1
|
Janarthanan R, Jayakumar R, Iyer S. Injectable Pectin-Alginate Hydrogels for Improving Vascularization and Adipogenesis of Human Fat Graft. J Funct Biomater 2023; 14:409. [PMID: 37623654 PMCID: PMC10455938 DOI: 10.3390/jfb14080409] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/26/2023] Open
Abstract
Autologous fat grafting (AFG) is the most prevailing tool for soft tissue regeneration in clinics, although efficiency is limited to unpredictable volume resorption due to poor vascularization and eventual necrosis. This study sought to improve the AFG efficiency using a hydrogel as a carrier for human fat graft (F) with and without platelet-rich plasma (PRP). PRP is clinically well known for the local release of several endogenous growth factors and has been in clinical use already. A human-fat-graft-encapsulated pectin-alginate hydrogel (FG) was developed and characterized. PRP was added to F to develop a human fat graft with PRP (FP). FP was admixed with a pectin-alginate hydrogel to develop FGP. FG and FGP showed the smooth injectable, elastic, and shear-thinning properties. FG and FGP groups showed enhanced cell viability and proliferation compared to the control F in vitro. We also investigated the in vivo angiogenesis and neo-adipogenesis ability of F, FG, FGP, and FP in nude mice after subcutaneous injection. After 2 and 4 weeks, an MRI of the mice was conducted, followed by graft explantation. The explanted grafts were also assessed histologically and with immunohistochemistry (IHC) studies. MRI and histology results revealed better vascularity of the FG and FGP system compared to fat graft alone. Further, the IHC studies, CD 31, and perilipin staining also revealed better vasculature and adipogenesis of FG and FGP systems. These results indicate the enhanced angiogenesis and adipogenesis of FG and FGP. Thus, developed pectin-alginate hydrogel-based fat graft systems FG and FGP replenish the native microenvironment by mediating angiogenesis and adipogenesis, thereby maximizing the clinical outcomes of autologous fat grafting.
Collapse
Affiliation(s)
- Ramu Janarthanan
- Department of Plastic and Reconstructive Surgery, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi 682041, India;
| | - Rangasamy Jayakumar
- Polymeric Biomaterials Lab, School of Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi 682041, India;
| | - Subramania Iyer
- Department of Plastic and Reconstructive Surgery, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi 682041, India;
| |
Collapse
|
2
|
Zhang HJ, Li FS, Wang F, Wang H, He TC, Reid RR, He BC, Xia Q. Transgenic PDGF-BB sericin hydrogel potentiates bone regeneration of BMP9-stimulated mesenchymal stem cells through a crosstalk of the Smad-STAT pathways. Regen Biomater 2022; 10:rbac095. [PMID: 36683747 PMCID: PMC9847547 DOI: 10.1093/rb/rbac095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/08/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
Silk as a natural biomaterial is considered as a promising bone substitute in tissue regeneration. Sericin and fibroin are the main components of silk and display unique features for their programmable mechanical properties, biocompatibility, biodegradability and morphological plasticity. It has been reported that sericin recombinant growth factors (GFs) can support cell proliferation and induce stem cell differentiation through cross-talk of signaling pathways during tissue regeneration. The transgenic technology allows the productions of bioactive heterologous GFs as fusion proteins with sericin, which are then fabricated into solid matrix or hydrogel format. Herein, using an injectable hydrogel derived from transgenic platelet-derived GF (PDGF)-BB silk sericin, we demonstrated that the PDGF-BB sericin hydrogel effectively augmented osteogenesis induced by bone morphogenetic protein (BMP9)-stimulated mesenchymal stem cells (MSCs) in vivo and in vitro, while inhibiting adipogenic differentiation. Further gene expression and protein-protein interactions studies demonstrated that BMP9 and PDGF-BB synergistically induced osteogenic differentiation through the cross-talk between Smad and Stat3 pathways in MSCs. Thus, our results provide a novel strategy to encapsulate osteogenic factors and osteoblastic progenitors in transgenic sericin-based hydrogel for robust bone tissue engineering.
Collapse
Affiliation(s)
- Hui-Jie Zhang
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
| | - Fu-Shu Li
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Department of Pharmacy, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing 400014, China
| | - Feng Wang
- Biological Science Research Center, Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing 400715, China
| | - Han Wang
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Department of Pharmacy, Panzhou People’s Hospital, Guizhou 553599, China
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Russell R Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Laboratory of Craniofacial Biology and Development, Department of Surgery Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Bai-Cheng He
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Qingyou Xia
- Biological Science Research Center, Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing 400715, China
| |
Collapse
|
3
|
Xia B, Chen G. Research progress of natural tissue-derived hydrogels for tissue repair and reconstruction. Int J Biol Macromol 2022; 214:480-491. [PMID: 35753517 DOI: 10.1016/j.ijbiomac.2022.06.137] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/05/2022] [Accepted: 06/20/2022] [Indexed: 12/26/2022]
Abstract
There are many different grafts to repair damaged tissue. Various types of biological scaffolds, including films, fibers, microspheres, and hydrogels, can be used for tissue repair. A hydrogel, which is composed a natural or synthetic polymer network with high water absorption capacity, can provide a microenvironment closely resembling the extracellular matrix (ECM) of natural tissues to stimulate cell adhesion, proliferation, and differentiation. It has been shown to have great application potential in the field of tissue repair and regeneration. Hydrogels derived from natural tissues retain a variety of proteins and growth factors in optimal proportions, which is beneficial for the regeneration of specific tissues. This article reviews the latest research advances in the field of hydrogels from a variety of natural tissue sources, including bone tissue, blood vessels, nerve tissue, adipose tissue, skin tissue, and muscle tissue, including preparation methods, advantages, and applications in tissue engineering and regenerative medicine. Finally, it summarizes and discusses the challenges faced by natural tissue-derived hydrogels used in tissue repair, as well as future research and application directions.
Collapse
Affiliation(s)
- Bin Xia
- Engineering Research Center for Waste Oil Recovery Technology and Equipment, Ministry of Education, Chongqing Technology and Business University, Chongqing 400067, PR China
| | - Guobao Chen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China; Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, PR China.
| |
Collapse
|
4
|
Sheng H, Pan C, Wang S, Yang C, Zhang J, Hu C, Hu H, Feng X, Yang M, Lei Z, Gao Y, Wang Z, Ma Y. Weighted Gene Co-Expression Network Analysis Identifies Key Modules and Central Genes Associated With Bovine Subcutaneous Adipose Tissue. Front Vet Sci 2022; 9:914848. [PMID: 35812879 PMCID: PMC9257221 DOI: 10.3389/fvets.2022.914848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background Fat deposition is an important economic trait in livestock and poultry production. However, the relationship between various genes and signal pathways of fat deposition is still unclear to a large extent. The purpose of this study is to analyze the potential molecular targets and related molecular pathways in bovine subcutaneous adipose tissue. Results We downloaded the GSE116775 microarray dataset from Gene Expression Omnibus (GEO). The weighted gene co-expression network (WGCNA) was used to analyze the gene expression profile, and the key gene modules with the highest correlation with subcutaneous adipose tissue were identified, and the functional enrichment of the key modules was analyzed. Then, the “real” Hub gene was screened by in-module analysis and protein–protein interaction network (PPI), and its expression level in tissue samples and adipocytes was verified. The study showed that a total of nine co-expression modules were identified, and the number of genes in these modules ranged from 101 to 1,509. Among them, the blue module is most closely related to subcutaneous adipose tissue, containing 1,387 genes. These genes were significantly enriched in 10 gene ontologies including extracellular matrix organization, biological adhesion, and collagen metabolic process, and were mainly involved in pathways including ECM-receptor interaction, focal adhesion, cAMP signaling pathway, PI3K-AKT signaling pathway, and regulation of lipolysis in adipocytes. In the PPI network and coexpression network, five genes (CAV1, ITGA5, COL5A1, ABL1, and HSPG2) were identified as “real” Hub genes. Analysis of Hub gene expression by dataset revealed that the expression of these Hub genes was significantly higher in subcutaneous adipose tissue than in other tissues. In addition, real-time fluorescence quantitative PCR (qRT-PCR) analysis based on tissue samples and adipocytes also confirmed the above results. Conclusion In this study, five key genes related to subcutaneous adipose tissue were discovered, which laid a foundation for further study of the molecular regulation mechanism of subcutaneous adipose tissue development and adipose deposition.
Collapse
Affiliation(s)
- Hui Sheng
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China
| | - Cuili Pan
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China
| | - Shuzhe Wang
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China
| | - Chaoyun Yang
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China
| | - Junxing Zhang
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China
| | - Chunli Hu
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China
| | - Honghong Hu
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China
| | - Xue Feng
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China
| | - Mengli Yang
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China
| | - Zhaoxiong Lei
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China
| | - Yuhong Gao
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China
| | - Zhong Wang
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China
| | - Yun Ma
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan, China
- College of Life Sciences, Xinyang Normal University, Xinyang, China
- *Correspondence: Yun Ma
| |
Collapse
|
5
|
Zhang G, Ci H, Ma C, Li Z, Jiang W, Chen L, Wang Z, Zhou M, Sun J. Additive manufactured macroporous chambers facilitate large volume soft tissue regeneration from adipose-derived extracellular matrix. Acta Biomater 2022; 148:90-105. [PMID: 35671873 DOI: 10.1016/j.actbio.2022.05.053] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 05/12/2022] [Accepted: 05/31/2022] [Indexed: 12/12/2022]
Abstract
Breast tissue engineering is a promising alternative intervention for breast reconstruction. Due to their low immunogenicity and well-preserved adipogenic microenvironment, decellularized adipose tissue (DAT) can potentially regenerate adipose tissue in vivo. However, the volume of adipose tissue regenerated from DAT can hardly satisfy the demand for breast reconstruction. Tissue engineering chamber (TEC) is an effective technique for generation of large adipose tissue volumes. However, TEC applications necessitate reoperation to remove non-degradable plastic chambers and harvest autologous tissue flaps, which prolongs the operation time and causes potential damage to donor sites. We improved the TEC strategy by combining bioresorbable polycaprolactone (PCL) chambers and decellularized adipose tissues (DAT). A miniaturized porous PCL chamber was fabricated based on scaling differences between human and rabbit chests, and basic fibroblast growth factor (bFGF)-loaded DAT successfully prepared. In rabbit models, a highly vascularized adipose tissue that nearly filled up the PCL chamber (5 mL) was generated de novo from 0.5 mL bFGF-loaded DAT. The newly formed tissue had significantly high expressions of adipogenic genes, compared to the endogenous adipose tissue. The concept described here can be exploited for breast tissue engineering. STATEMENT OF SIGNIFICANCE: Decellularized adipose tissue (DAT), which provides infiltrated cells adipogenic microenvironment, can potentially regenerate adipose tissue in vivo. Nevertheless, the volume of regenerated adipose tissue is insufficient to repair large sized tissue defect. Tissue engineering chamber (TEC) could provide a protective space for in situ regeneration of large volume tissue. Herein, a new strategy by combining biodegradable polycaprolactone chambers and basic fibroblast growth factor-loaded decellularized adipose tissue is proposed. In rabbit model, newly formed adipose tissue regenerated from DAT successfully filled the dome shaped chamber with ten folds higher volume than DAT, which is proportionally similar to women breast. This work highlighted the importance of adipogenic microenvironment and protective space for adipose tissue regeneration.
Collapse
Affiliation(s)
- Guo Zhang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan 430022, China
| | - Hai Ci
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan 430022, China; Department of Burn and Plastic Surgery, the First Affiliated Hospital of Medical College of Shihezi University, Shihezi, Xinjiang 832008, China
| | - Chenggong Ma
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhipeng Li
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan 430022, China
| | - Wenbin Jiang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan 430022, China
| | - Lifeng Chen
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan 430022, China
| | - Zhenxing Wang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan 430022, China
| | - Muran Zhou
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan 430022, China.
| | - Jiaming Sun
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan 430022, China.
| |
Collapse
|
6
|
Tang W, Qi J, Wang Q, Qu Y, Fu S, Luan J. Investigating the Adipogenic Effects of Different Tissue-Derived Decellularized Matrices. Front Bioeng Biotechnol 2022; 10:872897. [PMID: 35497363 PMCID: PMC9046558 DOI: 10.3389/fbioe.2022.872897] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/29/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: Decellularized adipose-derived matrix (DAM) can promote adipogenic differentiation and adipose tissue remodeling, but the biological impact of tissue origin on DAM remains unknown. The present study aimed to investigate the effects of tissue origins on the adipogenic capacity of the decellularized matrix by comparing the cellular and tissue responses of DAM versus acellular dermal matrix (ADM). Methods: The in vitro response of adipose-derived stem/stromal cells (ADSCs) to DAM and ADM was characterized by proliferation and differentiation. The in vivo remodeling response was evaluated in the subcutaneous injection model of immunocompromised mice, using histology, protein expression, and transcriptome analysis. Results: Both DAM and ADM exhibited excellent decellularization effects and cytocompatibility. In the absence of exogenous stimuli, DAM could induce adipogenic differentiation of ADSCs compared with ADM. In the animal model, the levels of PDGF, VEGF, and ACRP30 were higher in the DAM groups than in the ADM group, and more neovascularization and extensive adipose tissue remodeling were observed. The mRNA-seq analysis indicated that the DAM implant regulated tissue remodeling by modulating Lat1/2 expression along with Hippo Signaling pathway in the early stage. Conclusion: Tissue origin can influence the biological response of the decellularized matrix. DAM can retain favorable tissue-specific characteristics after the decellularization process and have unique adipogenic effects in vitro and vivo, which can be fully utilized for soft tissue repair and regeneration.
Collapse
Affiliation(s)
| | | | | | | | - Su Fu
- *Correspondence: Su Fu, ; Jie Luan,
| | - Jie Luan
- *Correspondence: Su Fu, ; Jie Luan,
| |
Collapse
|
7
|
Chen X, Lu F, Yuan Y. The Application and Mechanism of Action of External Volume Expansion in Soft Tissue Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2021; 27:181-197. [PMID: 32821009 DOI: 10.1089/ten.teb.2020.0137] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Xihang Chen
- Department of Plastic and Cosmetic Surgery, Southern Medical University, Nanfang Hospital, Guangzhou, China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Southern Medical University, Nanfang Hospital, Guangzhou, China
| | - Yi Yuan
- Department of Plastic and Cosmetic Surgery, Southern Medical University, Nanfang Hospital, Guangzhou, China
| |
Collapse
|
8
|
Yang J, Zhou C, Fu J, Yang Q, He T, Tan Q, Lv Q. In situ Adipogenesis in Biomaterials Without Cell Seeds: Current Status and Perspectives. Front Cell Dev Biol 2021; 9:647149. [PMID: 33763426 PMCID: PMC7982583 DOI: 10.3389/fcell.2021.647149] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/08/2021] [Indexed: 02/05/2023] Open
Abstract
For cosmetic and reconstructive purposes in the setting of small-volume adipose tissue damage due to aging, traumatic defects, oncological resections, and degenerative diseases, the current strategies for soft tissue replacement involve autologous fat grafts and tissue fillers with synthetic, bioactive, or tissue-engineered materials. However, they all have drawbacks such as volume shrinkage and foreign-body responses. Aiming to regenerate bioactive vascularized adipose tissue on biomaterial scaffolds, adipose tissue engineering (ATE) has emerged as a suitable substitute for soft tissue repair. The essential components of ATE include scaffolds as support, cells as raw materials for fat formation, and a tolerant local environment to allow regeneration to occur. The commonly loaded seeding cells are adipose-derived stem cells (ASCs), which are expected to induce stable and predictable adipose tissue formation. However, defects in stem cell enrichment, such as donor-site sacrifice, limit their wide application. As a promising alternative approach, cell-free bioactive scaffolds recruit endogenous cells for adipogenesis. In biomaterials without cell seeds, the key to sufficient adipogenesis relies on the recruitment of endogenous host cells and continuous induction of cell homing to scaffolds. Regeneration, rather than repair, is the fundamental dominance of an optimal mature product. To induce in situ adipogenesis, many researchers have focused on the mechanical and biochemical properties of scaffolds. In addition, efforts to regulate an angiogenic and adipogenic microenvironment in cell-free settings involve integrating growth factors or extracellular matrix (ECM) proteins onto bioactive scaffolds. Despite the theoretical feasibility and encouraging results in animal models, few of the reported cell-free biomaterials have been tested in humans, and failures of decellularized adipose tissues in adipogenesis have also been reported. In these cases, the most likely reason was the lack of supporting vasculature. This review summarizes the current status of biomaterials without cell seeds. Related mechanisms and influencing factors of in situ adipogenesis in cell-free biomaterials, dilemma in the development of biomaterials, and future perspectives are also addressed.
Collapse
Affiliation(s)
- Jiqiao Yang
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Tumor Targeted and Immune Therapy, Clinical Research Center for Breast Disease, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chen Zhou
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jingyang Fu
- West China School of Medicine/West China Hospital, Sichuan University, Chengdu, China
| | - Qianru Yang
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Tao He
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Qiuwen Tan
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qing Lv
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Xia Z, Guo X, Yu N, Zeng A, Si L, Long F, Zhang W, Wang X, Zhu L, Liu Z. The Application of Decellularized Adipose Tissue Promotes Wound Healing. Tissue Eng Regen Med 2020; 17:863-874. [PMID: 33165769 PMCID: PMC7710820 DOI: 10.1007/s13770-020-00286-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/07/2020] [Accepted: 07/20/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Due to adipose-derived stem cells (ASCs) being easy to obtain, their rapid proliferation rate, and their multidirectional differentiation capabilities, they have been widely used in the field of regenerative medicine. With the progress of decellularized adipose tissue (DAT) and adipose tissue engineering research, the role of DAT in promoting angiogenesis has gradually been emphasized. METHODS We examined the biological characteristics and biosafety of DAT and evaluated the stem cell maintenance ability and promotion of growth factor secretion through conducting in vitro and in vivo studies. RESULTS The tested ASCs showed high rat:es of proliferation and adhered well to DAT. The expression levels of essential genes for cell stem maintenance, including OCT4, SOX2, and Nanog were low at 2-24 h and much higher at 48 and 96 h. The Adipogenic expression level of markers for ASCs proliferation including PPARγ, C/EPBα, and LPL increased from 2 to 96 h. Co-culture of ASCs and DAT increased the secretion of local growth factors, such as VEGF, PDGF-bb, bFGF, HGF, EGF, and FDGF-bb, and secretion gradually increased from 0 to 48 h. A model of full-thickness skin defects on the back of nude mice was established, and the co-culture of ASCs and DAT showed the best in vivo treatment effect. CONCLUSION The application of DAT promotes wound healing, and DAT combined with ASCs may be a promising material in adipose tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Zenan Xia
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Dongcheng District, Beijing, 100730, China
| | - Xiao Guo
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Dongcheng District, Beijing, 100730, China
| | - Nanze Yu
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Dongcheng District, Beijing, 100730, China
| | - Ang Zeng
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Dongcheng District, Beijing, 100730, China
| | - Loubin Si
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Dongcheng District, Beijing, 100730, China
| | - Fei Long
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Dongcheng District, Beijing, 100730, China
| | - Wenchao Zhang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Dongcheng District, Beijing, 100730, China
| | - Xiaojun Wang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Dongcheng District, Beijing, 100730, China
| | - Lin Zhu
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Dongcheng District, Beijing, 100730, China.
| | - Zhifei Liu
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
10
|
Robb KP, Juignet L, Morissette Martin P, Walker JT, Brooks CR, Barreira C, Dekaban GA, Flynn LE. Adipose Stromal Cells Enhance Decellularized Adipose Tissue Remodeling Through Multimodal Mechanisms. Tissue Eng Part A 2020; 27:618-630. [PMID: 32873224 DOI: 10.1089/ten.tea.2020.0180] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Decellularized adipose tissue (DAT) scaffolds represent a promising cell-instructive platform for soft tissue engineering. While recent work has highlighted that mesenchymal stromal cells, including adipose-derived stromal cells (ASCs), can be combined with decellularized scaffolds to augment tissue regeneration, the mechanisms involved require further study. The objective of this work was to probe the roles of syngeneic donor ASCs and host-derived macrophages in tissue remodeling of DAT scaffolds within an immunocompetent mouse model. Dual transgenic reporter mouse strains were employed to track and characterize the donor ASCs and host macrophages within the DAT implants. More specifically, ASCs isolated from dsRed mice were seeded on DAT scaffolds, and the seeded and unseeded control scaffolds were implanted subcutaneously into MacGreen transgenic mice for up to 8 weeks. ASC seeding was shown to augment cell infiltration into the DAT implants at 8 weeks, and this was linked to significantly enhanced angiogenesis relative to the unseeded controls. Immunohistochemical staining demonstrated long-term retention of the syngeneic donor ASCs over the duration of the 8-week study, providing evidence that the DAT scaffolds are a cell-supportive delivery platform. Notably, newly formed adipocytes within the DAT implants were not dsRed+, indicating that the donor ASCs supported fat formation through indirect mechanisms. Immunohistochemical tracking of host macrophages through costaining for enhanced green fluorescent protein with the macrophage marker Iba1 revealed that ASC seeding significantly increased the number of infiltrating macrophages within the DAT implants at 3 weeks, while the fraction of macrophages relative to the total cellular infiltrate was similar between the groups at 1, 3, and 8 weeks. Consistent with the tissue remodeling response that was observed, western blotting demonstrated that there was significantly augmented expression of CD163 and CD206, markers of constructive M2-like macrophages, within the ASC-seeded DAT implants. Overall, our results demonstrate that exogenous ASCs enhance tissue regeneration within DAT scaffolds indirectly through multimodal mechanisms that include host cell recruitment and immunomodulation. These data provide further evidence to support the use of decellularized scaffolds as a delivery platform for ASCs in tissue engineering.
Collapse
Affiliation(s)
- Kevin P Robb
- School of Biomedical Engineering, University of Western Ontario, London, Canada
| | - Laura Juignet
- Department of Anatomy and Cell Biology and Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Pascal Morissette Martin
- Department of Anatomy and Cell Biology and Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - John T Walker
- Department of Anatomy and Cell Biology and Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Courtney R Brooks
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Christy Barreira
- Molecular Medicine Research Laboratories, Robarts Research Institute, University of Western Ontario, London, Canada
| | - Gregory A Dekaban
- Molecular Medicine Research Laboratories, Robarts Research Institute, University of Western Ontario, London, Canada.,Department of Microbiology & Immunology and University of Western Ontario, London, Canada
| | - Lauren E Flynn
- School of Biomedical Engineering, University of Western Ontario, London, Canada.,Department of Anatomy and Cell Biology and Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada.,Department of Chemical and Biochemical Engineering, University of Western Ontario, London, Canada.,Bone and Joint Institute, University of Western Ontario, London, Canada
| |
Collapse
|
11
|
Debels H, Palmer J, Han XL, Poon C, Abberton K, Morrison W. In vivo tissue engineering of an adipose tissue flap using fat grafts and Adipogel. J Tissue Eng Regen Med 2020; 14:633-644. [PMID: 32090506 DOI: 10.1002/term.3027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 01/27/2020] [Accepted: 02/04/2020] [Indexed: 12/17/2022]
Abstract
For decades, plastic surgeons have spent considerable effort exploring anatomical regions for free flap design. More recently, tissue-engineering approaches have been utilised in an attempt to grow transplantable tissue flaps in vivo. The aim of this study was to engineer a fat flap with a vascular pedicle by combining autologous fat grafts and a novel acellular hydrogel (Adipogel) in an established tissue-engineering model comprising a chamber and blood vessel loop. An arteriovenous loop was created in the rat groin from the femoral vessels and positioned inside a perforated polycarbonate chamber. In Group 1, the chamber contained minced, centrifuged autologous fat; in Group 2, Adipogel was added to the graft; and in Group 3, Adipogel alone was used. Constructs were histologically examined at 6 and 12 weeks. In all groups, new tissue was generated. Adipocytes, although appearing viable in the graft at the time of insertion, were predominantly nonviable at 6 weeks. However, by 12 weeks, new fat had formed in all groups and was significantly greater in the combined fat/Adipogel group. No significant difference was seen in final construct total volume or construct neovascularisation between the groups. This study demonstrated that a pedicled adipose flap can be generated in rats by combining a blood vessel loop, an adipogenic hydrogel, and a lipoaspirate equivalent. Success appears to be based on adipogenesis rather than on adipocyte survival, and consistent with our previous work, this adipogenesis occurred subsequent to graft death and remodelling. The regenerative process was significantly enhanced in the presence of Adipogel.
Collapse
Affiliation(s)
- Heidi Debels
- O'Brien Institute Department, St. Vincent's Institute, Fitzroy, Victoria, Australia.,Department of Plastic and Reconstructive Surgery, Free University Brussels (VUB), Belgium.,Department of Plastic Surgery, Maastricht University, Maastricht, The Netherlands
| | - Jason Palmer
- O'Brien Institute Department, St. Vincent's Institute, Fitzroy, Victoria, Australia.,University of Melbourne Department of Surgery, St. Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia
| | - Xiao-Lian Han
- O'Brien Institute Department, St. Vincent's Institute, Fitzroy, Victoria, Australia
| | - Christopher Poon
- O'Brien Institute Department, St. Vincent's Institute, Fitzroy, Victoria, Australia.,University of Melbourne Department of Surgery, St. Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia
| | - Keren Abberton
- O'Brien Institute Department, St. Vincent's Institute, Fitzroy, Victoria, Australia.,University of Melbourne Department of Surgery, St. Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia.,Faculty of Health Sciences, Australian Catholic University, Fitzroy, Victoria, Australia
| | - Wayne Morrison
- O'Brien Institute Department, St. Vincent's Institute, Fitzroy, Victoria, Australia.,University of Melbourne Department of Surgery, St. Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia.,Faculty of Health Sciences, Australian Catholic University, Fitzroy, Victoria, Australia
| |
Collapse
|
12
|
Zhang Z, Cai J, Li Y, He Y, Dong Z, Dai J, Lu F. External Volume Expansion Adjusted Adipose Stem Cell by Shifting the Ratio of Fibronectin to Laminin. Tissue Eng Part A 2020; 26:66-77. [PMID: 31347463 DOI: 10.1089/ten.tea.2019.0095] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Ziang Zhang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, P.R. China
| | - Junrong Cai
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Ye Li
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Yunfan He
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Ziqing Dong
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Jingxing Dai
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, P.R. China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| |
Collapse
|
13
|
Abstract
The ability to generate new microvessels in desired numbers and at desired locations has been a long-sought goal in vascular medicine, engineering, and biology. Historically, the need to revascularize ischemic tissues nonsurgically (so-called therapeutic vascularization) served as the main driving force for the development of new methods of vascular growth. More recently, vascularization of engineered tissues and the generation of vascularized microphysiological systems have provided additional targets for these methods, and have required adaptation of therapeutic vascularization to biomaterial scaffolds and to microscale devices. Three complementary strategies have been investigated to engineer microvasculature: angiogenesis (the sprouting of existing vessels), vasculogenesis (the coalescence of adult or progenitor cells into vessels), and microfluidics (the vascularization of scaffolds that possess the open geometry of microvascular networks). Over the past several decades, vascularization techniques have grown tremendously in sophistication, from the crude implantation of arteries into myocardial tunnels by Vineberg in the 1940s, to the current use of micropatterning techniques to control the exact shape and placement of vessels within a scaffold. This review provides a broad historical view of methods to engineer the microvasculature, and offers a common framework for organizing and analyzing the numerous studies in this area of tissue engineering and regenerative medicine. © 2019 American Physiological Society. Compr Physiol 9:1155-1212, 2019.
Collapse
Affiliation(s)
- Joe Tien
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
- Division of Materials Science and Engineering, Boston University, Brookline, Massachusetts, USA
| |
Collapse
|
14
|
Biocompatible Interface-Modified Tissue Engineering Chamber Reduces Capsular Contracture and Enlarges Regenerated Adipose Tissue. ACS Biomater Sci Eng 2019; 5:3440-3447. [PMID: 33405728 DOI: 10.1021/acsbiomaterials.8b00930] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
15
|
Chang Q, Cai J, Wang Y, Yang R, Xing M, Lu F. Large adipose tissue generation in a mussel-inspired bioreactor of elastic-mimetic cryogel and platelets. J Tissue Eng 2018; 9:2041731418808633. [PMID: 30505425 PMCID: PMC6259050 DOI: 10.1177/2041731418808633] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 09/26/2018] [Indexed: 12/16/2022] Open
Abstract
Soft tissue generation, especially in large tissue, is a major challenge in reconstructive surgery to treat congenital deformities, posttraumatic repair, and cancer rehabilitation. The concern is along with the donor site morbidity, donor tissue shortage, and flap necrosis. Here, we report a dissection-free adipose tissue chamber-based novel guided adipose tissue regeneration strategy in a bioreactor of elastic gelatin cryogel and polydopamine-assisted platelet immobilization intended to improve angiogenesis and generate large adipose tissue in situ. In order to have matched tissue mechanics, we used 5% gelatin cryogel as growth substrate of bioreactor. Platelets from the platelet-rich plasma were then immobilized onto the gelatin cryogel with the aid of polydopamine to form a biomimetic bioreactor (polydopamine/gelatin cryogel/platelet). Platelets on the substrate led to a sustained high release in both platelet-derived growth factor and vascular endothelial growth factor compared with non-polydopamine-assisted group. The formed bioreactor was then transferred to a tissue engineering chamber and then inserted above inguinal fat pad of rats without flap dissection. This integrate strategy significantly boomed the vessel density, stimulated cellular proliferation, and upregulated macrophage infiltration. There was a noticeable rise in the expression of dual-angiogenic growth factors (platelet-derived growth factor and vascular endothelial growth factor) in chamber fluid; host cell migration and host fibrous protein secretion coordinated with gelatin cryogel degradation. The regenerated adipose tissue volume gained threefold larger than control group (p < 0.05) with less fibrosis tissue. These results indicate that a big well-vascularized three-dimensional mature adipose tissue can be regenerated using elastic gel, polydopamine, platelets, and small fat tissue.
Collapse
Affiliation(s)
- Qiang Chang
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB, Canada
| | - Junrong Cai
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ying Wang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Ruijia Yang
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB, Canada
| | - Malcolm Xing
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB, Canada.,State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Feng Lu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
16
|
Yap KK, Yeoh GC, Morrison WA, Mitchell GM. The Vascularised Chamber as an In Vivo Bioreactor. Trends Biotechnol 2018; 36:1011-1024. [DOI: 10.1016/j.tibtech.2018.05.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 05/25/2018] [Accepted: 05/29/2018] [Indexed: 02/06/2023]
|
17
|
Ginsenoside Rg1 and platelet-rich fibrin enhance human breast adipose-derived stem cell function for soft tissue regeneration. Oncotarget 2018; 7:35390-403. [PMID: 27191987 PMCID: PMC5085237 DOI: 10.18632/oncotarget.9360] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 04/11/2016] [Indexed: 02/01/2023] Open
Abstract
Adipose-derived stem cells (ASCs) can be used to repair soft tissue defects, wounds, burns, and scars and to regenerate various damaged tissues. The cell differentiation capacity of ASCs is crucial for engineered adipose tissue regeneration in reconstructive and plastic surgery. We previously reported that ginsenoside Rg1 (G-Rg1 or Rg1) promotes proliferation and differentiation of ASCs in vitro and in vivio. Here we show that both G-Rg1 and platelet-rich fibrin (PRF) improve the proliferation, differentiation, and soft tissue regeneration capacity of human breast adipose-derived stem cells (HBASCs) on collagen type I sponge scaffolds in vitro and in vivo. Three months after transplantation, tissue wet weight, adipocyte number, intracellular lipid, microvessel density, and gene and protein expression of VEGF, HIF-1α, and PPARγ were higher in both G-Rg1- and PRF-treated HBASCs than in control grafts. More extensive new adipose tissue formation was evident after treatment with G-Rg1 or PRF. In summary, G-Rg1 and/or PRF co-administration improves the function of HBASCs for soft tissue regeneration engineering.
Collapse
|
18
|
Brett E, Chung N, Leavitt WT, Momeni A, Longaker MT, Wan DC. A Review of Cell-Based Strategies for Soft Tissue Reconstruction. TISSUE ENGINEERING PART B-REVIEWS 2017; 23:336-346. [PMID: 28372485 DOI: 10.1089/ten.teb.2016.0455] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Soft tissue reconstruction to restore volume to damaged or deficient tissue beneath the skin remains a challenging endeavor. Current techniques are centered around autologous fat transfer, or the use of synthetic substitutes, however, a great deal of scientific inquiry has been made into both the molecular mechanisms involved in, and limitations of, de novo adipogenesis, that is, the formation of new adipose tissue from precursor cells. To best comprehend these mechanisms, an understanding of defined markers for adipogenic differentiation, and knowledge of both commercially available and primary cell lines that enable in vitro and in vivo studies is necessary. We review the growth factors, proteins, cytokines, drugs, and molecular pathways that have shown promise in enhancing adipogenesis and vasculogenesis, in addition to the multitude of scaffolds that act as delivery vehicles to support these processes. While progress continues on these fronts, equally important is how researchers are optimizing clinically employed strategies such as autologous fat transfer through cell-based intervention, and the potential to augment this approach through isolation of preferentially adipogenic or angiogenic precursor subpopulations, which exists on the horizon. This review will highlight the novel molecular and synthetic modifications currently being studied for inducing adipose tissue regeneration on a cellular level, which will expand our arsenal of techniques for approaching soft tissue reconstruction.
Collapse
Affiliation(s)
- Elizabeth Brett
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Natalie Chung
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - William Tripp Leavitt
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Arash Momeni
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Michael T Longaker
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine , Stanford, California.,2 Institute for Stem Cell Biology and Regenerative Medicine, Stanford University , Stanford, California
| | - Derrick C Wan
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine , Stanford, California
| |
Collapse
|
19
|
The Combination of Tissue Dissection and External Volume Expansion Generates Large Volumes of Adipose Tissue. Plast Reconstr Surg 2017; 139:888e-899e. [DOI: 10.1097/prs.0000000000003212] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
20
|
Luo L, He Y, Chang Q, Xie G, Zhan W, Wang X, Zhou T, Xing M, Lu F. Polycaprolactone nanofibrous mesh reduces foreign body reaction and induces adipose flap expansion in tissue engineering chamber. Int J Nanomedicine 2016; 11:6471-6483. [PMID: 27980405 PMCID: PMC5147407 DOI: 10.2147/ijn.s114295] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Tissue engineering chamber technique can be used to generate engineered adipose tissue, showing the potential for the reconstruction of soft tissue defects. However, the consequent foreign body reaction induced by the exogenous chamber implantation causes thick capsule formation on the surface of the adipose flap following capsule contracture, which may limit the internal tissue expansion. The nanotopographical property and architecture of nanofibrous scaffold may serve as a promising method for minimizing the foreign body reaction. Accordingly, electrospinning porous polycaprolactone (PCL) nanofibrous mesh, a biocompatible synthetic polymer, was attached to the internal surface of the chamber for the reducing local foreign body reaction. Adipose flap volume, level of inflammation, collagen quantification, capsule thickness, and adipose tissue-specific gene expression in chamber after implantation were evaluated at different time points. The in vivo study revealed that the engineered adipose flaps in the PCL group had a structure similar to that in the controls and normal adipose tissue structure but with a larger flap volume. Interleukin (IL)-1β, IL-6, and transforming growth factor-β expression decreased significantly in the PCL group compared with the control. Moreover, the control group had much more collagen deposition and thicker capsule than that observed in the PCL group. These results indicate that the unique nanotopographical effect of electrospinning PCL nanofiber can reduce foreign body reaction in a tissue engineering chamber, which maybe a promising new method for generating a larger volume of mature, vascularized, and stable adipose tissue.
Collapse
Affiliation(s)
- Lin Luo
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Yunfan He
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
- Department of Mechanical Engineering, Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Qiang Chang
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
- Department of Mechanical Engineering, Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Gan Xie
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Weiqing Zhan
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Xuecen Wang
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Tao Zhou
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Malcolm Xing
- Department of Mechanical Engineering, Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
- Children’s Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Feng Lu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
21
|
Generating an Engineered Adipose Tissue Flap Using an External Suspension Device. Plast Reconstr Surg 2016; 138:109-120. [DOI: 10.1097/prs.0000000000002305] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
22
|
Zhan W, Marre D, Mitchell GM, Morrison WA, Lim SY. Tissue Engineering by Intrinsic Vascularization in an In Vivo Tissue Engineering Chamber. J Vis Exp 2016. [PMID: 27286267 DOI: 10.3791/54099] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
In reconstructive surgery, there is a clinical need for an alternative to the current methods of autologous reconstruction which are complex, costly and trade one defect for another. Tissue engineering holds the promise to address this increasing demand. However, most tissue engineering strategies fail to generate stable and functional tissue substitutes because of poor vascularization. This paper focuses on an in vivo tissue engineering chamber model of intrinsic vascularization where a perfused artery and a vein either as an arteriovenous loop or a flow-through pedicle configuration is directed inside a protected hollow chamber. In this chamber-based system angiogenic sprouting occurs from the arteriovenous vessels and this system attracts ischemic and inflammatory driven endogenous cell migration which gradually fills the chamber space with fibro-vascular tissue. Exogenous cell/matrix implantation at the time of chamber construction enhances cell survival and determines specificity of the engineered tissues which develop. Our studies have shown that this chamber model can successfully generate different tissues such as fat, cardiac muscle, liver and others. However, modifications and refinements are required to ensure target tissue formation is consistent and reproducible. This article describes a standardized protocol for the fabrication of two different vascularized tissue engineering chamber models in vivo.
Collapse
Affiliation(s)
- Weiqing Zhan
- O'Brien Institute Department, St Vincent's Institute of Medical Research
| | - Diego Marre
- O'Brien Institute Department, St Vincent's Institute of Medical Research
| | - Geraldine M Mitchell
- O'Brien Institute Department, St Vincent's Institute of Medical Research; Department of Surgery, University of Melbourne; Faculty of Health Sciences, Australia Catholic University
| | - Wayne A Morrison
- O'Brien Institute Department, St Vincent's Institute of Medical Research; Department of Surgery, University of Melbourne; Faculty of Health Sciences, Australia Catholic University
| | - Shiang Y Lim
- O'Brien Institute Department, St Vincent's Institute of Medical Research; Department of Surgery, University of Melbourne;
| |
Collapse
|
23
|
Adipose-Derived Stem Cell Delivery for Adipose Tissue Engineering: Current Status and Potential Applications in a Tissue Engineering Chamber Model. Stem Cell Rev Rep 2016; 12:484-91. [DOI: 10.1007/s12015-016-9653-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
24
|
Morrison WA, Marre D, Grinsell D, Batty A, Trost N, O'Connor AJ. Creation of a Large Adipose Tissue Construct in Humans Using a Tissue-engineering Chamber: A Step Forward in the Clinical Application of Soft Tissue Engineering. EBioMedicine 2016; 6:238-245. [PMID: 27211566 PMCID: PMC4856786 DOI: 10.1016/j.ebiom.2016.03.032] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 02/18/2016] [Accepted: 03/02/2016] [Indexed: 11/30/2022] Open
Abstract
Tissue engineering is currently exploring new and exciting avenues for the repair of soft tissue and organ defects. Adipose tissue engineering using the tissue engineering chamber (TEC) model has yielded promising results in animals; however, to date, there have been no reports on the use of this device in humans. Five female post mastectomy patients ranging from 35 to 49 years old were recruited and a pedicled thoracodorsal artery perforator fat flap ranging from 6 to 50 ml was harvested, transposed onto the chest wall and covered by an acrylic perforated dome-shaped chamber ranging from 140 to 350 cm3. Magnetic resonance evaluation was performed at three and six months after chamber implantation. Chambers were removed at six months and samples were obtained for histological analysis. In one patient, newly formed tissue to a volume of 210 ml was generated inside the chamber. One patient was unable to complete the trial and the other three failed to develop significant enlargement of the original fat flap, which, at the time of chamber explantation, was encased in a thick fibrous capsule. Our study provides evidence that generation of large well-vascularized tissue engineered constructs using the TEC is feasible in humans. Tissue engineering has the potential to offer exciting alternatives for the repair of soft tissues and organ development, The tissue-engineering chamber has been shown to support tissue growth and the generation of specialized organs in animals. Here we report on the use of this chamber in humans for the successful generation of new adipose tissue.
Tissue engineering has traditionally relied on the combination of cells and scaffolds, which are subsequently implanted into the patient. However, such paradigm is limited to tissues that are thin enough to rely on diffusion for survival or that have a low metabolic rate. The tissue-engineering chamber represents an interesting approach to circumvent these obstacles, as it is able to support the growth of well-vascularized blocks of specialized tissues of varying kinds. This work presents the use of such chamber in five human patients, one of which generated a large three-dimensional well-vascularized piece of adipose tissue, probably the largest and thickest engineered tissue construct reported to date.
Collapse
Affiliation(s)
- Wayne A Morrison
- O'Brien Institute of Regenerative Surgery, St. Vincent's Institute, Melbourne, Australia; Department of Plastic and Reconstructive Surgery, St. Vincent's Hospital Melbourne, Australia; Department of Surgery, St. Vincent's Hospital, University of Melbourne, Australia.
| | - Diego Marre
- O'Brien Institute of Regenerative Surgery, St. Vincent's Institute, Melbourne, Australia
| | - Damien Grinsell
- Department of Plastic and Reconstructive Surgery, St. Vincent's Hospital Melbourne, Australia; Department of Surgery, St. Vincent's Hospital, University of Melbourne, Australia
| | | | - Nicholas Trost
- Radiology Department, St. Vincent's Hospital Melbourne, Australia
| | - Andrea J O'Connor
- Department of Chemical and Biomolecular Engineering, Particulate Fluids Processing Centre, University of Melbourne, Australia
| |
Collapse
|
25
|
Volz AC, Huber B, Kluger PJ. Adipose-derived stem cell differentiation as a basic tool for vascularized adipose tissue engineering. Differentiation 2016; 92:52-64. [PMID: 26976717 DOI: 10.1016/j.diff.2016.02.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 01/08/2016] [Accepted: 02/10/2016] [Indexed: 12/13/2022]
Abstract
The development of in vitro adipose tissue constructs is highly desired to cope with the increased demand for substitutes to replace damaged soft tissue after high graded burns, deformities or tumor removal. To achieve clinically relevant dimensions, vascularization of soft tissue constructs becomes inevitable but still poses a challenge. Adipose-derived stem cells (ASCs) represent a promising cell source for the setup of vascularized fatty tissue constructs as they can be differentiated into adipocytes and endothelial cells in vitro and are thereby available in sufficiently high cell numbers. This review summarizes the currently known characteristics of ASCs and achievements in adipogenic and endothelial differentiation in vitro. Further, the interdependency of adipogenesis and angiogenesis based on the crosstalk of endothelial cells, stem cells and adipocytes is addressed at the molecular level. Finally, achievements and limitations of current co-culture conditions for the construction of vascularized adipose tissue are evaluated.
Collapse
Affiliation(s)
- Ann-Cathrin Volz
- Process Analysis and Technology (PA&T), Reutlingen University, Alteburgstraße 150, 72762 Reutlingen, Germany
| | - Birgit Huber
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Nobelstraße 12, 70569 Stuttgart, Germany
| | - Petra J Kluger
- Process Analysis and Technology (PA&T), Reutlingen University, Alteburgstraße 150, 72762 Reutlingen, Germany; Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Nobelstraße 12, 70569 Stuttgart, Germany
| |
Collapse
|
26
|
Development of Synthetic and Natural Materials for Tissue Engineering Applications Using Adipose Stem Cells. Stem Cells Int 2016; 2016:5786257. [PMID: 26977158 PMCID: PMC4764745 DOI: 10.1155/2016/5786257] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 01/09/2016] [Accepted: 01/12/2016] [Indexed: 12/12/2022] Open
Abstract
Adipose stem cells have prominent implications in tissue regeneration due to their abundance and relative ease of harvest from adipose tissue and their abilities to differentiate into mature cells of various tissue lineages and secrete various growth cytokines. Development of tissue engineering techniques in combination with various carrier scaffolds and adipose stem cells offers great potential in overcoming the existing limitations constraining classical approaches used in plastic and reconstructive surgery. However, as most tissue engineering techniques are new and highly experimental, there are still many practical challenges that must be overcome before laboratory research can lead to large-scale clinical applications. Tissue engineering is currently a growing field of medical research; in this review, we will discuss the progress in research on biomaterials and scaffolds for tissue engineering applications using adipose stem cells.
Collapse
|
27
|
NADPH oxidase 2 plays a role in experimental corneal neovascularization. Clin Sci (Lond) 2016; 130:683-96. [PMID: 26814205 DOI: 10.1042/cs20150103] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 01/26/2016] [Indexed: 12/14/2022]
Abstract
Corneal neovascularization, the growth of new blood vessels in the cornea, is a leading cause of vision impairment after corneal injury. Neovascularization typically occurs in response to corneal injury such as that caused by infection, physical trauma, chemical burns or in the setting of corneal transplant rejection. The NADPH oxidase enzyme complex is involved in cell signalling for wound-healing angiogenesis, but its role in corneal neovascularization has not been studied. We have now analysed the role of the Nox2 isoform of NADPH oxidase in corneal neovascularization in mice following chemical injury. C57BL/6 mice aged 8-14 weeks were cauterized with an applicator coated with 75% silver nitrate and 25% potassium nitrate for 8 s. Neovascularization extending radially from limbal vessels was observed in corneal whole-mounts from cauterized wild type mice and CD31+ vessels were identified in cauterized corneal sections at day 7. In contrast, in Nox2 knockout (Nox2 KO) mice vascular endothelial growth factor-A (Vegf-A), Flt1 mRNA expression, and the extent of corneal neovascularization were all markedly reduced compared with their wild type controls. The accumulation of Iba-1+ microglia and macrophages in the cornea was significantly less in Nox2 KO than in wild type mice. In conclusion, we have demonstrated that Nox2 is implicated in the inflammatory and neovascular response to corneal chemical injury in mice and clearly VEGF is a mediator of this effect. This work raises the possibility that therapies targeting Nox2 may have potential for suppressing corneal neovascularization and inflammation in humans.
Collapse
|
28
|
Salo T, Sutinen M, Hoque Apu E, Sundquist E, Cervigne NK, de Oliveira CE, Akram SU, Ohlmeier S, Suomi F, Eklund L, Juusela P, Åström P, Bitu CC, Santala M, Savolainen K, Korvala J, Paes Leme AF, Coletta RD. A novel human leiomyoma tissue derived matrix for cell culture studies. BMC Cancer 2015; 15:981. [PMID: 26673244 PMCID: PMC4682271 DOI: 10.1186/s12885-015-1944-z] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 11/19/2015] [Indexed: 01/29/2023] Open
Abstract
Background The composition of the matrix molecules is important in in vitro cell culture experiments of e.g. human cancer invasion and vessel formation. Currently, the mouse Engelbreth-Holm-Swarm (EHS) sarcoma -derived products, such as Matrigel®, are the most commonly used tumor microenvironment (TME) mimicking matrices for experimental studies. However, since Matrigel® is non-human in origin, its molecular composition does not accurately simulate human TME. We have previously described a solid 3D organotypic myoma disc invasion assay, which is derived from human uterus benign leiomyoma tumor. Here, we describe the preparation and analyses of a processed, gelatinous leiomyoma matrix, named Myogel. Methods A total protein extract, Myogel, was formulated from myoma. The protein contents of Myogel were characterized and its composition and properties compared with a commercial mouse Matrigel®. Myogel was tested and compared to Matrigel® in human cell adhesion, migration, invasion, colony formation, spheroid culture and vessel formation experiments, as well as in a 3D hanging drop video image analysis. Results We demonstrated that only 34 % of Myogel’s molecular content was similar to Matrigel®. All test results showed that Myogel was comparable with Matrigel®, and when mixed with low-melting agarose (Myogel-LMA) it was superior to Matrigel® in in vitro Transwell® invasion and capillary formation assays. Conclusions In conclusion, we have developed a novel Myogel TME matrix, which is recommended for in vitro human cell culture experiments since it closely mimics the human tumor microenvironment of solid cancers. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1944-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tuula Salo
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, PO Box 5281, FI-90014, Oulu, Finland. .,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, FI-90014, Oulu, Finland. .,Department of Oral and Maxillofacial Diseases, University of Helsinki, FI-00014, Helsinki, Finland.
| | - Meeri Sutinen
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, PO Box 5281, FI-90014, Oulu, Finland. .,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, FI-90014, Oulu, Finland.
| | - Ehsanul Hoque Apu
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, PO Box 5281, FI-90014, Oulu, Finland. .,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, FI-90014, Oulu, Finland.
| | - Elias Sundquist
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, PO Box 5281, FI-90014, Oulu, Finland. .,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, FI-90014, Oulu, Finland.
| | - Nilva K Cervigne
- Clinical Department, Faculty of Medicine of Jundiai (FMJ), Jundiai, São Paulo, SP-13202-550, Brazil. .,Department of Oral Diagnosis, Oral Pathology Division, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, SP-13414-903, Brazil.
| | - Carine Ervolino de Oliveira
- Department of Oral Diagnosis, Oral Pathology Division, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, SP-13414-903, Brazil.
| | - Saad Ullah Akram
- Center for Machine Vision Research, University of Oulu, FI-90014, Oulu, Finland. .,Biocenter Oulu, University of Oulu, FI-90014, Oulu, Finland.
| | - Steffen Ohlmeier
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014, Oulu, Finland. .,Proteomics Core Facility, Biocenter Oulu, University of Oulu, FI-90014, Oulu, Finland.
| | - Fumi Suomi
- Biocenter Oulu, University of Oulu, FI-90014, Oulu, Finland. .,Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014, Oulu, Finland.
| | - Lauri Eklund
- Biocenter Oulu, University of Oulu, FI-90014, Oulu, Finland. .,Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014, Oulu, Finland.
| | - Pirjo Juusela
- Department of Oral and Maxillofacial Diseases, University of Helsinki, FI-00014, Helsinki, Finland.
| | - Pirjo Åström
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, PO Box 5281, FI-90014, Oulu, Finland. .,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, FI-90014, Oulu, Finland.
| | - Carolina Cavalcante Bitu
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, PO Box 5281, FI-90014, Oulu, Finland. .,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, FI-90014, Oulu, Finland.
| | - Markku Santala
- Department of Obstetrics and Gynecology, Oulu University Hospital and University of Oulu, FI-90029, Oulu, Finland.
| | - Kalle Savolainen
- Department of Obstetrics and Gynecology, Tampere University Hospital and University of Tampere, FI-33521, Tampere, Finland.
| | - Johanna Korvala
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, PO Box 5281, FI-90014, Oulu, Finland. .,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, FI-90014, Oulu, Finland.
| | | | - Ricardo D Coletta
- Department of Oral Diagnosis, Oral Pathology Division, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, SP-13414-903, Brazil.
| |
Collapse
|
29
|
Han TTY, Toutounji S, Amsden BG, Flynn LE. Adipose-derived stromal cells mediate in vivo adipogenesis, angiogenesis and inflammation in decellularized adipose tissue bioscaffolds. Biomaterials 2015; 72:125-37. [PMID: 26360790 DOI: 10.1016/j.biomaterials.2015.08.053] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 08/25/2015] [Accepted: 08/28/2015] [Indexed: 12/20/2022]
Abstract
Decellularized adipose tissue (DAT) has shown promise as an adipogenic bioscaffold for soft tissue augmentation and reconstruction. The objective of the current study was to investigate the effects of allogeneic adipose-derived stem/stromal cells (ASCs) on in vivo fat regeneration in DAT bioscaffolds using an immunocompetent rat model. ASC seeding significantly enhanced angiogenesis and adipogenesis, with cell tracking studies indicating that the newly-forming tissues were host-derived. Incorporating ASCs also mediated the inflammatory response and promoted a more constructive macrophage phenotype. A fraction of the CD163(+) macrophages in the implants expressed adipogenic markers, with higher levels of this "adipocyte-like" phenotype in proximity to the developing adipose tissues. Our results indicate that the combination of ASCs and adipose extracellular matrix (ECM) provides an inductive microenvironment for adipose regeneration mediated by infiltrating host cell populations. The DAT scaffolds are a useful tissue-specific model system for investigating the mechanisms of in vivo adipogenesis that may help to develop a better understanding of this complex process in the context of both regeneration and disease. Overall, combining adipose-derived matrices with ASCs is a highly promising approach for the in situ regeneration of host-derived adipose tissue.
Collapse
Affiliation(s)
- Tim Tian Y Han
- Department of Chemical Engineering, Queen's University, 19 Division Street, Kingston, Ontario, Canada, K7L 3N6; Human Mobility Research Centre, Kingston General Hospital, 76 Stuart Street, Kingston, Ontario, Canada, K7L 2V7
| | - Sandra Toutounji
- Department of Chemical Engineering, Queen's University, 19 Division Street, Kingston, Ontario, Canada, K7L 3N6
| | - Brian G Amsden
- Department of Chemical Engineering, Queen's University, 19 Division Street, Kingston, Ontario, Canada, K7L 3N6; Human Mobility Research Centre, Kingston General Hospital, 76 Stuart Street, Kingston, Ontario, Canada, K7L 2V7
| | - Lauren E Flynn
- Department of Chemical Engineering, Queen's University, 19 Division Street, Kingston, Ontario, Canada, K7L 3N6; Human Mobility Research Centre, Kingston General Hospital, 76 Stuart Street, Kingston, Ontario, Canada, K7L 2V7; Department of Chemical and Biochemical Engineering, Thompson Engineering Building, The University of Western Ontario, London, Ontario, Canada, N6A 5B9; Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1.
| |
Collapse
|
30
|
Zhan W, Chang Q, Xiao X, Dong Z, Zeng Z, Gao J, Lu F. Self-synthesized extracellular matrix contributes to mature adipose tissue regeneration in a tissue engineering chamber. Wound Repair Regen 2015; 23:443-52. [PMID: 25847278 DOI: 10.1111/wrr.12292] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 03/26/2015] [Indexed: 12/01/2022]
Affiliation(s)
- Weiqing Zhan
- Department of Plastic and Cosmetic Surgery; Nanfang Hospital, Southern Medical University; Guangzhou Guangdong People's Republic of China
| | - Qiang Chang
- Department of Plastic and Cosmetic Surgery; Nanfang Hospital, Southern Medical University; Guangzhou Guangdong People's Republic of China
| | - Xiaolian Xiao
- Department of Plastic and Cosmetic Surgery; Nanfang Hospital, Southern Medical University; Guangzhou Guangdong People's Republic of China
| | - Ziqing Dong
- Department of Plastic and Cosmetic Surgery; Nanfang Hospital, Southern Medical University; Guangzhou Guangdong People's Republic of China
| | - Zhaowei Zeng
- Department of Plastic and Cosmetic Surgery; Nanfang Hospital, Southern Medical University; Guangzhou Guangdong People's Republic of China
| | - Jianhua Gao
- Department of Plastic and Cosmetic Surgery; Nanfang Hospital, Southern Medical University; Guangzhou Guangdong People's Republic of China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery; Nanfang Hospital, Southern Medical University; Guangzhou Guangdong People's Republic of China
| |
Collapse
|