mL of the microgel dispersions is 2000 Pa, which is similar to that of the native adipose tissue. The spheroidal stem cell shape and extensive cell-cell connections can be formed in the present microgels to promote adipogenic differentiation and realize adipose tissue regeneration. After injection in vitro, the microgels can maintain high stem cell viability up to 14 days. The extensive Oil red O staining is observed after adipogenic induction for 14 days. After 12 weeks postimplantation, adipose tissues can be regenerated well. Blood vessels are formed in the neogenerated tissues. The degradation rate of microgels roughly matches with the adipose tissue formation rate. The study offers an applicable microgel system to boost the adipose tissue regeneration.
Collapse
Affiliation(s)
- Pengfei Xia
- Department of Polymer Materials, Shanghai University , 99 Shangda Road, Shanghai 200444, People's Republic of China
| | - Kunxi Zhang
- Department of Polymer Materials, Shanghai University , 99 Shangda Road, Shanghai 200444, People's Republic of China
| | - Yan Gong
- Department of Polymer Materials, Shanghai University , 99 Shangda Road, Shanghai 200444, People's Republic of China
| | - Guifei Li
- Department of Polymer Materials, Shanghai University , 99 Shangda Road, Shanghai 200444, People's Republic of China
| | - Shifeng Yan
- Department of Polymer Materials, Shanghai University , 99 Shangda Road, Shanghai 200444, People's Republic of China
| | - Jingbo Yin
- Department of Polymer Materials, Shanghai University , 99 Shangda Road, Shanghai 200444, People's Republic of China
| |
Collapse
|
36
|
O’Halloran N, Courtney D, Kerin MJ, Lowery AJ. Adipose-Derived Stem Cells in Novel Approaches to Breast Reconstruction: Their Suitability for Tissue Engineering and Oncological Safety. Breast Cancer (Auckl) 2017; 11:1178223417726777. [PMID: 29104428 PMCID: PMC5562338 DOI: 10.1177/1178223417726777] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 07/13/2017] [Indexed: 12/13/2022] Open
Abstract
Adipose-derived stem cells (ADSCs) are rapidly becoming the gold standard cell source for tissue engineering strategies and hold great potential for novel breast reconstruction strategies. However, their use in patients with breast cancer is controversial and their oncological safety, particularly in relation to local disease recurrence, has been questioned. In vitro, in vivo, and clinical studies using ADSCs report conflicting data on their suitability for adipose tissue regeneration in patients with cancer. This review aims to provide an overview of the potential role for ADSCs in breast reconstruction and to examine the evidence relating to the oncologic safety of their use in patients with breast cancer.
Collapse
Affiliation(s)
- Niamh O’Halloran
- Discipline of Surgery, Lambe Institute for Translational Research, National University of Ireland, Galway, Galway, Ireland
| | - Donald Courtney
- Discipline of Surgery, Lambe Institute for Translational Research, National University of Ireland, Galway, Galway, Ireland
| | - Michael J Kerin
- Discipline of Surgery, Lambe Institute for Translational Research, National University of Ireland, Galway, Galway, Ireland
| | - Aoife J Lowery
- Graduate Entry Medical School, University of Limerick, Limerick, Ireland
| |
Collapse
|