51
|
Tamai M, Adachi E, Kawase M, Tagawa YI. Syngeneic implantation of mouse hepatic progenitor cell-derived three-dimensional liver tissue with dense collagen fibrils. World J Gastroenterol 2022; 28:1444-1454. [PMID: 35582675 PMCID: PMC9048472 DOI: 10.3748/wjg.v28.i14.1444] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/10/2021] [Accepted: 09/02/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Liver transplantation is a therapy for irreversible liver failure; however, at present, donor organs are in short supply. Cell transplantation therapy for liver failure is still at the developmental stage and is critically limited by a shortage of human primary hepatocytes.
AIM To investigate the possibility that hepatic progenitor cells (HPCs) prepared from the portal branch-ligated hepatic lobe may be used in regenerative medicine, we attempted to enable the implantation of extracellular matrices containing organoids consisting of HPC-derived hepatocytes and non-parenchymal cells.
METHODS In vitro liver organoid tissue has been generated by accumulating collagen fibrils, fibroblasts, and HPCs on a mesh of polylactic acid fabric using a bioreactor; this was subsequently implanted into syngeneic wild-type mice.
RESULTS The in vitro liver organoid tissues generated transplantable tissues in the condensed collagen fibril matrix and were obtained from the mouse through partial hepatectomy.
CONCLUSION Liver organoid tissue was produced from expanded HPCs using an originally designed bioreactor system. This tissue was comparable to liver lobules, and with fibroblasts embedded in the network collagen fibrils of this artificial tissue, it is useful for reconstructing the hepatic interstitial structure.
Collapse
Affiliation(s)
- Miho Tamai
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama-shi 226-8501, Japan
- Faculty of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan
| | - Eijiro Adachi
- Department of Molecular Morphology, Kitasato University, Yokohama-shi 319-3526, Japan
- Long-Term Care Health Facility Yasuragi, Ibaraki Zip or Postal Code, Japan
| | - Masaya Kawase
- Nagahama Institute of Bio-Science and Technology, Shiga 526-0829, Japan
| | - Yoh-ichi Tagawa
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama-shi 226-8501, Japan
| |
Collapse
|
52
|
Zhang X, Chen X, Hong H, Hu R, Liu J, Liu C. Decellularized extracellular matrix scaffolds: Recent trends and emerging strategies in tissue engineering. Bioact Mater 2022; 10:15-31. [PMID: 34901526 PMCID: PMC8637010 DOI: 10.1016/j.bioactmat.2021.09.014] [Citation(s) in RCA: 236] [Impact Index Per Article: 118.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/24/2021] [Accepted: 09/08/2021] [Indexed: 01/09/2023] Open
Abstract
The application of scaffolding materials is believed to hold enormous potential for tissue regeneration. Despite the widespread application and rapid advance of several tissue-engineered scaffolds such as natural and synthetic polymer-based scaffolds, they have limited repair capacity due to the difficulties in overcoming the immunogenicity, simulating in-vivo microenvironment, and performing mechanical or biochemical properties similar to native organs/tissues. Fortunately, the emergence of decellularized extracellular matrix (dECM) scaffolds provides an attractive way to overcome these hurdles, which mimic an optimal non-immune environment with native three-dimensional structures and various bioactive components. The consequent cell-seeded construct based on dECM scaffolds, especially stem cell-recellularized construct, is considered an ideal choice for regenerating functional organs/tissues. Herein, we review recent developments in dECM scaffolds and put forward perspectives accordingly, with particular focus on the concept and fabrication of decellularized scaffolds, as well as the application of decellularized scaffolds and their combinations with stem cells (recellularized scaffolds) in tissue engineering, including skin, bone, nerve, heart, along with lung, liver and kidney.
Collapse
Affiliation(s)
| | | | - Hua Hong
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Rubei Hu
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Jiashang Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| |
Collapse
|
53
|
Modular Bioreactor Design for Directed Tendon/Ligament Tissue Engineering. Bioengineering (Basel) 2022; 9:bioengineering9030127. [PMID: 35324816 PMCID: PMC8945228 DOI: 10.3390/bioengineering9030127] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 11/30/2022] Open
Abstract
Functional tissue-engineered tendons and ligaments remain to be prepared in a reproducible and scalable manner. This study evaluates an acellular 3D extracellular matrix (ECM) scaffold for tendon/ligament tissue engineering and their ability to support strain-induced gene regulation associated with the tenogenesis of cultured mesenchymal stromal cells. Preliminary data demonstrate unique gene regulation patterns compared to other scaffold forms, in particular in Wnt signaling. However, the need for a robust bioreactor system that minimizes process variation was also evident. A design control process was used to design and verify the functionality of a novel bioreactor. The system accommodates 3D scaffolds with clinically-relevant sizes, is capable of long-term culture with customizable mechanical strain regimens, incorporates in-line load measurement for continuous monitoring and feedback control, and allows a variety of scaffold configurations through a unique modular grip system. All critical functional specifications were met, including verification of physiological strain levels from 1–10%, frequency levels from 0.2–0.5 Hz, and accurate load measurement up to 50 N, which can be expanded on the basis of load cell capability. The design process serves as a model for establishing statistical functionality and reliability of investigative systems. This work sets the stage for detailed analyses of ECM scaffolds to identify critical differentiation signaling responses and essential matrix composition and cell–matrix interactions.
Collapse
|
54
|
Krishnan A, Wang H, MacArthur JW. Applications of Tissue Decellularization Techniques in Ventricular Myocardial Biofabrication. Front Bioeng Biotechnol 2022; 10:802283. [PMID: 35265593 PMCID: PMC8899393 DOI: 10.3389/fbioe.2022.802283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/21/2022] [Indexed: 11/13/2022] Open
Abstract
Ischemic heart disease is the leading cause of death around the world, and though the advent of coronary revascularization has revolutionized its treatment, many patients who sustain ischemic injury to the heart will go on to develop heart failure. Biofabrication of ventricular myocardium for replacement of irreversibly damaged ischemic myocardium is sought after as a potential therapy for ischemic heart failure, though challenges in reliably producing this biomaterial have limited its clinical application. One method that shows promise for generation of functional myocardium is the use of tissue decellularization to serve as a scaffold for biofabrication. This review outlines the methods, materials, challenges, and prospects of tissue decellularization techniques for ventricular myocardium biofabrication. Decellularization aims to preserve the architecture and composition of the extracellular matrix of the tissue it is applied to, allowing for the subsequent implantation of stem cells of the desired cell type. Decellularization can be achieved with multiple reagents, most of which have detergent properties. A variety of cell types can be implanted in the resulting scaffold, including cardiac progenitor cells, and embryonic or induced pluripotent stem cells to generate a range of tissue, from patches to beating myocardium. The future of this biofabrication method will likely emphasize patient specific tissue engineering to generate complex 3-dimensional constructs that can replace dysfunctional cardiac structures.
Collapse
Affiliation(s)
- Aravind Krishnan
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Hanjay Wang
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - John Ward MacArthur
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
55
|
Zhai Y, Wang Q, Zhu Z, Zheng W, Ma S, Hao Y, Yang L, Cheng G. Cell-derived extracellular matrix enhanced by collagen-binding domain-decorated exosomes to promote neural stem cells neurogenesis. Biomed Mater 2021; 17. [PMID: 34874314 DOI: 10.1088/1748-605x/ac4089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/06/2021] [Indexed: 11/11/2022]
Abstract
Enhancing neurogenesis of neural stem cells (NSCs) is crucial in stem cell therapy for neurodegenerative diseases. Within the extracellular microenvironment, extracellular matrix (ECM) plays a pivotal role in modulating cell behaviors. However, a single ECM biomaterial is not sufficient to establish an ideal microenvironment. As multifunctional nanocarriers, exosomes display tremendous advantages for the treatments of various diseases. Herein, collagen binding domain peptide-modified exosomes (CBD-Exo) were obtained from the SH-SY5Y cell line infected with lentivirus particles encoding CBD-lysosome associated membrane glycoprotein 2b (CBD-Lamp2b) to improve the binding efficiency of exosomes and ECM. An exosomes-functionalized ECM (CBD-Exo/ECM) was then constructed via the interaction between CBD and collagen in ECM. Then, CBD-Exo/ECM was employed as a carrier for NSCs culture. The results showed that CBD-Exo/ECM can support the neurogenesis of NSCs with the percentage of proliferation marker EdU-positive (35.8% ± 0.47% vs 21.9% ± 2.32%) and neuron maker Tuj-1-positive (55.8% ± 0.47% vs 30.6% ± 2.62%) were both significantly increased in the exosomes-functionalized ECM system. This exosomes-functionalized ECM was capable to promote the cell proliferation and accelerate neuronal differentiation of NSCs, providing a potential biomedical material for stem cell application in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Yuanxin Zhai
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, Anhui 230026, People's Republic of China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, Jiangsu 215123, People's Republic of China
| | - Quanwei Wang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, Jiangsu 215123, People's Republic of China.,School of Life Sciences, Shanghai University, Shanghai 200444, People's Republic of China
| | - Zhanchi Zhu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, Anhui 230026, People's Republic of China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, Jiangsu 215123, People's Republic of China
| | - Wenlong Zheng
- Suzhou Kowloon Hospital, Shanghai Jiaotong University Medical School, Suzhou, Jiangsu 215028, People's Republic of China
| | - Sancheng Ma
- Suzhou Kowloon Hospital, Shanghai Jiaotong University Medical School, Suzhou, Jiangsu 215028, People's Republic of China
| | - Ying Hao
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, Anhui 230026, People's Republic of China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, Jiangsu 215123, People's Republic of China.,Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Guangdong Branch, Foshan, Guangdong 528225, People's Republic of China
| | - Lingyan Yang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, Anhui 230026, People's Republic of China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, Jiangsu 215123, People's Republic of China.,Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Guangdong Branch, Foshan, Guangdong 528225, People's Republic of China
| | - Guosheng Cheng
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, Anhui 230026, People's Republic of China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, Jiangsu 215123, People's Republic of China.,Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Guangdong Branch, Foshan, Guangdong 528225, People's Republic of China
| |
Collapse
|
56
|
Biomaterials and Their Biomedical Applications: From Replacement to Regeneration. Processes (Basel) 2021. [DOI: 10.3390/pr9111949] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The history of biomaterials dates back to the mists of time: human beings had always used exogenous materials to facilitate wound healing and try to restore damaged tissues and organs. Nowadays, a wide variety of materials are commercially available and many others are under investigation to both maintain and restore bodily functions. Emerging clinical needs forced the development of new biomaterials, and lately discovered biomaterials allowed for the performing of new clinical applications. The definition of biomaterials as materials specifically conceived for biomedical uses was raised when it was acknowledged that they have to possess a fundamental feature: biocompatibility. At first, biocompatibility was mainly associated with biologically inert substances; around the 1970s, bioactivity was first discovered and the definition of biomaterials was consequently extended. At present, it also includes biologically derived materials and biological tissues. The present work aims at walking across the history of biomaterials, looking towards the scientific literature published on this matter. Finally, some current applications of biomaterials are briefly depicted and their future exploitation is hypothesized.
Collapse
|
57
|
Ma YH, Shi HJ, Wei QS, Deng QW, Sun JH, Liu Z, Lai BQ, Li G, Ding Y, Niu WT, Zeng YS, Zeng X. Developing a mechanically matched decellularized spinal cord scaffold for the in situ matrix-based neural repair of spinal cord injury. Biomaterials 2021; 279:121192. [PMID: 34700225 DOI: 10.1016/j.biomaterials.2021.121192] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 12/16/2022]
Abstract
Tissue engineering is a promising strategy to repair spinal cord injury (SCI). However, a bioscaffold with mechanical properties that match those of the pathological spinal cord tissue and a pro-regenerative matrix that allows robust neurogenesis for overcoming post-SCI scar formation has yet to be developed. Here, we report that a mechanically enhanced decellularized spinal cord (DSC) scaffold with a thin poly (lactic-co-glycolic acid) (PLGA) outer shell may fulfill the requirements for effective in situ neuroengineering after SCI. Using chemical extraction and electrospinning methods, we successfully constructed PLGA thin shell-ensheathed DSC scaffolds (PLGA-DSC scaffolds) in a way that removed major inhibitory components while preserving the permissive matrix. The DSCs exhibited good cytocompatibility with neural stem cells (NSCs) and significantly enhanced their differentiation toward neurons in vitro. Due to the mechanical reinforcement, the implanted PLGA-DSC scaffolds showed markedly increased resilience to infiltration by myofibroblasts and the deposition of dense collagen matrix, thereby creating a neurogenic niche favorable for the targeted migration, residence and neuronal differentiation of endogenous NSCs after SCI. Furthermore, PLGA-DSC presented a mild immunogenic property but prominent ability to polarize macrophages from the M1 phenotype to the M2 phenotype, leading to significant tissue regeneration and functional restoration after SCI. Taken together, the results demonstrate that the mechanically matched PLGA-DSC scaffolds show promise for effective tissue repair after SCI.
Collapse
Affiliation(s)
- Yuan-Huan Ma
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China; Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong Province, 510080, China; Guangdong Key Laboratory of Age-Related Cardiocerebral Diseases, Institute of Neurology, Guangdong Medical University, Zhanjiang, Guangdong Province, 524023, China; Guangzhou Institute of Clinical Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong Province, 510180, PR China
| | - Hui-Juan Shi
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong Province, 510080, China
| | - Qing-Shuai Wei
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong Province, 510080, China
| | - Qing-Wen Deng
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China
| | - Jia-Hui Sun
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China
| | - Zhou Liu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China; Guangdong Key Laboratory of Age-Related Cardiocerebral Diseases, Institute of Neurology, Guangdong Medical University, Zhanjiang, Guangdong Province, 524023, China
| | - Bi-Qin Lai
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China; Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong Province, 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China
| | - Ge Li
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China; Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong Province, 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China
| | - Ying Ding
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China; Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong Province, 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China
| | - Wan-Ting Niu
- Department of Orthopedics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yuan-Shan Zeng
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China; Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong Province, 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China; Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, Guangdong Province, 510120, China
| | - Xiang Zeng
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China; Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong Province, 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China; Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, Guangdong Province, 510120, China.
| |
Collapse
|
58
|
Snider C, Grant D, Grant SA. Investigation of an injectable gold nanoparticle extracellular matrix. J Biomater Appl 2021; 36:1289-1300. [PMID: 34672227 DOI: 10.1177/08853282211051586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Post-traumatic osteoarthritis (PTOA) is a progressive articular degenerative disease that degrades articular cartilage and stimulates apoptosis in chondrocyte cells. An injectable decellularized, extracellular matrix (ECM) scaffold, that might be able to combat the effects of PTOA, was developed where the ECM was conjugated with 20 nm gold nanoparticles (AuNP) and supplemented with curcumin and hyaluronic acid (HA). Porcine diaphragm ECM was decellularized and homogenized; AuNPs were conjugated using chemical crosslinking followed by mixing with curcumin and/or HA. Injection force testing and scanning electron microscopy with energy-dispersive X-ray spectroscopy were utilized to characterize the ECM scaffolds. In vitro testing with L929 murine fibroblasts, equine synovial fibroblasts, and Human Chondrocytes were used to determine biocompatibility, reactive oxygen species (ROS) reduction, and chondroprotective ability. The results demonstrated that conjugation of 20 nm AuNPs to the ECM was successful without significantly altering the physical properties as noted in the low injection force. In vitro work provided evidence of biocompatibility with a propensity to reduce intracellular ROS and an ability to mitigate apoptosis of chondrocyte cells stimulated with IL-1β, a known apoptosis inducing cytokine. It was concluded that an injectable AuNP-ECM may have the ability to mitigate inflammation and apoptosis.
Collapse
Affiliation(s)
- Colten Snider
- Department of Bioengineering, 14716University of Missouri, Columbia, MO, USA
| | - David Grant
- Department of Bioengineering, 14716University of Missouri, Columbia, MO, USA
| | - Sheila A Grant
- Department of Bioengineering, 14716University of Missouri, Columbia, MO, USA
| |
Collapse
|
59
|
Li M, Bai Y, Li M, Zhou J. [Performance evaluation of two antigen-extracted xenogeneic ostein and experimental study on repairing skull defects in rats]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2021; 35:1303-1310. [PMID: 34651485 DOI: 10.7507/1002-1892.202103177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Objective To evaluate the physical and chemical properties, immunogenicity, and osteogenesis of two antigen-extracted xenogeneic bone scaffolds-decalcified bone matrix (DBM) and calcined bone. Methods By removing the inorganic and organic components of adult pig femus, xenogeneic DBM and calcined bone were prepared respectively. The density and pH value of the two materials were measured and calculated, the material morphology and pore diameter were observed by scanning electron microscope, and the surface contact angle was measured by automatic contact angle measuring instrument. The safety, osteogenic activity, and immunogenicity of the two materials were evaluated by cytotoxicity test, osteoblast proliferation test, DNA residue test, and human peripheral blood lymphocyte proliferation test. The two materials were implanted into the 5 mm full-thickness skull defect of 6-week-old male Sprague Dawley rats (the blank control group was not implanted with materials). The materials were taken at 4 and 8 weeks after operation, the repair effect of the materials on the rat skull was observed and evaluated by gross observation, Micro-CT scanning, and HE staining observation. Results Compared with calcined bone, DBM has lower density and poor hydrophilicity; the pH value of the two materials was 5.5-6.1, and the pore diameter was 160-800 μm. The two materials were non-cytotoxic and could promote the proliferation of osteoblasts. The absorbance ( A) values of osteoblast proliferation at 1, 4, and 7 days in the DBM group were significantly higher than those in the calcined bone group ( P<0.05). The DNA residues of the two materials were much lower than 50 ng/mg dry weight, and neither of them could stimulate the proliferation and differentiation of human peripheral blood lymphocytes. The results of animal experiments in vivo showed that the bone volume/total volume (BV/TV) in DBM group and calcined bone group were significantly higher than that in blank control group at 4 weeks after operation ( P<0.05), and that in calcined bone group was significantly higher than that in DBM group ( P<0.05); at 8 weeks after operation, there was no significant difference in BV/TV between groups ( P>0.05). HE staining showed that at 4 and 8 weeks after operation, the defect in the blank control group was filled with fibrous connective tissue, the defect was obvious, and no bone growth was found; the defect in DBM group and calcined bone group had been repaired to varying degrees, and a large number of new bone formation could be seen. The material degradability of DBM group was better than that of calcined bone group. Conclusion The physical and chemical properties and degradability of the two kinds of xenogeneic bone scaffolds were slightly different, both of them have no immunogenicity and can promote the repair and reconstruction of skull defects in rats.
Collapse
Affiliation(s)
- Mao Li
- Department of Orthopedics, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, P.R.China
| | - Yulong Bai
- Shanghai Gencong Biomedical Materials Research Center, Shanghai, 201201, P.R.China.,Shanghai Yapeng Biotechnology Co., Ltd, Shanghai, 201201, P.R.China
| | - Miao Li
- Shanghai Yapeng Biotechnology Co., Ltd, Shanghai, 201201, P.R.China
| | - Jianwei Zhou
- Department of Orthopedics, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, P.R.China
| |
Collapse
|
60
|
Li M, Yang S, Song J, Fu T, Liang P, Gao Z, Tang J, Guo L. Different grinding speeds affect induced regeneration capacity of human treated dentin matrix. J Biomed Mater Res B Appl Biomater 2021; 110:755-767. [PMID: 34637601 DOI: 10.1002/jbm.b.34954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 09/18/2021] [Accepted: 09/29/2021] [Indexed: 11/07/2022]
Abstract
Human-treated dentin matrix (hTDM) is a biomaterial scaffold, which can induce implant cells to differentiate into odontoblasts and then form neo-dentin. However, hTDM with long storage or prepared by high-speed handpiece would not to form neo-dentin. In this research, we developed two fresh hTDM with different grinding speeds, which were low-speed hTDM (LTDM) with maximum speed of 500 rpm and high-speed hTDM (HTDM) with a speed of 3,80,000 rpm. Here, we aim to understand whether there were induced regeneration capacity differences between LTDM and HTDM. Scanning electron microscope showed that DFCs grew well on both materials, but the morphology of DFCs and the extracellular matrix was different. Especially, the secreted extracellular matrixes on the inner surface of LTDM were regular morphology and ordered arrangement around the dentin tubules. The transcription-quantitative polymerase chain reaction (qRT-PCR), western blot and immunofluorescence assay showed that the dentin markers DSPP and DMP-1 were about 2× greater in DFCs induced by LTDM than by HTDM, and osteogenic marker BSP was about 2× greater in DFCs induced by HTDM than by LTDM. Histological examinations of the harvested grafts observed the formation of neo-tissue were different, and there were neo-dentin formed on the inner surface of LTDM and neo-cementum formed on the outer surface of HTDM. In summary, it found that the induction abilities of LTDM and HTDM are different, and the dentin matrix is directional. This study lays a necessary foundation for searching the key factors of dentin regeneration in future.
Collapse
Affiliation(s)
- Min Li
- Department of Stomatology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Medical Cosmetology, Suining Central Hospital, Suining, China.,Department of Stomatology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Sen Yang
- Stomatology Center, Suining Central Hospital, Suining, China
| | - Jinlin Song
- Chongqing Medical University Stomatology College, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Tiwei Fu
- Chongqing Medical University Stomatology College, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Panpan Liang
- Chongqing Medical University Stomatology College, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Zhi Gao
- Department of Stomatology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Tang
- Department of Stomatology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lijuan Guo
- Department of Medical Cosmetology, Suining Central Hospital, Suining, China
| |
Collapse
|
61
|
Guan Y, Yang B, Xu W, Li D, Wang S, Ren Z, Zhang J, Zhang T, Liu XZ, Li J, Li C, Meng F, Han F, Wu T, Wang Y, Peng J. Cell-derived extracellular matrix materials for tissue engineering. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:1007-1021. [PMID: 34641714 DOI: 10.1089/ten.teb.2021.0147] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The involvement of cell-derived extracellular matrix (CDM) in assembling tissue engineering scaffolds has yielded significant results. CDM possesses excellent characteristics, such as ideal cellular microenvironment mimicry and good biocompatibility, which make it a popular research direction in the field of bionanomaterials. CDM has significant advantages as an expansion culture substrate for stem cells, including stabilization of phenotype, reversal of senescence, and guidance of specific differentiation. In addition, the applications of CDM-assembled tissue engineering scaffolds for disease simulation and tissue organ repair are comprehensively summarized; the focus is mainly on bone and cartilage repair, skin defect or wound healing, engineered blood vessels, peripheral nerves, and periodontal tissue repair. We consider CDM a highly promising bionic biomaterial for tissue engineering applications and propose a vision for its comprehensive development.
Collapse
Affiliation(s)
- Yanjun Guan
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, Beijing, China;
| | - Boyao Yang
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, Beijing, China;
| | - Wenjing Xu
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, Beijing, China;
| | - Dongdong Li
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, Beijing, China;
| | - Sidong Wang
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, Beijing, China;
| | - Zhiqi Ren
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Jian Zhang
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Tieyuan Zhang
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Xiu-Zhi Liu
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Junyang Li
- Nankai University School of Medicine, 481107, Tianjin, Tianjin, China.,Chinese PLA General Hospital, 104607, Beijing, Beijing, China;
| | - Chaochao Li
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Fanqi Meng
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China.,Peking University People's Hospital, 71185, Department of spine surgery, Beijing, China;
| | - Feng Han
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Tong Wu
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Yu Wang
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China.,Nantong University, 66479, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu, China;
| | - Jiang Peng
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China.,Nantong University, 66479, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu, China;
| |
Collapse
|
62
|
Wang JN, Kan CD, Lin SH, Chang KC, Tsao S, Wong TW. Potential of Autologous Progenitor Cells and Decellularized Porcine Artery Matrix in Construction of Tissue-engineered Vascular Grafts. Organogenesis 2021; 17:72-84. [PMID: 34405770 PMCID: PMC9208767 DOI: 10.1080/15476278.2021.1963603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/14/2021] [Accepted: 07/30/2021] [Indexed: 10/20/2022] Open
Abstract
To develop a tissue-engineered vascular graft, we used pericardial effusion-derived progenitor cells (PEPCs) collected from drained fluid after open-heart surgery in children with congenital heart diseases to repopulate a decellularized porcine pulmonary artery. The PEPCs were compared with human fibroblasts (HS68) and human umbilical vein endothelial cells (HUVECs) in cell growth and migration. They were cultured with the matrices via an inner approach (intima), lateral approach (media), and outer approach (adventitia). PEPCs grew and migrated better than the other two cells 14 days after seeding in the decellularized vessel. In immunofluorescence assays, PEPCs expressed CD90 and CD105 indicating a vascular differentiation. PEPCs grew in a decellularized porcine pulmonary artery matrix may have the potential for producing tissue-engineered vascular grafts.
Collapse
Affiliation(s)
- Jieh-Neng Wang
- Departments Of Pediatrics, National Cheng Kung University Hospital, College Of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chung-Dann Kan
- Departments Of Surgery, National Cheng Kung University Hospital, College Of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shao-Hsien Lin
- Departments Of Pediatrics, National Cheng Kung University Hospital, College Of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ko-Chi Chang
- Departments Of Pediatrics, National Cheng Kung University Hospital, College Of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Stephanie Tsao
- Department Of Dermatology, National Cheng Kung University Hospital, College Of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tak-Wah Wong
- Department Of Dermatology, National Cheng Kung University Hospital, College Of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department Of Biochemistry And Molecular Biology, College Of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center Of Applied Nanomedicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
63
|
Yadav S, Majumder A. Biomimicked hierarchical 2D and 3D structures from natural templates: applications in cell biology. Biomed Mater 2021; 16. [PMID: 34438385 DOI: 10.1088/1748-605x/ac21a7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 08/26/2021] [Indexed: 11/11/2022]
Abstract
Intricate structures of natural surfaces and materials have amazed people over the ages. The unique properties of various surfaces also created interest and curiosity in researchers. In the recent past, with the advent of superior microscopy techniques, we have started to understand how these complex structures provide superior properties. With that knowledge, scientists have developed various biomimicked and bio-inspired surfaces for different non-biological applications. In the last two decades, we have also started to learn how structures of the tissue microenvironment influence cell function and behaviour, both in physiological and pathological conditions. Hence, it became essential to decipher the role and importance of structural hierarchy in the cellular context. With advances in microfabricated techniques, such complex structures were made by superimposing features of different dimensions. However, the fabricated topographies are far from matching the complexities presentin vivo. Hence, the need of biomimicking the natural surfaces for cellular applications was felt. In this review, we discuss a few examples of hierarchical surfaces found in plants, insects, and vertebrates. Such structures have been widely biomimicked for various applications but rarely studied for cell-substrate interaction and cellular response. Here, we discuss the research work wherein 2D hierarchical substrates were prepared using biomimicking to understand cellular functions such as adhesion, orientation, differentiation, and formation of spheroids. Further, we also present the status of ongoing research in mimicking 3D tissue architecture using de-cellularized plant-based and tissue/organ-based scaffolds. We will also discuss 3D printing for fabricating 2D and 3D hierarchical structures. The review will end by highlighting the various advantages and research challenges in this approach. The biomimickedin-vivolike substrate can be used to better understand cellular physiology, and for tissue engineering.
Collapse
Affiliation(s)
- Shital Yadav
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Abhijit Majumder
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| |
Collapse
|
64
|
Kanda H, Ando D, Oya K, Wahyudiono, Goto M. Surfactant-free preparation of an ostrich carotid artery scaffold using liquefied dimethyl ether and DNase. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2021.103280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
65
|
Martínez-Hernández NJ, Díaz-Cuevas A, Milián-Medina L, Sancho-Tello M, Roselló-Ferrando J, Morcillo-Aixelá A, Campo-Cañaveral JL, Roig-Bataller A, Mata-Roig M. Decellularized tracheal prelamination implant: A proposed bilateral double organ technique. Artif Organs 2021; 45:1491-1500. [PMID: 34310703 DOI: 10.1111/aor.14043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 01/08/2023]
Abstract
In tracheal replacement transplantation, prelamination is a critical stage. Nowadays, the most widely used prelamination technique is the prethoracic fascia flap with lateral thoracic artery. We propose a flap based on the internal thoracic artery, which allows a relatively non-aggressive double organ implant, and we have tested its efficacy in decellularized tracheas. Tracheas of albino New Zealand rabbits were decellularized following a protocol that uses detergents and cryogenization, sterilized with 1kGy gamma radiation, and tutorized with a stent. Bilateral pedicled flaps made of pectoral fascia and a muscular component were harvested through a longitudinal 3-cm central thoracic incision, wrapping the tracheas with them in 16 rabbits, remaining them implanted for 2, 4, 8, and 12 weeks. The tracheas were then studied histologically using standard stainings plus immunohistochemistry (CD31). The models were adjusted with Bayesian statistics using ordinal regression; results as odds ratios and credibility intervals. All analysis were performed using R software. Acute inflammatory cell invasion was observed at 2 weeks, which almost disappeared at week 8 after implant. Only macrophages and giant cells increased between Weeks 8 and 12 (OR 10.487, CI [1.603-97.327]). The cartilage maintained its structure, with slight signs of ischemia in a few cases. New CD31-positive vessels were observed from Week 2 and increasing thereafter, reaching a maximum peak at Week 8. We propose a bilateral implant technique that is viable and effective as a prelamination option for two concurrent tracheas, achieving perfect vascularization and integration of the organ with hardly any inflammatory response in the medium or long term.
Collapse
Affiliation(s)
| | | | - Lara Milián-Medina
- Pathology Department, Medicine and Odontology Faculty, Universitat de València, València, Spain
| | | | | | | | | | | | - Manuel Mata-Roig
- Pathology Department, Medicine and Odontology Faculty, Universitat de València, València, Spain.,Networking Research Center on Respiratory Diseases (CIBERER), ISCIII, Madrid, Spain
| |
Collapse
|
66
|
Pereira AT, Schneider KH, Henriques PC, Grasl C, Melo SF, Fernandes IP, Kiss H, Martins MCL, Bergmeister H, Gonçalves IC. Graphene Oxide Coating Improves the Mechanical and Biological Properties of Decellularized Umbilical Cord Arteries. ACS APPLIED MATERIALS & INTERFACES 2021; 13:32662-32672. [PMID: 34240610 DOI: 10.1021/acsami.1c04028] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The lack of small-diameter vascular grafts (inner diameter <5 mm) to substitute autologous grafts in arterial bypass surgeries has a massive impact on the prognosis and progression of cardiovascular diseases, the leading cause of death globally. Decellularized arteries from different sources have been proposed as an alternative, but their poor mechanical performance and high collagen exposure, which promotes platelet and bacteria adhesion, limit their successful application. In this study, these limitations were surpassed for decellularized umbilical cord arteries through the coating of their lumen with graphene oxide (GO). Placental and umbilical cord arteries were decellularized and perfused with a suspension of GO (C/O ratio 2:1) with ∼1.5 μm lateral size. A homogeneous GO coating that completely covered the collagen fibers was obtained for both arteries, with improvement of mechanical properties being achieved for umbilical cord decellularized arteries. GO coating increased the maximum force in 27%, the burst pressure in 29%, the strain in 25%, and the compliance in 10%, compared to umbilical cord decellularized arteries. The achieved theoretical burst pressure (1960 mmHg) and compliance (13.9%/100 mmHg) are similar to the human saphenous vein and mammary artery, respectively, which are used nowadays as the gold standard in coronary and peripheral artery bypass surgeries. Furthermore, and very importantly, coatings with GO did not compromise the endothelial cell adhesion but decreased platelet and bacteria adhesion to decellularized arteries, which will impact on the prevention of thrombosis and infection, until full re-endothetialization is achieved. Overall, our results reveal that GO coating has an effective role in the improvement of decellularized umbilical cord artery performance, which is a huge step toward their application as a small-diameter vascular graft.
Collapse
Affiliation(s)
- Andreia T Pereira
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- Center for Biomedical Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Karl H Schneider
- Center for Biomedical Research, Medical University of Vienna, 1090 Vienna, Austria
- Ludwig Boltzmann Institute of Cardiovascular Research, 1090 Vienna, Austria
| | - Patrícia C Henriques
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Christian Grasl
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria
| | - Sofia F Melo
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Inês P Fernandes
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Herbert Kiss
- Department of Obstetrics and Gynecology, Division of Obstetrics and Feto-Maternal Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - M Cristina L Martins
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Helga Bergmeister
- Center for Biomedical Research, Medical University of Vienna, 1090 Vienna, Austria
- Ludwig Boltzmann Institute of Cardiovascular Research, 1090 Vienna, Austria
| | - Inês C Gonçalves
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- FEUP-Faculdade de Engenharia da Universidade do Porto, 4200-465 Porto, Portugal
| |
Collapse
|
67
|
Ahmed E, Saleh T, Xu M. Recellularization of Native Tissue Derived Acellular Scaffolds with Mesenchymal Stem Cells. Cells 2021; 10:cells10071787. [PMID: 34359955 PMCID: PMC8304639 DOI: 10.3390/cells10071787] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/03/2021] [Accepted: 07/12/2021] [Indexed: 12/22/2022] Open
Abstract
The functionalization of decellularized scaffolds is still challenging because of the recellularization-related limitations, including the finding of the most optimal kind of cell(s) and the best way to control their distribution within the scaffolds to generate native mimicking tissues. That is why researchers have been encouraged to study stem cells, in particular, mesenchymal stem cells (MSCs), as alternative cells to repopulate and functionalize the scaffolds properly. MSCs could be obtained from various sources and have therapeutic effects on a wide range of inflammatory/degenerative diseases. Therefore, in this mini-review, we will discuss the benefits using of MSCs for recellularization, the factors affecting their efficiency, and the drawbacks that may need to be overcome to generate bioengineered transplantable organs.
Collapse
Affiliation(s)
- Ebtehal Ahmed
- Department of Pathology, Faculty of Veterinary Medicine, Assiut University, Assiut 71515, Egypt;
| | - Tarek Saleh
- Department of Animal Surgery, Faculty of Veterinary Medicine, Assiut University, Assiut 71515, Egypt;
| | - Meifeng Xu
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
- Correspondence: or ; Tel.: +1-513-558-4725; Fax: +1-513-558-2141
| |
Collapse
|
68
|
Parihar A, Pandita V, Kumar A, Parihar DS, Puranik N, Bajpai T, Khan R. 3D Printing: Advancement in Biogenerative Engineering to Combat Shortage of Organs and Bioapplicable Materials. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2021; 8:173-199. [PMID: 34230892 PMCID: PMC8252697 DOI: 10.1007/s40883-021-00219-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/26/2021] [Accepted: 06/08/2021] [Indexed: 02/06/2023]
Abstract
Abstract Organ or cell transplantation is medically evaluated for end-stage failure saving or extending the lives of thousands of patients who are suffering from organ failure disorders. The unavailability of adequate organs for transplantation to meet the existing demand is a major challenge in the medical field. This led to day-day-increase in the number of patients on transplant waiting lists as well as in the number of patients dying while on the queue. Recently, technological advancements in the field of biogenerative engineering have the potential to regenerate tissues and, in some cases, create new tissues and organs. In this context, major advances and innovations are being made in the fields of tissue engineering and regenerative medicine which have a huge impact on the scientific community is three-dimensional bioprinting (3D bioprinting) of tissues and organs. Besides this, the decellularization of organs and using this as a scaffold for generating new organs through the recellularization process shows promising results. This review discussed about current approaches for tissue and organ engineering including methods of scaffold designing, recent advances in 3D bioprinting, organs regenerated successfully using 3D printing, and extended application of 3D bioprinting technique in the field of medicine. Besides this, information about commercially available 3D printers has also been included in this article. Lay Summary Today's need for organs for the transplantation process in order to save a patient's life or to enhance the survival rate of diseased one is the prime concern among the scientific community. Recent, advances in the field of biogenerative engineering have the potential to regenerate tissues and create organs compatible with the patient's body. In this context, major advances and innovations are being made in the fields of tissue engineering and regenerative medicine which have a huge impact on the scientific community is three-dimensional bioprinting (3D bioprinting) of tissues and organs. Besides this, the decellularization of organs and using this as a scaffold for generating new organs through the recellularization process shows promising results. This review dealt with the current approaches for tissue and organ engineering including methods of scaffold designing, recent advances in 3D bioprinting, organs regenerated successfully using 3D printing, and extended application of 3D bioprinting technique in the field of medicine. Furthermore, information about commercially available 3D printers has also been included in this article.
Collapse
Affiliation(s)
- Arpana Parihar
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, Madhya Pradesh 462026 India
- Microfluidics & MEMS Centre, CSIR-Advanced Materials and Processes Research Institute (AMPRI), Hoshangabad Road Bhopal, 462026 India
| | - Vasundhara Pandita
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, Madhya Pradesh 462026 India
| | - Avinash Kumar
- Department of Mechanical Engineering, Indian Institute of Information Technology, Design & Manufacturing (IIITD&M), Kancheepuram, 600127 India
| | - Dipesh Singh Parihar
- Engineering College Tuwa , At. & Post. Tuwa, Taluka Godhra, Dist. Panchmahal, Godhra, Gujarat 388713 India
| | - Nidhi Puranik
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, Madhya Pradesh 462026 India
| | - Tapas Bajpai
- Department of Mechanical Engineering, Malaviya National Institute of Technology, Jaipur, 302017 India
| | - Raju Khan
- Microfluidics & MEMS Centre, CSIR-Advanced Materials and Processes Research Institute (AMPRI), Hoshangabad Road Bhopal, 462026 India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-AMPRI, Bhopal, 462026 India
| |
Collapse
|
69
|
Kanda H, Ando D, Hoshino R, Yamamoto T, Wahyudiono, Suzuki S, Shinohara S, Goto M. Surfactant-Free Decellularization of Porcine Aortic Tissue by Subcritical Dimethyl Ether. ACS OMEGA 2021; 6:13417-13425. [PMID: 34056489 PMCID: PMC8158793 DOI: 10.1021/acsomega.1c01549] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/07/2021] [Indexed: 05/07/2024]
Abstract
Porcine aortic tissue was decellularized by subcritical dimethyl ether (DME) used as an alternative to the surfactant sodium dodecyl sulfate. The process included three steps. For the first step, lipids were extracted from the porcine aorta using subcritical DME at 23 °C with a DME pressure of 0.56 MPa. Next, DME was evaporated from the aorta under atmospheric pressure and temperature. The second step involved DNA fragmentation by DNase, which was primarily identical to the common method. For the third step, similar to the common method, DNA fragments were removed by washing with water and ethanol. After 3 days of DNase treatment, the amount of DNA remaining in the porcine aorta was 40 ng/dry-mg, which was lower than the standard value of 50 ng/mg-dry. Hematoxylin and eosin staining showed that most cell nuclei were removed from the aorta. These results demonstrate that subcritical DME eliminates the need to utilize surfactants.
Collapse
Affiliation(s)
- Hideki Kanda
- Department
of Materials Process Engineering, Nagoya University, Nagoya 464-8603, Japan
| | - Daigo Ando
- Department
of Materials Process Engineering, Nagoya University, Nagoya 464-8603, Japan
| | - Rintaro Hoshino
- Department
of Materials Process Engineering, Nagoya University, Nagoya 464-8603, Japan
| | - Tetsuya Yamamoto
- Department
of Chemical Systems Engineering, Nagoya
University, Nagoya 464-8603, Japan
| | - Wahyudiono
- Department
of Materials Process Engineering, Nagoya University, Nagoya 464-8603, Japan
| | - Shogo Suzuki
- Eco
Business Development Center, Ricoh, 1-10 Komakado, Gotemba 412-0038, Japan
| | - Satoshi Shinohara
- Eco
Business Development Center, Ricoh, 1-10 Komakado, Gotemba 412-0038, Japan
| | - Motonobu Goto
- Department
of Materials Process Engineering, Nagoya University, Nagoya 464-8603, Japan
| |
Collapse
|
70
|
Palomares D, Ammann KR, Saldana Perez JJ, Gomez A, Barreda A, Russell-Cheung A, Martin A, Tran PL, Hossainy S, Slepian RC, Hossainy SF, Slepian MJ. Patterned Electrospinning: A Method of Generating Defined Fibrous Constructs Influencing Cell Adhesion and Retention. ACS APPLIED BIO MATERIALS 2021; 4:4084-4093. [DOI: 10.1021/acsabm.0c01311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Daniel Palomares
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona 85721-0072, United States
| | - Kaitlyn R. Ammann
- Department of Medicine, University of Arizona, Tucson, Arizona 85721-0072, United States
- Sarver Heart Center, University of Arizona, Tucson, Arizona 85721-0001, United States
| | - Javier J. Saldana Perez
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona 85721-0072, United States
| | - Alexan Gomez
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona 85721-0072, United States
| | - Adriana Barreda
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona 85721-0072, United States
| | - Andrew Russell-Cheung
- Department of Biological & Biomedical Sciences, University of Arizona, Tucson, Arizona 85721-0001, United States
| | - Adriana Martin
- Sarver Heart Center, University of Arizona, Tucson, Arizona 85721-0001, United States
| | - Phat Le Tran
- Department of Bioengineering, University of California, Berkeley, California 94720, United States
| | - Sahir Hossainy
- Sarver Heart Center, University of Arizona, Tucson, Arizona 85721-0001, United States
| | - Rebecca C. Slepian
- Sarver Heart Center, University of Arizona, Tucson, Arizona 85721-0001, United States
| | - Syed F.A. Hossainy
- Department of Bioengineering, University of California, Berkeley, California 94720, United States
| | - Marvin J. Slepian
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona 85721-0072, United States
- Department of Medicine, University of Arizona, Tucson, Arizona 85721-0072, United States
- Sarver Heart Center, University of Arizona, Tucson, Arizona 85721-0001, United States
| |
Collapse
|
71
|
Suvarnapathaki S, Nguyen MA, Goulopoulos AA, Lantigua D, Camci-Unal G. Engineering calcium peroxide based oxygen generating scaffolds for tissue survival. Biomater Sci 2021; 9:2519-2532. [PMID: 33565527 PMCID: PMC11442008 DOI: 10.1039/d0bm02048f] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Oxygen supply is essential for the long-term viability and function of tissue engineered constructs in vitro and in vivo. The integration with the host blood supply as the primary source of oxygen to cells requires 4 to 5 weeks in vivo and involves neovascularization stages to support the delivery of oxygenated blood to cells. Consequently, three-dimensional (3D) encapsulated cells during this process are prone to oxygen deprivation, cellular dysfunction, damage, and hypoxia-induced necrosis. Here we demonstrate the use of calcium peroxide (CaO2) and polycaprolactone (PCL), as part of an emerging paradigm of oxygen-generating scaffolds that substitute the host oxygen supply via hydrolytic degradation. The 35-day in vitro study showed predictable oxygen release kinetics that achieved 5% to 29% dissolved oxygen with increasing CaO2 loading. As a biomaterial, the iterations of 0 mg, 40 mg, and 60 mg of CaO2 loaded scaffolds yielded modular mechanical behaviors, ranging from 5-20 kPa in compressive strength. The other controlled physiochemical features included swelling capacities of 22-33% and enzymatic degradation rates of 0.8% to 60% remaining mass. The 3D-encapsulation experiments of NIH/3T3 fibroblasts, L6 rat myoblasts, and primary cardiac fibroblasts in these scaffolds showed enhanced cell survival, proliferation, and function under hypoxia. During continuous oxygen release, the scaffolds maintained a stable tissue culture system between pH 8 to 9. The broad basis of this work supports prospects in the expansion of robust and clinically translatable tissue constructs.
Collapse
Affiliation(s)
- Sanika Suvarnapathaki
- Biomedical Engineering and Biotechnology Program, University of Massachusetts Lowell, One University Avenue, Lowell, MA 01854, USA
| | | | | | | | | |
Collapse
|
72
|
Nakayama-Iwatsuki K, Yanagisawa K, Tanaka D, Hirabayashi M, Negishi J, Hochi S. Acellular matrix derived from rat liver improves the functionality of rat pancreatic islets before or after vitrification. Cryobiology 2021; 100:90-95. [PMID: 33757759 DOI: 10.1016/j.cryobiol.2021.03.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/26/2021] [Accepted: 03/16/2021] [Indexed: 12/18/2022]
Abstract
Cryopreservation of pancreatic islets can overcome the severe shortage of islet donors in clinical islet transplantation, but the impaired quality of post-warm islets need improvement. This present study was conducted to investigate whether the pre- or post-treatment of rat islets with liver decellularized matrix (LDM) for vitrification can improve the viability (FDA/PI double staining) and the functionality (glucose-stimulated insulin secretion [GSIS] assay). Rat LDM was prepared by high-hydrostatic pressure, lyophilization, and re-suspension in saline. Co-culturing of isolated islets with 0 (control), 30, 60, or 90 μg/ml LDM for 24 h resulted in the comparable viability among the 4 groups (98.7-99.6%) and the higher insulin secretion potential in 30 and 60 μg/ml LDM treatment groups than the control group (stimulation index [SI]: 12.1 and 12.7, respectively, vs. 6.5 in the control group, P < 0.05). When the islets co-cultured with 60 μg/ml LDM were vitrified-warmed on a nylon mesh cryodevice, the viability and the GSIS of the post-warm islets were not improved. Post-treatment of vitrified-warmed islets with 60 μg/ml LDM during the recovery culture for 12 h resulted in the comparable clearance of degenerating cell debris from the post-warm islets, while their insulin secretion potential was improved (SI: 5.0 vs. 3.5 in the control group, P < 0.05). These findings indicate that the components in LDM can enhance the insulin secretion potential of rat islets suffering damage by enzymatic stress during the islet isolation process or by cryoinjuries during the vitrification-warming process.
Collapse
Affiliation(s)
- Kenyu Nakayama-Iwatsuki
- Graduate School of Science and Technology, Shinshu University, Ueda, Nagano, 386-8567, Japan; National Institute for Physiological Sciences, Okazaki, Aichi, 444-8787, Japan
| | - Kotaro Yanagisawa
- Graduate School of Science and Technology, Shinshu University, Ueda, Nagano, 386-8567, Japan
| | - Dan Tanaka
- Graduate School of Science and Technology, Shinshu University, Ueda, Nagano, 386-8567, Japan
| | - Masumi Hirabayashi
- National Institute for Physiological Sciences, Okazaki, Aichi, 444-8787, Japan; School of Life Science, The Graduate University for Advanced Studies, Okazaki, Aichi, 444-8787, Japan
| | - Jun Negishi
- Graduate School of Science and Technology, Shinshu University, Ueda, Nagano, 386-8567, Japan; Faculty of Textile Science and Technology, Shinshu University, Ueda, Nagano, 386-8567, Japan
| | - Shinichi Hochi
- Graduate School of Science and Technology, Shinshu University, Ueda, Nagano, 386-8567, Japan; Faculty of Textile Science and Technology, Shinshu University, Ueda, Nagano, 386-8567, Japan.
| |
Collapse
|
73
|
Elashry MI, Baulig N, Wagner AS, Klymiuk MC, Kruppke B, Hanke T, Wenisch S, Arnhold S. Combined macromolecule biomaterials together with fluid shear stress promote the osteogenic differentiation capacity of equine adipose-derived mesenchymal stem cells. Stem Cell Res Ther 2021; 12:116. [PMID: 33579348 PMCID: PMC7879632 DOI: 10.1186/s13287-021-02146-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 01/06/2021] [Indexed: 11/10/2022] Open
Abstract
Background Combination of mesenchymal stem cells (MSCs) and biomaterials is a rapidly growing approach in regenerative medicine particularly for chronic degenerative disorders including osteoarthritis and osteoporosis. The present study examined the effect of biomaterial scaffolds on equine adipose-derived MSC morphology, viability, adherence, migration, and osteogenic differentiation. Methods MSCs were cultivated in conjunction with collagen CultiSpher-S Microcarrier (MC), nanocomposite xerogels B30 and combined B30 with strontium (B30Str) biomaterials in osteogenic differentiation medium either under static or mechanical fluid shear stress (FSS) culture conditions. The data were generated by histological means, live cell imaging, cell viability, adherence and migration assays, semi-quantification of alkaline phosphatase (ALP) activity, and quantification of the osteogenic markers runt-related transcription factor 2 (Runx2) and alkaline phosphatase (ALP) expression. Results The data revealed that combined mechanical FSS with MC but not B30 enhanced MSC viability and promoted their migration. Combined osteogenic medium with MC, B30, and B30Str increased ALP activity compared to cultivation in basal medium. Osteogenic induction with MC, B30, and B30Str resulted in diffused matrix mineralization. The combined osteogenic induction with biomaterials under mechanical FSS increased Runx2 protein expression either in comparison to those cells cultivated in BM or those cells induced under static culture. Runx2 and ALP expression was upregulated following combined osteogenic differentiation together with B30 and B30Str regardless of static or FSS culture. Conclusions Taken together, the data revealed that FSS in conjunction with biomaterials promoted osteogenic differentiation of MSCs. This combination may be considered as a marked improvement for clinical applications to cure bone defects. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02146-7.
Collapse
Affiliation(s)
- Mohamed I Elashry
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, Frankfurter Str. 98, 35392, Giessen, Germany.
| | - Nadine Baulig
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, Frankfurter Str. 98, 35392, Giessen, Germany
| | - Alena-Svenja Wagner
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, 35392, Giessen, Germany.,Institute of Veterinary Physiology and Biochemistry, Justus Liebig University of Giessen, 35392, Giessen, Germany
| | - Michele C Klymiuk
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, Frankfurter Str. 98, 35392, Giessen, Germany
| | - Benjamin Kruppke
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Str. 27, 01069, Dresden, Germany
| | - Thomas Hanke
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Str. 27, 01069, Dresden, Germany
| | - Sabine Wenisch
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, 35392, Giessen, Germany
| | - Stefan Arnhold
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, Frankfurter Str. 98, 35392, Giessen, Germany
| |
Collapse
|
74
|
Kamalvand M, Biazar E, Daliri-Joupari M, Montazer F, Rezaei-Tavirani M, Heidari-Keshel S. Design of a decellularized fish skin as a biological scaffold for skin tissue regeneration. Tissue Cell 2021; 71:101509. [PMID: 33621947 DOI: 10.1016/j.tice.2021.101509] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 02/03/2021] [Accepted: 02/03/2021] [Indexed: 01/02/2023]
Abstract
The use of decellularized natural skin as an extracellular matrix (ECM) may be a great candidate to regenerate damaged tissues. In this study, decellularized scaffolds from fish skin were designed by different techniques (physical, chemical, and enzymatic methods) and investigated by analyses such as Differential Scanning Calorimetry (DSC), Scanning Electron Microscopy (SEM), Tensile strength, Degradability, Histological studies, Toxicity test, and Determination of DNA content. Results showed that the best sample is related to the decellularized skin by hypertonic & hypotonic technique and Triton X100 solutions. Structural and mechanical results were demonstrated that samples have similar properties to human skin to regenerate it. The cytotoxicity results showed that decellularized skin by hypertonic & hypotonic method and Triton solution is non-toxic with minimal amount of genetic materials. Cellular results with epithelial cells indicated good adhesion on decellularized matrix, so it can be a suitable candidate for skin tissue regeneration.
Collapse
Affiliation(s)
- Mahshad Kamalvand
- Tissue Engineering Group, Department of Biomedical Engineering, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Esmaeil Biazar
- Tissue Engineering Group, Department of Biomedical Engineering, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran.
| | | | - Fatemeh Montazer
- Pathology Department, Firoozabadi Clinical Research Development Unit, Iran University of Medical Sciences, Tehran, Iran
| | | | - Saeed Heidari-Keshel
- Medical Nanotechnology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
75
|
Rabbani M, Zakian N, Alimoradi N. Contribution of Physical Methods in Decellularization of Animal Tissues. JOURNAL OF MEDICAL SIGNALS & SENSORS 2021; 11:1-11. [PMID: 34026585 PMCID: PMC8043117 DOI: 10.4103/jmss.jmss_2_20] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/19/2020] [Accepted: 07/19/2020] [Indexed: 01/12/2023]
Abstract
Biologic scaffolds composed of extracellular matrix (ECM) are frequently used for clinical purposes of tissue regeneration. Different methods have been developed for this purpose. All methods of decellularization including chemical and physical approaches leave some damage on the ECM; however, the effects of these methods are different which make some of these procedures more proper to maintain ECM structure than other methods. This review is aimed to introduce and compare new physical methods for the decellularization of different tissues and organs in tissue engineering. All recent reports and research that have used at least one physical method in the procedure of decellularization, were included and evaluated in this paper. The advantages and drawbacks of each method were examined and compared considering the effectiveness. This review tried to highlight the prospective potentials and benefits of applying physical methods for decellularization protocols in tissue engineering instead of the current chemical methods. These chemical methods are harsh in nature and were shown to be destructive and harmful to essential substances of ECM and scaffold structure. Therefore, using physical methods as a partial or even a whole protocol could save time, costs, and quality of the final acellular tissue in complicated decellularization procedures. Moreover, regarding the control factor that could be achieved easily with physical methods, optimization of different decellularization protocols would be quite satisfactory. Combined methods take advantage of both chemical and physical approaches.
Collapse
Affiliation(s)
- Mohsen Rabbani
- Department of Biomedical Engineering, University of Isfahan, Isfahan, Iran
| | - Nasrin Zakian
- Department of Biomedical Engineering, University of Isfahan, Isfahan, Iran
| | - Nima Alimoradi
- Department of Biomedical Engineering, University of Isfahan, Isfahan, Iran
| |
Collapse
|
76
|
Feng Y, Jiang Z, Zhang Y, Miao X, Yu Q, Xie Z, Yang G. Stem-cell-derived ECM sheet-implant complexes for enhancing osseointegration. Biomater Sci 2020; 8:6647-6656. [PMID: 33074268 DOI: 10.1039/d0bm00980f] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Numerous treatment methods have been developed to modify the surface of dental implants to improve cell migration and proliferation, removal torque, and osseointegration. Recent studies have constructed cell sheet-implant complexes with enhanced osteogenic capabilities. However, these complexes have some limitations, such as requirements for complex preparation processes, cell vitality maintenance, strict preservation conditions, and the induction of immunogenicity. Extracellular matrix (ECM) sheets without cells may be a more desirable material. To date, the effect of ECM sheets on implant osseointegration has not been reported. In this study, we fabricated ECM sheet-implant complexes through the combination of rat bone marrow mesenchymal stem cell (BMSC)-derived ECM sheets with sandblasted, large-grit, acid-etched (SLA) implants. These complexes were characterized by light microscopy, scanning electron microscopy (SEM), and immunofluorescence (IF) assays. The adhesion, proliferation, and osteogenic differentiation of BMSCs cultured on ECM sheets were detected in vitro. Then, the ECM sheet-implant complexes were transplanted into the metaphysis of the tibias of rats to evaluate the implant osseointegration in vivo. The results showed that ECM sheets were successfully constructed and showed significantly improved adhesion and proliferation. BMSCs cultured on ECM sheets upregulated the expression levels of the osteogenic-related genes alkaline phosphatase (ALP), bone morphogenetic protein 2 (BMP2), and runt-related transcription factor 2 (Runx2) compared to controls. In vivo, ECM sheet-implant complexes demonstrated superior new bone formation. Our findings proved that the BMSC-derived ECM sheets promoted osseointegration in vitro and in vivo. The current study indicated that the ECM sheet could be an ideal tissue engineering material, and ECM sheet-implant complexes could provide a strategy with low immunogenicity and easy storage and transportation. This research provides a novel strategy for the development of implant surface modification approaches.
Collapse
Affiliation(s)
- Yuting Feng
- Department of Implantology, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, No. 395, Yan'an Road, Xia-Cheng Region, Hangzhou, Zhejiang 310006, China.
| | | | | | | | | | | | | |
Collapse
|
77
|
Rashid S, Qazi REM, Malick TS, Salim A, Khan I, Ilyas A, Haneef K. Effect of valproic acid on the hepatic differentiation of mesenchymal stem cells in 2D and 3D microenvironments. Mol Cell Biochem 2020; 476:909-919. [PMID: 33111212 DOI: 10.1007/s11010-020-03955-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs) have multi-lineage differentiation potential which make them an excellent source for cell-based therapies. Histone modification is one of the major epigenetic regulations that play central role in stem cell differentiation. Keeping in view their ability to maintain gene expression essential for successful differentiation, it was interesting to examine the effects of valproic acid (VPA), a histone deacetylase inhibitor, in the hepatic differentiation of MSCs within the 3D scaffold. MSCs were treated with the optimized concentration of VPA in the 3D collagen scaffold. Analyses of hepatic differentiation potential of treated MSCs were performed by qPCR, immunostaining and periodic acid Schiff assay. Our results demonstrate that MSCs differentiate into hepatic-like cells when treated with 5 mM VPA for 24 h. The VPA-treated MSCs have shown significant upregulation in the expression of hepatic genes, CK-18 (P < 0.05), TAT (P < 0.01), and AFP (P < 0.001), and hepatic proteins, AFP (P < 0.05) and ALB (P < 0.01). In addition, acetylation of histones (H3 and H4) was significantly increased (P < 0.001) in VPA-pretreated cells. Further analysis showed that VPA treatment significantly enhanced (P < 0.01) glycogen storage, an important functional aspect of hepatic cells. The present study revealed the effectiveness of VPA in hepatic differentiation within the 3D collagen scaffold. These hepatic-like cells may have an extended clinical applicability in future for successful liver regeneration.
Collapse
Affiliation(s)
- Saman Rashid
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi, 75270, Pakistan
| | - Rida-E-Maria Qazi
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Tuba Shakil Malick
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Asmat Salim
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Irfan Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Amber Ilyas
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi, 75270, Pakistan
| | - Kanwal Haneef
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
78
|
Priester C, MacDonald A, Dhar M, Bow A. Examining the Characteristics and Applications of Mesenchymal, Induced Pluripotent, and Embryonic Stem Cells for Tissue Engineering Approaches across the Germ Layers. Pharmaceuticals (Basel) 2020; 13:E344. [PMID: 33114710 PMCID: PMC7692540 DOI: 10.3390/ph13110344] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
The field of regenerative medicine utilizes a wide array of technologies and techniques for repairing and restoring function to damaged tissues. Among these, stem cells offer one of the most potent and promising biological tools to facilitate such goals. Implementation of mesenchymal stem cells (MSCs), induced pluripotent stem cells (iPSCs), and embryonic stem cells (ESCs) offer varying advantages based on availability and efficacy in the target tissue. The focus of this review is to discuss characteristics of these three subset stem cell populations and examine their utility in tissue engineering. In particular, the development of therapeutics that utilize cell-based approaches, divided by germinal layer to further assess research targeting specific tissues of the mesoderm, ectoderm, and endoderm. The combinatorial application of MSCs, iPSCs, and ESCs with natural and synthetic scaffold technologies can enhance the reparative capacity and survival of implanted cells. Continued efforts to generate more standardized approaches for these cells may provide improved study-to-study variations on implementation, thereby increasing the clinical translatability of cell-based therapeutics. Coupling clinically translatable research with commercially oriented methods offers the potential to drastically advance medical treatments for multiple diseases and injuries, improving the quality of life for many individuals.
Collapse
Affiliation(s)
- Caitlin Priester
- Department of Animal Science, University of Tennessee, Knoxville, TN 37998, USA;
| | - Amber MacDonald
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA; (A.M.); (M.D.)
| | - Madhu Dhar
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA; (A.M.); (M.D.)
| | - Austin Bow
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA; (A.M.); (M.D.)
| |
Collapse
|
79
|
Farhat W, Chatelain F, Marret A, Faivre L, Arakelian L, Cattan P, Fuchs A. Trends in 3D bioprinting for esophageal tissue repair and reconstruction. Biomaterials 2020; 267:120465. [PMID: 33129189 DOI: 10.1016/j.biomaterials.2020.120465] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/15/2020] [Accepted: 10/18/2020] [Indexed: 02/08/2023]
Abstract
In esophageal pathologies, such as esophageal atresia, cancers, caustic burns, or post-operative stenosis, esophageal replacement is performed by using parts of the gastrointestinal tract to restore nutritional autonomy. However, this surgical procedure most often does not lead to complete functional recovery and is instead associated with many complications resulting in a decrease in the quality of life and survival rate. Esophageal tissue engineering (ETE) aims at repairing the defective esophagus and is considered as a promising therapeutic alternative. Noteworthy progress has recently been made in the ETE research area but strong challenges remain to replicate the structural and functional integrity of the esophagus with the approaches currently being developed. Within this context, 3D bioprinting is emerging as a new technology to facilitate the patterning of both cellular and acellular bioinks into well-organized 3D functional structures. Here, we present a comprehensive overview of the recent advances in tissue engineering for esophageal reconstruction with a specific focus on 3D bioprinting approaches in ETE. Current biofabrication techniques and bioink features are highlighted, and these are discussed in view of the complexity of the native esophagus that the designed substitute needs to replace. Finally, perspectives on recent strategies for fabricating other tubular organ substitutes via 3D bioprinting are discussed briefly for their potential in ETE applications.
Collapse
Affiliation(s)
- Wissam Farhat
- Université de Paris, Inserm, U976 HIPI, F-75006, Paris, France; AP-HP, Hôpital Saint-Louis, 1 avenue Vellefaux, F-75010, Paris, France; CEA, IRIG, F-38000, Grenoble, France
| | - François Chatelain
- Université de Paris, Inserm, U976 HIPI, F-75006, Paris, France; AP-HP, Hôpital Saint-Louis, 1 avenue Vellefaux, F-75010, Paris, France; CEA, IRIG, F-38000, Grenoble, France
| | - Auriane Marret
- Université de Paris, Inserm, U976 HIPI, F-75006, Paris, France; AP-HP, Hôpital Saint-Louis, 1 avenue Vellefaux, F-75010, Paris, France; CEA, IRIG, F-38000, Grenoble, France
| | - Lionel Faivre
- Université de Paris, Inserm, U976 HIPI, F-75006, Paris, France; Assistance Publique - Hôpitaux de Paris, Unité de Thérapie Cellulaire, Hôpital Saint-Louis, Paris, France
| | - Lousineh Arakelian
- Université de Paris, Inserm, U976 HIPI, F-75006, Paris, France; Assistance Publique - Hôpitaux de Paris, Unité de Thérapie Cellulaire, Hôpital Saint-Louis, Paris, France
| | - Pierre Cattan
- Université de Paris, Inserm, U976 HIPI, F-75006, Paris, France; Assistance Publique - Hôpitaux de Paris, Service de Chirurgie Digestive, Hôpital Saint-Louis, Paris, France
| | - Alexandra Fuchs
- Université de Paris, Inserm, U976 HIPI, F-75006, Paris, France; AP-HP, Hôpital Saint-Louis, 1 avenue Vellefaux, F-75010, Paris, France; CEA, IRIG, F-38000, Grenoble, France.
| |
Collapse
|
80
|
Nakamura N, Saito K, Kimura T, Kishida A. Recellularization of decellularized cancellous bone scaffolds using low-temperature cell seeding. Tissue Cell 2020; 66:101385. [DOI: 10.1016/j.tice.2020.101385] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 05/13/2020] [Accepted: 05/15/2020] [Indexed: 12/25/2022]
|
81
|
Li Y, Zhu T, Wang L, Jiang J, Xie G, Huangfu X, Dong S, Zhao J. Tissue-Engineered Decellularized Allografts for Anterior Cruciate Ligament Reconstruction. ACS Biomater Sci Eng 2020; 6:5700-5710. [PMID: 33320573 DOI: 10.1021/acsbiomaterials.0c00269] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Anterior cruciate ligament (ACL) reconstruction with allografts is limited by high immunogenicity, poor cellularization, and delayed tendon-bone healing. Decellularized tendons (DAs) have been used as bioscaffolds to reconstruct ligaments with variable success. In the study, four kinds of decellularized allogeneic hamstring tendons were prepared and their microstructure and cytocompatibility were examined in vitro. The results showed that decellularized allografts neutralized by 5% calcium bicarbonate had typical reticular and porous microstructures with optical cytocompatibility. Tissue-engineering decellularized allografts (TEDAs) were prepared with the selected decellularized allografts and tendon stem/progenitor cells and used for ACL reconstruction in a rabbit model. Histological staining showed that the TEDAs promoted cellular infiltration and new vessel formation significantly and improved tendon-bone healing moderately compared to decellularized allografts. Better macroscopic scores and biomechanical results were observed in TEDA groups, but there were no significant differences between DA and TEDA groups at months 1, 2, and 3 postoperatively. Immunohistochemical data showed that the tissue-engineering decellularized allografts enhanced the expression of collagen I at each timepoint and collagen III at months 1 and 2. ELISA analysis showed that the tissue-engineering decellularized allografts reduced the secretion of IgE and IL-1β within 1 month and promoted the secretion of IL-2, IL-4, IL-10, and IL-17 after 1 month. The results showed that tissue-engineering decellularized allografts strengthened intra-articular graft remodeling significantly and provided moderate improvements in tendon-bone healing by creating more suitable immune responses than decellularized allografts. The study revealed that tissue-engineering decellularized allografts as a promising option for ACL reconstruction could achieve more favorable outcomes.
Collapse
Affiliation(s)
- Yamin Li
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Tonghe Zhu
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Liren Wang
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Jia Jiang
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Guoming Xie
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Xiaoqiao Huangfu
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Shikui Dong
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Jinzhong Zhao
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| |
Collapse
|
82
|
Novoseletskaya E, Grigorieva O, Nimiritsky P, Basalova N, Eremichev R, Milovskaya I, Kulebyakin K, Kulebyakina M, Rodionov S, Omelyanenko N, Efimenko A. Mesenchymal Stromal Cell-Produced Components of Extracellular Matrix Potentiate Multipotent Stem Cell Response to Differentiation Stimuli. Front Cell Dev Biol 2020; 8:555378. [PMID: 33072743 PMCID: PMC7536557 DOI: 10.3389/fcell.2020.555378] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 08/31/2020] [Indexed: 12/13/2022] Open
Abstract
Extracellular matrix (ECM) provides both structural support and dynamic microenvironment for cells regulating their behavior and fate. As a critical component of stem cell niche ECM maintains stem cells and activates their proliferation and differentiation under specific stimuli. Mesenchymal stem/stromal cells (MSCs) regulate tissue-specific stem cell functions locating in their immediate microenvironment and producing various bioactive factors, including ECM components. We evaluated the ability of MSC-produced ECM to restore stem and progenitor cell microenvironment in vitro and analyzed the possible mechanisms of its effects. Human MSC cell sheets were decellularized by different agents (detergents, enzymes, and apoptosis inductors) to select the optimized combination (CHAPS and DNAse I) based on the conservation of decellularized ECM (dECM) structure and effectiveness of DNA removal. Prepared dECM was non-immunogenic, supported MSC proliferation and formation of larger colonies in colony-forming unit-assay. Decellularized ECM effectively promoted MSC trilineage differentiation (adipogenic, osteogenic, and chondrogenic) compared to plastic or plastic covered by selected ECM components (collagen, fibronectin, laminin). Interestingly, dECM produced by human fibroblasts could not enhance MSC differentiation like MSC-produced dECM, indicating cell-specific functionality of dECM. We demonstrated the significant integrin contribution in dECM-cell interaction by blocking the stimulatory effects of dECM with RGD peptide and suggested the involvement of key intracellular signaling pathways activation (pERK/ERK and pFAK/FAK axes, pYAP/YAP and beta-catenin) in the observed processes based on the results of inhibitory analysis. Taken together, we suppose that MSC-produced dECM may mimic stem cell niche components in vitro and maintain multipotent progenitor cells to insure their effective response to external differentiating stimuli upon activation. The obtained data provide more insights into the possible role of MSC-produced ECM in stem and progenitor cell regulation within their niches. Our results are also useful for the developing of dECM-based cell-free products for regenerative medicine.
Collapse
Affiliation(s)
- Ekaterina Novoseletskaya
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia.,Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Olga Grigorieva
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | - Peter Nimiritsky
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia.,Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Nataliya Basalova
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia.,Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Roman Eremichev
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | - Irina Milovskaya
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Konstantin Kulebyakin
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia.,Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Maria Kulebyakina
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Sergei Rodionov
- N.N. Priorov National Medical Research Center of Traumatology and Orthopedics, Moscow, Russia
| | - Nikolai Omelyanenko
- N.N. Priorov National Medical Research Center of Traumatology and Orthopedics, Moscow, Russia
| | - Anastasia Efimenko
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia.,Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
83
|
Peng W, Peng Z, Tang P, Sun H, Lei H, Li Z, Hui D, Du C, Zhou C, Wang Y. Review of Plastic Surgery Biomaterials and Current Progress in Their 3D Manufacturing Technology. MATERIALS 2020; 13:ma13184108. [PMID: 32947925 PMCID: PMC7560273 DOI: 10.3390/ma13184108] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 02/05/2023]
Abstract
Plastic surgery is a broad field, including maxillofacial surgery, skin flaps and grafts, liposuction and body contouring, breast surgery, and facial cosmetic procedures. Due to the requirements of plastic surgery for the biological safety of materials, biomaterials are widely used because of its superior biocompatibility and biodegradability. Currently, there are many kinds of biomaterials clinically used in plastic surgery and their applications are diverse. Moreover, with the rise of three-dimensional printing technology in recent years, the macroscopically more precise and personalized bio-scaffolding materials with microporous structure have made good progress, which is thought to bring new development to biomaterials. Therefore, in this paper, we reviewed the plastic surgery biomaterials and current progress in their 3D manufacturing technology.
Collapse
Affiliation(s)
- Wei Peng
- Department of Palliative Care, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China;
- Occupational Health Emergency Key Laboratory of West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiyu Peng
- Department of Thoracic Surgery, West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Pei Tang
- Department of Burn and Plastic Surgery, West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; (P.T.); (Z.L.)
| | - Huan Sun
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; (H.S.); (H.L.); (C.Z.)
| | - Haoyuan Lei
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; (H.S.); (H.L.); (C.Z.)
| | - Zhengyong Li
- Department of Burn and Plastic Surgery, West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; (P.T.); (Z.L.)
| | - Didi Hui
- Innovatus Oral Cosmetic & Surgical Institute, Norman, OK 73069, USA; (D.H.); (C.D.)
| | - Colin Du
- Innovatus Oral Cosmetic & Surgical Institute, Norman, OK 73069, USA; (D.H.); (C.D.)
| | - Changchun Zhou
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; (H.S.); (H.L.); (C.Z.)
| | - Yongwei Wang
- Department of Palliative Care, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China;
- Occupational Health Emergency Key Laboratory of West China Fourth Hospital, Sichuan University, Chengdu 610041, China
- Correspondence:
| |
Collapse
|
84
|
Alshaikh AB, Padma AM, Dehlin M, Akouri R, Song MJ, Brännström M, Hellström M. Decellularization and recellularization of the ovary for bioengineering applications; studies in the mouse. Reprod Biol Endocrinol 2020; 18:75. [PMID: 32703228 PMCID: PMC7376865 DOI: 10.1186/s12958-020-00630-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 07/17/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Fertility preservation is particularly challenging in young women diagnosed with hematopoietic cancers, as transplantation of cryopreserved ovarian cortex in these women carries the risk for re-introducing cancer cells. Therefore, the construction of a bioengineered ovary that can accommodate isolated small follicles was proposed as an alternative to minimize the risk of malignancy transmission. Various options for viable bioengineered scaffolds have been reported in the literature. Previously, we reported three protocols for producing mouse ovarian scaffolds with the decellularization technique. The present study examined these scaffolds further, specifically with regards to their extracellular composition, biocompatibility and ability to support recellularization with mesenchymal stem cells. MATERIAL AND METHODS Three decellularization protocols based on 0.5% sodium dodecyl sulfate (Protocol 1; P1), or 2% sodium deoxycholate (P2), or a combination of the two detergents (P3) were applied to produce three types of scaffolds. The levels of collagen, elastin and sulfated glycosaminoglycans (sGAGs) were quantified in the remaining extracellular matrix. Detailed immunofluorescence and scanning electron microscopy imaging were conducted to assess the morphology and recellularization efficiency of the constructs after 14 days in vitro utilizing red fluorescent protein-labelled mesenchymal stem cells. RESULTS All protocols efficiently removed the DNA while the elastin content was not significantly reduced during the procedures. The SDS-protocol (P1) reduced the sGAG and the collagen content more than the SDC-protocol (P2). All scaffolds were biocompatible and recellularization was successful, particularly in several P2-derived scaffolds. The cells were extensively distributed throughout the constructs, with a denser distribution observed towards the ovarian cortex. The cell density was not significantly different (400 to 550 cells/mm2) between scaffold types. However, there was a tendency towards a higher cell density in the SDC-derived constructs. Scanning electron microscope images showed fibrous scaffolds with a dense repopulated surface structure. CONCLUSIONS While there were differences in the key structural macromolecules between protocols, all scaffolds were biocompatible and showed effective recellularization. The results indicate that our SDC-protocol might be better than our SDS-protocol. However, additional studies are necessary to determine their suitability for attachment of small follicles and folliculogenesis.
Collapse
Affiliation(s)
- Ahmed Baker Alshaikh
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Kvinnokliniken, Blå stråket 6, SE-413 45, Göteborg, Sweden
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Arvind Manikantan Padma
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Kvinnokliniken, Blå stråket 6, SE-413 45, Göteborg, Sweden
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Matilda Dehlin
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Kvinnokliniken, Blå stråket 6, SE-413 45, Göteborg, Sweden
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Randa Akouri
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Kvinnokliniken, Blå stråket 6, SE-413 45, Göteborg, Sweden
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Min Jong Song
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Kvinnokliniken, Blå stråket 6, SE-413 45, Göteborg, Sweden
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Obstetrics & Gynecology, Yeouido St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mats Brännström
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Kvinnokliniken, Blå stråket 6, SE-413 45, Göteborg, Sweden
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Stockholm IVF-EUGIN, Stockholm, Sweden
| | - Mats Hellström
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Kvinnokliniken, Blå stråket 6, SE-413 45, Göteborg, Sweden.
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
85
|
Liu D, Cheng F, Pan S, Liu Z. Stem cells: a potential treatment option for kidney diseases. Stem Cell Res Ther 2020; 11:249. [PMID: 32586408 PMCID: PMC7318741 DOI: 10.1186/s13287-020-01751-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 05/26/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023] Open
Abstract
The prevalence of kidney diseases is emerging as a public health problem. Stem cells (SCs), currently considered as a promising tool for therapeutic application, have aroused considerable interest and expectations. With self-renewal capabilities and great potential for proliferation and differentiation, stem cell therapy opens new avenues for the development of renal function and structural repair in kidney diseases. Mounting evidence suggests that stem cells exert a therapeutic effect mainly by replacing damaged tissues and paracrine pathways. The benefits of various types of SCs in acute kidney disease and chronic kidney disease have been demonstrated in preclinical studies, and preliminary results of clinical trials present its safety and tolerability. This review will focus on the stem cell-based therapy approaches for the treatment of kidney diseases, including various cell sources used, possible mechanisms involved, and outcomes that are generated so far, along with prospects and challenges in clinical application.
Collapse
Affiliation(s)
- Dongwei Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, People's Republic of China
- Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, 450052, People's Republic of China
| | - Fei Cheng
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, People's Republic of China
- Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, 450052, People's Republic of China
| | - Shaokang Pan
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, People's Republic of China
- Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, 450052, People's Republic of China
| | - Zhangsuo Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, People's Republic of China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, People's Republic of China.
- Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, 450052, People's Republic of China.
| |
Collapse
|
86
|
Nyström H. Extracellular matrix proteins in metastases to the liver - Composition, function and potential applications. Semin Cancer Biol 2020; 71:134-142. [PMID: 32526353 DOI: 10.1016/j.semcancer.2020.06.004] [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: 02/29/2020] [Revised: 06/04/2020] [Accepted: 06/04/2020] [Indexed: 02/08/2023]
Abstract
The rising evidence of the tumor microenvironment (TME) and its role in cancer have made this an area of increased research efforts. The focus is both on the primary tumor but also on the metastatic setting. The TME though, does not only consist of the non-malignant cells of a tumor, but also of the acellular compartment: The Extracellular Matrix (ECM). The liver is a common organ for metastasis of many cancers and for some of these cancers' liver surgery is a standard treatment with long-term cure, whereas for other cancers not considered meaningful. Blood supply and anatomical reasons plays one part for the establishment of liver metastasis. It is however a well-known fact that the "soil" of a metastatic organ is of utter importance in the process of metastasis. The "soil" consists of the TME where the ECM is a critical and active part. This review focuses what is known about the normal ECM of the human liver, what is known about ECM proteins in human liver metastasis, challenges of studying the ECM in liver metastases and lastly, potential applications of this field of knowledge.
Collapse
Affiliation(s)
- Hanna Nyström
- Department of Surgical and Perioperative Sciences, Surgery, Umeå University, Sweden; Associated Researcher Wallenberg Centre for Molecular Medicine, Umeå University, Sweden.
| |
Collapse
|
87
|
Çankirili NK, Kart D, Çelebi-Saltik B. Evaluation of the biofilm formation of Staphylococcus aureus and Pseudomonas aeruginosa on human umbilical cord CD146+ stem cells and stem cell-based decellularized matrix. Cell Tissue Bank 2020; 21:215-231. [PMID: 32020424 DOI: 10.1007/s10561-020-09815-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 01/30/2020] [Indexed: 02/06/2023]
Abstract
This study aims to evaluate the CD146+ stem cells obtained from the human umbilical cord and their extracellular matrix proteins on in vitro Pseudomonas aeruginosa and Staphylococcus aureus biofilms to understand their possible antimicrobial activity. CD146+ stem cells were determined according to cell surface markers and differentiation capacity. Characterization of the decellularized matrix was done with DAPI, Masson's Trichrome staining and proteome analysis. Cell viability/proliferation of cells in co-cultures was evaluated by WST-1 and crystal-violet staining. The effects of cells and decellularized matrix proteins on biofilms were investigated on a drip flow biofilm reactor and their effects on gene expression were determined by RT-qPCR. We observed that CD146/105+ stem cells could differentiate adipogenically and decellularized matrix showed negative DAPI and positive collagen staining with Masson' s Trichrome. Proteome analysis of the decellularized matrix revealed some matrix components and growth factors. Although the decellularized matrix significantly reduced the cell counts of P. aeruginosa, no significant difference was observed for S. aureus cells in both groups. Supporting data was obtained from the gene expression results of P. aeruginosa with the significant down-regulation of rhlR and lasR. For S. aureus, icaADBC genes were significantly up-regulated when grown on the decellularized matrix.
Collapse
Affiliation(s)
- Nur Kübra Çankirili
- Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, 06100, Sihhiye, Ankara, Turkey
- Center for Stem Cell Research and Development, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - Didem Kart
- Department of Pharmaceutical Microbiology, Hacettepe University Faculty of Pharmacy, 06100, Sihhiye, Ankara, Turkey
| | - Betül Çelebi-Saltik
- Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, 06100, Sihhiye, Ankara, Turkey.
- Center for Stem Cell Research and Development, Hacettepe University, 06100, Sihhiye, Ankara, Turkey.
| |
Collapse
|
88
|
Lyu J, Chen L, Zhang J, Kang X, Wang Y, Wu W, Tang H, Wu J, He Z, Tang K. A microfluidics-derived growth factor gradient in a scaffold regulates stem cell activities for tendon-to-bone interface healing. Biomater Sci 2020; 8:3649-3663. [PMID: 32458839 DOI: 10.1039/d0bm00229a] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Treatment of tendon-to-bone interface injury has long been challenging in sports medicine. The major obstacle lies with the complicated three-layer structure of the tissue that consists of a bone region with osteocytes, a tendon region with tenocytes and a transitional region with chondrocytes. Conventional tissue engineering approaches using simply biomaterial scaffolds, stem cells and combinations of them had limited abilities to reconstruct the gradient structure with normal biomechanical properties. We herein aim to construct a three-layer structure with bone marrow-derived stem cells and tendon stem cells cultured in a decellularized tendon scaffold, through application of a gradient of biological cues in the longitudinal direction of the scaffold that guides the stem cells to differentiate and remodel the extracellular matrix in response to different medium concentrations in different regions. A microfluidic chip, on which a tree-like flow pattern was implemented, was adopted to create the concentration gradient in a dichotomous manner. We screened for an optimized seeding ratio between the two stem cell types before incubation of the scaffold in the medium concentration gradient and surgical implantation. Histology and immunohistochemistry assessments, both qualitatively and semi-quantitatively, showed that the microfluidic system provided desired guidance to the seeded stem cells that the healing at 8-week post-implantation presented a similar structure to that of a normal tendon-to-bone interface, which was outstanding compared to treatments without gradient guidance, stem cells or scaffolds where chaotic and fibrotic structures were obtained. This strategy offers a potentially translational tissue engineering approach for better outcomes in tendon-to-bone healing.
Collapse
Affiliation(s)
- Jingtong Lyu
- Center of Sports Medicine of Orthopaedic Department, Southwest hospital, Third Military Medical University, Chongqing 400038, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Zhao X, Zhu Y, Laslett AL, Chan HF. Hepatic Differentiation of Stem Cells in 2D and 3D Biomaterial Systems. Bioengineering (Basel) 2020; 7:E47. [PMID: 32466173 PMCID: PMC7356247 DOI: 10.3390/bioengineering7020047] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/18/2020] [Accepted: 05/22/2020] [Indexed: 02/06/2023] Open
Abstract
A critical shortage of donor livers for treating end-stage liver failure signifies the urgent need for alternative treatment options. Hepatocyte-like cells (HLC) derived from various stem cells represent a promising cell source for hepatocyte transplantation, liver tissue engineering, and development of a bioartificial liver assist device. At present, the protocols of hepatic differentiation of stem cells are optimized based on soluble chemical signals introduced in the culture medium and the HLC produced typically retain an immature phenotype. To promote further hepatic differentiation and maturation, biomaterials can be designed to recapitulate cell-extracellular matrix (ECM) interactions in both 2D and 3D configurations. In this review, we will summarize and compare various 2D and 3D biomaterial systems that have been applied to hepatic differentiation, and highlight their roles in presenting biochemical and physical cues to different stem cell sources.
Collapse
Affiliation(s)
- Xiaoyu Zhao
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China; (X.Z.); (Y.Z.)
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Yanlun Zhu
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China; (X.Z.); (Y.Z.)
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Andrew L. Laslett
- CSIRO Manufacturing, Clayton, Victoria 3168, Australia;
- Australian Regenerative Medicine Institute, Monash University, Victoria 3800, Australia
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China; (X.Z.); (Y.Z.)
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
| |
Collapse
|
90
|
Tang SW, Tong WY, Pang SW, Voelcker NH, Lam YW. Deconstructing, Replicating, and Engineering Tissue Microenvironment for Stem Cell Differentiation. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:540-554. [PMID: 32242476 DOI: 10.1089/ten.teb.2020.0044] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
One of the most crucial components of regenerative medicine is the controlled differentiation of embryonic or adult stem cells into the desired cell lineage. Although most of the reported protocols of stem cell differentiation involve the use of soluble growth factors, it is increasingly evident that stem cells also undergo differentiation when cultured in the appropriate microenvironment. When cultured in decellularized tissues, for instance, stem cells can recapitulate the morphogenesis and functional specialization of differentiated cell types with speed and efficiency that often surpass the traditional growth factor-driven protocols. This suggests that the tissue microenvironment (TME) provides stem cells with a holistic "instructive niche" that harbors signals for cellular reprogramming. The translation of this into medical applications requires the decoding of these signals, but this has been hampered by the complexity of TME. This problem is often addressed by a reductionist approach, in which cells are exposed to substrates decorated with simple, empirically designed geometries, textures, and chemical compositions ("bottom-up" approach). Although these studies are invaluable in revealing the basic principles of mechanotransduction mechanisms, their physiological relevance is often uncertain. This review examines the recent progress of an alternative, "top-down" approach, in which the TME is treated as a holistic biological entity. This approach is made possible by recent advances in systems biology and fabrication technologies that enable the isolation, characterization, and reconstitution of TME. It is hoped that these new techniques will elucidate the nature of niche signals so that they can be extracted, replicated, and controlled. This review summarizes these emerging techniques and how the data they generated are changing our view on TME. Impact statement This review summarizes the current state of art of the understanding of instructive niche in the field of tissue microenvironment. Not only did we survey the use of different biochemical preparations as stimuli of stem cell differentiation and summarize the recent effort in dissecting the biochemical composition of these preparations, through the application of extracellular matrix (ECM) arrays and proteomics, but we also introduce the use of open-source, high-content immunohistochemistry projects in contributing to the understanding of tissue-specific composition of ECM. We believe this review would be highly useful for our peer researching in the same field. "Mr. Tulkinghorn is always the same… so oddly out of place and yet so perfectly at home." -Charles Dickens, Bleak House.
Collapse
Affiliation(s)
- Sze Wing Tang
- Department of Chemistry, City University of Hong Kong, Hong Kong, Hong Kong
| | - Wing Yin Tong
- Melbourne Center for Nanofabrication, Victorian Node of the Australian National Fabrication, Clayton, Australia.,Drug Delivery Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Stella W Pang
- Department of Electrical Engineering, City University of Hong Kong, Hong Kong, Hong Kong
| | - Nicolas H Voelcker
- Melbourne Center for Nanofabrication, Victorian Node of the Australian National Fabrication, Clayton, Australia.,Drug Delivery Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Yun Wah Lam
- Department of Chemistry, City University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
91
|
Shen W, Berning K, Tang SW, Lam YW. Rapid and Detergent-Free Decellularization of Cartilage. Tissue Eng Part C Methods 2020; 26:201-206. [PMID: 32126898 DOI: 10.1089/ten.tec.2020.0008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The use of decellularized tissues or organs as cell culture scaffolds has proven to be a promising approach for tissue engineering and regenerative medicine, as these decellularized tissues can provide the instructive niche for cell differentiation and functions. Cartilage is a largely avascular tissue with limited regenerative capacity. Lesions caused by arthritis can lead to severe cartilage degeneration. Previous studies have indicated that decellularized cartilage can be used as scaffolds that support the chondrogenic differentiation of adult stem cells. However, these decellularization protocols all require the use of denaturing agents, such as high salt and detergents, that lead to the artifactual disruption of the chemical and physical integrity of the tissue microenvironment. Here, we established a new decellularization method for cartilage, through a combined effect of freezing-thawing, sectioning, and sonication in water. This protocol achieved the complete removal of cells within minutes, instead of hours or days required by existing procedures, and does not use any detergent. The resulting decellularized cartilage preserved the native ultrastructure and biochemical contents, including glycosaminoglycans, which is typically depleted by traditional decellularization methods. Human mesenchymal stem cells could readily adhere onto the decellularized cartilage. Together, this work unveils a simple new method for decellularizing cartilage, which will be useful in studying how tissue microenvironment supports chondrocyte growth and functions. Impact statement In this study, we develop a simple, fast cartilage decellularization method that does not require any detergent, so that the decellularized cartilage chemistry is preserved. Traditional detergent-based decellularization removes the tissue biochemical contents (i.e., glycosaminoglycans). In this new water decellularization protocol, the biochemical contents of cartilage can be preserved. This allows the study of biochemistry and physical content in extracellular matrix as a whole, and this protocol would definitely be useful for studying the effect of tissue microenvironment in supporting chondrocyte growth and functions.
Collapse
Affiliation(s)
- Wei Shen
- Department of Chemistry, City University of Hong Kong, Kowloon, Hong Kong
| | - Karsten Berning
- Department of Chemistry, City University of Hong Kong, Kowloon, Hong Kong
| | - Sze Wing Tang
- Department of Chemistry, City University of Hong Kong, Kowloon, Hong Kong
| | - Yun Wah Lam
- Department of Chemistry, City University of Hong Kong, Kowloon, Hong Kong
| |
Collapse
|
92
|
Furuta T, Furuya K, Zheng YW, Oda T. Novel alternative transplantation therapy for orthotopic liver transplantation in liver failure: A systematic review. World J Transplant 2020; 10:64-78. [PMID: 32257850 PMCID: PMC7109592 DOI: 10.5500/wjt.v10.i3.64] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/10/2020] [Accepted: 03/24/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Orthotopic liver transplantation (OLT) is the only treatment for end-stage liver failure; however, graft shortage impedes its applicability. Therefore, studies investigating alternative therapies are plenty. Nevertheless, no study has comprehensively analyzed these therapies from different perspectives.
AIM To summarize the current status of alternative transplantation therapies for OLT and to support future research.
METHODS A systematic literature search was performed using PubMed, Cochrane Library and EMBASE for articles published between January 2010 and 2018, using the following MeSH terms: [(liver transplantation) AND cell] OR [(liver transplantation) AND differentiation] OR [(liver transplantation) AND organoid] OR [(liver transplantation) AND xenotransplantation]. Various types of studies describing therapies to replace OLT were retrieved for full-text evaluation. Among them, we selected articles including in vivo transplantation.
RESULTS A total of 89 studies were selected. There are three principle forms of treatment for liver failure: Xeno-organ transplantation, scaffold-based transplantation, and cell transplantation. Xeno-organ transplantation was covered in 14 articles, scaffold-based transplantation was discussed in 22 articles, and cell transplantation was discussed in 53 articles. Various types of alternative therapies were discussed: Organ liver, 25 articles; adult hepatocytes, 31 articles; fetal hepatocytes, three articles; mesenchymal stem cells (MSCs), 25 articles; embryonic stem cells, one article; and induced pluripotent stem cells, three articles and other sources. Clinical applications were discussed in 12 studies: Cell transplantation using hepatocytes in four studies, five studies using umbilical cord-derived MSCs, three studies using bone marrow-derived MSCs, and two studies using hematopoietic stem cells.
CONCLUSION The clinical applications are present only for cell transplantation. Scaffold-based transplantation is a comprehensive treatment combining organ and cell transplantations, which warrants future research to find relevant clinical applications.
Collapse
Affiliation(s)
- Tomoaki Furuta
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba-shi 305-8575, Ibaraki, Japan
| | - Kinji Furuya
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba-shi 305-8575, Ibaraki, Japan
| | - Yun-Wen Zheng
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba-shi 305-8575, Ibaraki, Japan
- Institute of Regenerative Medicine and Affiliated Hospital of Jiangsu University, Zhenjiang 212001, Jiangsu Province, China
- Department of Regenerative Medicine, School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
- Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Tatsuya Oda
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba-shi 305-8575, Ibaraki, Japan
| |
Collapse
|
93
|
Kaur S, Tripathi DM, Venugopal JR, Ramakrishna S. Advances in biomaterials for hepatic tissue engineering. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2020. [DOI: 10.1016/j.cobme.2020.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
94
|
The Fabrication and Evaluation of a Potential Biomaterial Produced with Stem Cell Sheet Technology for Future Regenerative Medicine. Stem Cells Int 2020; 2020:9567362. [PMID: 32104186 PMCID: PMC7035578 DOI: 10.1155/2020/9567362] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 11/04/2019] [Accepted: 11/29/2019] [Indexed: 12/17/2022] Open
Abstract
To date, the decellularized scaffold has been widely explored as a source of biological scaffolds for regenerative medicine. However, the acellular matrix derived from natural tissues and organs has a lot of defects, including the limited amount of autogenous tissue and surgical complication such as risk of blood loss, wound infection, pain, shock, and functional damage in the donor part of the body. In this study, we prepared acellular matrix using adipose-derived stem cell (ADSC) sheets and evaluate the cellular compatibility and immunoreactivity. The ADSC sheets were fabricated and subsequently decellularized using repeated freeze-thaw, Triton X-100 and SDS decellularization. Oral mucosal epithelial cells were seeded onto the decellularized ADSC sheets to evaluate the cell replantation ability, and silk fibroin was used as the control. Then, acellular matrix was transplanted onto subcutaneous tissue for 1 week or 3 weeks; H&E staining and immunohistochemical analysis of CD68 expression and quantitative real-time PCR (qPCR) were performed to evaluate the immunogenicity and biocompatibility. The ADSC sheet-derived ECM scaffolds preserved the three-dimensional architecture of ECM and retained the cytokines by Triton X-100 decellularization protocols. Compared with silk fibroin in vitro, the oral mucosal epithelial cells survived better on the decellularized ADSC sheets with an intact and consecutive epidermal cellular layer. Compared with porcine small intestinal submucosa (SIS) in vivo, the homogeneous decellularized ADSC sheets had less monocyte-macrophage infiltrating in vivo implantation. During 3 weeks after transplantation, the mRNA expression of cytokines, such as IL-4/IL-10, was obviously higher in decellularized ADSC sheets than that of porcine SIS. A Triton X-100 method can achieve effective cell removal, retain major ECM components, and preserve the ultrastructure of ADSC sheets. The decellularized ADSC sheets possess good recellularization capacity and excellent biocompatibility. This study demonstrated the potential suitability of utilizing acellular matrix from ADSC sheets for soft tissue regeneration and repair.
Collapse
|
95
|
Liu G, David BT, Trawczynski M, Fessler RG. Advances in Pluripotent Stem Cells: History, Mechanisms, Technologies, and Applications. Stem Cell Rev Rep 2020; 16:3-32. [PMID: 31760627 PMCID: PMC6987053 DOI: 10.1007/s12015-019-09935-x] [Citation(s) in RCA: 257] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Over the past 20 years, and particularly in the last decade, significant developmental milestones have driven basic, translational, and clinical advances in the field of stem cell and regenerative medicine. In this article, we provide a systemic overview of the major recent discoveries in this exciting and rapidly developing field. We begin by discussing experimental advances in the generation and differentiation of pluripotent stem cells (PSCs), next moving to the maintenance of stem cells in different culture types, and finishing with a discussion of three-dimensional (3D) cell technology and future stem cell applications. Specifically, we highlight the following crucial domains: 1) sources of pluripotent cells; 2) next-generation in vivo direct reprogramming technology; 3) cell types derived from PSCs and the influence of genetic memory; 4) induction of pluripotency with genomic modifications; 5) construction of vectors with reprogramming factor combinations; 6) enhancing pluripotency with small molecules and genetic signaling pathways; 7) induction of cell reprogramming by RNA signaling; 8) induction and enhancement of pluripotency with chemicals; 9) maintenance of pluripotency and genomic stability in induced pluripotent stem cells (iPSCs); 10) feeder-free and xenon-free culture environments; 11) biomaterial applications in stem cell biology; 12) three-dimensional (3D) cell technology; 13) 3D bioprinting; 14) downstream stem cell applications; and 15) current ethical issues in stem cell and regenerative medicine. This review, encompassing the fundamental concepts of regenerative medicine, is intended to provide a comprehensive portrait of important progress in stem cell research and development. Innovative technologies and real-world applications are emphasized for readers interested in the exciting, promising, and challenging field of stem cells and those seeking guidance in planning future research direction.
Collapse
Affiliation(s)
- Gele Liu
- Department of Neurosurgery, Rush University Medical College, 1725 W. Harrison St., Suite 855, Chicago, IL, 60612, USA.
| | - Brian T David
- Department of Neurosurgery, Rush University Medical College, 1725 W. Harrison St., Suite 855, Chicago, IL, 60612, USA
| | - Matthew Trawczynski
- Department of Neurosurgery, Rush University Medical College, 1725 W. Harrison St., Suite 855, Chicago, IL, 60612, USA
| | - Richard G Fessler
- Department of Neurosurgery, Rush University Medical College, 1725 W. Harrison St., Suite 855, Chicago, IL, 60612, USA
| |
Collapse
|
96
|
Ebrahimi Sadrabadi A, Baei P, Hosseini S, Baghaban Eslaminejad M. Decellularized Extracellular Matrix as a Potent Natural Biomaterial for Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1341:27-43. [PMID: 32166633 DOI: 10.1007/5584_2020_504] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Decellularization technique is a favorable method used to fabricate natural and tissue-like scaffolds. This technique is important because of its remarkable ability to perfectly mimic the natural extracellular matrix (ECM). ECM-based scaffolds/hydrogels provide structural support for cell differentiation and maturation. Therefore, novel natural-based bioinks, ECM-based hydrogels, and particulate forms of the ECM provide promising strategies for whole organ regeneration. Despite its efficacious characteristics, removal of residual detergent and the presence of various protocols make this technique challenging for scientists and regenerative medicine-related programs. This chapter reviews the most effective physical, chemical, and enzymatic protocols used to remove the cellular components and their challenges. We discuss the applications of decellularized ECM (dECM) in tissue engineering and regenerative medicine with an emphasis on hard tissues.
Collapse
Affiliation(s)
- Amin Ebrahimi Sadrabadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Payam Baei
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Samaneh Hosseini
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran. .,Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
97
|
Liao J, Xu B, Zhang R, Fan Y, Xie H, Li X. Applications of decellularized materials in tissue engineering: advantages, drawbacks and current improvements, and future perspectives. J Mater Chem B 2020; 8:10023-10049. [PMID: 33053004 DOI: 10.1039/d0tb01534b] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Decellularized materials (DMs) are attracting more and more attention in tissue engineering because of their many unique advantages, and they could be further improved in some aspects through various means.
Collapse
Affiliation(s)
- Jie Liao
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education
- School of Biological Science and Medical Engineering
- Beijing Advanced Innovation Center for Biomedical Engineering
- Beihang University
- Beijing 100083
| | - Bo Xu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education
- School of Biological Science and Medical Engineering
- Beijing Advanced Innovation Center for Biomedical Engineering
- Beihang University
- Beijing 100083
| | - Ruihong Zhang
- Department of Research and Teaching
- the Fourth Central Hospital of Baoding City
- Baoding 072350
- China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education
- School of Biological Science and Medical Engineering
- Beijing Advanced Innovation Center for Biomedical Engineering
- Beihang University
- Beijing 100083
| | - Huiqi Xie
- Laboratory of Stem Cell and Tissue Engineering
- State Key Laboratory of Biotherapy and Cancer Center
- West China Hospital
- Sichuan University and Collaborative Innovation Center of Biotherapy
- Chengdu 610041
| | - Xiaoming Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education
- School of Biological Science and Medical Engineering
- Beijing Advanced Innovation Center for Biomedical Engineering
- Beihang University
- Beijing 100083
| |
Collapse
|
98
|
Moreno-Manzano V, Mellado-López M, Morera-Esteve MJ, Alastrue-Agudo A, Bisbal-Velasco V, Forteza-Vila J, Serrano-Aroca Á, Vera-Donoso CD. Human adipose-derived mesenchymal stem cells accelerate decellularized neobladder regeneration. Regen Biomater 2019; 7:161-169. [PMID: 32296535 PMCID: PMC7147364 DOI: 10.1093/rb/rbz049] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/11/2019] [Accepted: 11/22/2019] [Indexed: 12/16/2022] Open
Abstract
Decellularized natural bladder matrices (neobladders) represent an exciting means to regenerate the bladder following bladder cancer-associated cystectomy. In this study, we compare the evolution of decellularized matrices with recellularized matrices by seeding it with human adipose-derived mesenchymal stem cells (ADSC) after implantation following partial cystectomy in rats. We discovered significant anatomical differences since 10 days after neobladder implantation with the ADSC-containing matrices promoting a significant recovery of mature p63- and cytokeratin 7-positive urothelium. We also discovered significantly induced expression of the vimentin mesoderm marker in the submucosal layer in ADSC-seeded matrices. Interestingly, we found a higher expression of smooth muscle actin in transversal and longitudinal smooth muscle layers with ADSC-seeded matrices. Furthermore, ADSC also showed increased vascularization and nerve innervation of the neobladder as determined by the distribution of CD31 and S100β reactivity, respectively. We believe that ADSC and their paracrine-acting pro-regenerative secretome within decellularized matrices represent an efficient bladder substitution strategy; however, we require a fuller understanding of the mechanisms involved before clinical studies can begin.
Collapse
Affiliation(s)
- Victoria Moreno-Manzano
- Neuronal and Tissue Regeneration Lab, Centro de Investigación Príncipe Felipe, Valencia, Spain.,Facultad de Veterinaria y Ciencias Experimentales, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, Valencia 46001, Spain
| | | | | | - Ana Alastrue-Agudo
- Neuronal and Tissue Regeneration Lab, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Viviana Bisbal-Velasco
- Neuronal and Tissue Regeneration Lab, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Jerónimo Forteza-Vila
- Molecular Pathology and Translational Research in Oncology, Unidad Mixta Universidad Católica de Valencia y Centro de Investigación Príncipe Felipe, Spain
| | - Ángel Serrano-Aroca
- Facultad de Veterinaria y Ciencias Experimentales, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, Valencia 46001, Spain
| | - César David Vera-Donoso
- Department of Urology, La Fe University and Polytechnic Hospital and Health Research Institute, Hospital La Fe, Valencia 46026, Spain
| |
Collapse
|
99
|
Mukhamadiyarov RA, Rutkovskaya NV, Mil'to IV, Sidorova OD, Barbarash LS. [The cellular composition of explanted bioprosthetic heart valves in infective endocarditis]. Arkh Patol 2019; 81:16-23. [PMID: 31851188 DOI: 10.17116/patol20198106116] [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] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To perform immunohistochemical typing of cells as a component of bioprosthetic (BP) heart valves explanted during reoperations for prosthetic valve endocarditis. MATERIAL AND METHODS The authors investigated 8 models of KemCor and PeriCor artificial heart valves produced by NeoCor Company (Kemerovo, Russia), which were explanted from the mitral position due to infection of xenogeneic implanted material. The following markers: CD3 (T-lymphocytes), CD20 (B-lymphocytes), CD34 and VEGFR2 (endotheliocytes), CD68 (monocytes/macrophages), vimentin (fibroblasts), and α-smooth muscle actin (smooth muscle cells), were used for immunohistochemical typing of cells as a component of the analyzed samples. RESULTS Recipient cells were found to colonize devitalized BP tissues in infective endocarditis. This process simultaneously involved several types of cells performing their functions in infectious lesion and its initiation of BP remodeling. Macrophages contributed to the sanitation of the foci of infection and destruction of BP xenotissue; endotheliocytes ensured neovascularization and resistance of the implanted valve surface to infection; fibroblasts played a role in the neoplastic transformation of collagen, and smooth muscle cells were likely to take on the role in forming the elastic framework of a leaflet and in ensuring the mechanical properties of the bioprosthesis. CONCLUSION In the time course of development of prosthetic endocarditis, the recipient cells populate xenovalve leaflets that are a modified extracellular matrix obtained from the porcine aortic valve complex. This process is a consequence of the destruction of the BP surface and deep components. The observed cellular reactions are likely to be adaptive and to be aimed at eliminating microorganisms and regenerating structural damages.
Collapse
Affiliation(s)
- R A Mukhamadiyarov
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - N V Rutkovskaya
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - I V Mil'to
- Siberian State Medical University, Ministry of Health of Russia, Tomsk, Russia; National Research Tomsk Polytechnic University, Tomsk, Russia
| | - O D Sidorova
- Kemerovo State Medical University, Ministry of Health of Russia, Kemerovo, Russia
| | - L S Barbarash
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| |
Collapse
|
100
|
Nikolova MP, Chavali MS. Recent advances in biomaterials for 3D scaffolds: A review. Bioact Mater 2019; 4:271-292. [PMID: 31709311 PMCID: PMC6829098 DOI: 10.1016/j.bioactmat.2019.10.005] [Citation(s) in RCA: 427] [Impact Index Per Article: 85.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/07/2019] [Accepted: 10/15/2019] [Indexed: 02/06/2023] Open
Abstract
Considering the advantages and disadvantages of biomaterials used for the production of 3D scaffolds for tissue engineering, new strategies for designing advanced functional biomimetic structures have been reviewed. We offer a comprehensive summary of recent trends in development of single- (metal, ceramics and polymers), composite-type and cell-laden scaffolds that in addition to mechanical support, promote simultaneous tissue growth, and deliver different molecules (growth factors, cytokines, bioactive ions, genes, drugs, antibiotics, etc.) or cells with therapeutic or facilitating regeneration effect. The paper briefly focuses on divers 3D bioprinting constructs and the challenges they face. Based on their application in hard and soft tissue engineering, in vitro and in vivo effects triggered by the structural and biological functionalized biomaterials are underlined. The authors discuss the future outlook for the development of bioactive scaffolds that could pave the way for their successful imposing in clinical therapy.
Collapse
Affiliation(s)
- Maria P. Nikolova
- Department of Material Science and Technology, University of Ruse “A. Kanchev”, 8 Studentska Str., 7000, Ruse, Bulgaria
| | - Murthy S. Chavali
- Shree Velagapudi Ramakrishna Memorial College (PG Studies, Autonomous), Nagaram, 522268, Guntur District, India
- PG Department of Chemistry, Dharma Appa Rao College, Nuzvid, 521201, Krishna District, India
- MCETRC, Tenali, 522201, Guntur District, Andhra Pradesh, India
| |
Collapse
|