1
|
Puthiya Veettil J, Sasikumar Lolitha D, Payanam Ramachandra U. Combinatorial Decellularization as a Better Approach to Porcine Liver Extracellular Matrix Scaffold Fabrication With Preserved Bioactivity: A Comparative Evaluation. Xenotransplantation 2025; 32:e70025. [PMID: 39960357 DOI: 10.1111/xen.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Soft tissue repair patches of decellularized extracellular matrices (ECM) with inherently preserved structural components and biomacromolecules are desirable in regenerative applications. This study characterizes three detergent-based decellularization methods to fabricate acellular porcine liver matrices to remove antigenic determinants without compromising the structural integrity, glycosaminoglycans (GAG) content, and bound growth factors within the resulting ECM. Three detergents chosen for decellularization were sodium dodecyl sulfate (SDS), SDS with sodium deoxycholate (SDS + SDC-combinatorial method), and Triton X-100 followed by SDS. Combinatorial detergent decellularization effectively removed cellular components and retained intact collagenous structure with minimal residual DNA and protein. It also preserved significantly higher amounts of GAG, HGF, and bFGF. TX100 decellularization was highly destructive with the least preservation of GAG and GFs. The SDS method showed an intermediate level of preservation of biomolecules. The correlation obtained between GAG and GFs revealed quantification of GAG to be an indirect way of estimating the bound GFs preserved within the ECM. In vitro experiments revealed the noncytotoxic nature of the scaffolds. The study revealed that, among the three methods of decellularization, ECM scaffold fabricated by combinatorial detergent decellularization is extremely promising for use as a soft tissue repair patch with inherent bioactive molecules for scaffold-based regenerative therapy.
Collapse
Affiliation(s)
- Jesna Puthiya Veettil
- Division of In-Vivo Models and Testing, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Devika Sasikumar Lolitha
- Division of In-Vivo Models and Testing, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Umashankar Payanam Ramachandra
- Division of In-Vivo Models and Testing, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| |
Collapse
|
2
|
Bhatt S S, Krishna Kumar J, Laya S, Thakur G, Nune M. Scaffold-mediated liver regeneration: A comprehensive exploration of current advances. J Tissue Eng 2024; 15:20417314241286092. [PMID: 39411269 PMCID: PMC11475092 DOI: 10.1177/20417314241286092] [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: 06/28/2024] [Accepted: 09/08/2024] [Indexed: 10/19/2024] Open
Abstract
The liver coordinates over 500 biochemical processes crucial for maintaining homeostasis, detoxification, and metabolism. Its specialized cells, arranged in hexagonal lobules, enable it to function as a highly efficient metabolic engine. However, diseases such as cirrhosis, fatty liver disease, and hepatitis present significant global health challenges. Traditional drug development is expensive and often ineffective at predicting human responses, driving interest in advanced in vitro liver models utilizing 3D bioprinting and microfluidics. These models strive to mimic the liver's complex microenvironment, improving drug screening and disease research. Despite its resilience, the liver is vulnerable to chronic illnesses, injuries, and cancers, leading to millions of deaths annually. Organ shortages hinder liver transplantation, highlighting the need for alternative treatments. Tissue engineering, employing polymer-based scaffolds and 3D bioprinting, shows promise. This review examines these innovative strategies, including liver organoids and liver tissue-on-chip technologies, to address the challenges of liver diseases.
Collapse
Affiliation(s)
- Supriya Bhatt S
- Manipal Institute of Regenerative Medicine, Bengaluru, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Jayanthi Krishna Kumar
- Manipal Institute of Regenerative Medicine, Bengaluru, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Shurthi Laya
- Manipal Institute of Regenerative Medicine, Bengaluru, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
- Department of Biomedical Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Goutam Thakur
- Department of Biomedical Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Manasa Nune
- Manipal Institute of Regenerative Medicine, Bengaluru, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
3
|
Allu I, Sahi AK, Koppadi M, Gundu S, Sionkowska A. Decellularization Techniques for Tissue Engineering: Towards Replicating Native Extracellular Matrix Architecture in Liver Regeneration. J Funct Biomater 2023; 14:518. [PMID: 37888183 PMCID: PMC10607724 DOI: 10.3390/jfb14100518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
The process of tissue regeneration requires the utilization of a scaffold, which serves as a structural framework facilitating cellular adhesion, proliferation, and migration within a physical environment. The primary aim of scaffolds in tissue engineering is to mimic the structural and functional properties of the extracellular matrix (ECM) in the target tissue. The construction of scaffolds that accurately mimic the architecture of the extracellular matrix (ECM) is a challenging task, primarily due to the intricate structural nature and complex composition of the ECM. The technique of decellularization has gained significant attention in the field of tissue regeneration because of its ability to produce natural scaffolds by removing cellular and genetic components from the extracellular matrix (ECM) while preserving its structural integrity. The present study aims to investigate the various decellularization techniques employed for the purpose of isolating the extracellular matrix (ECM) from its native tissue. Additionally, a comprehensive comparison of these methods will be presented, highlighting their respective advantages and disadvantages. The primary objective of this study is to gain a comprehensive understanding of the anatomical and functional features of the native liver, as well as the prevalence and impact of liver diseases. Additionally, this study aims to identify the limitations and difficulties associated with existing therapeutic methods for liver diseases. Furthermore, the study explores the potential of tissue engineering techniques in addressing these challenges and enhancing liver performance. By investigating these aspects, this research field aims to contribute to the advancement of liver disease treatment and management.
Collapse
Affiliation(s)
- Ishita Allu
- Department of Biomedical Engineering, University College of Engineering (UCE), Osmania University, Hyderabad 500007, India; (I.A.); (M.K.)
| | - Ajay Kumar Sahi
- School of Medicine, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA;
| | - Meghana Koppadi
- Department of Biomedical Engineering, University College of Engineering (UCE), Osmania University, Hyderabad 500007, India; (I.A.); (M.K.)
| | - Shravanya Gundu
- Department of Biomedical Engineering, University College of Engineering (UCE), Osmania University, Hyderabad 500007, India; (I.A.); (M.K.)
| | - Alina Sionkowska
- Faculty of Chemistry, Nicolaus Copernicus University in Torun, Jurija Gagarina 11, 87-100 Torun, Poland
- Faculty of Health Sciences, Calisia University, Nowy Świat 4, 62-800 Kalisz, Poland
| |
Collapse
|
4
|
Afzal Z, Huguet EL. Bioengineering liver tissue by repopulation of decellularised scaffolds. World J Hepatol 2023; 15:151-179. [PMID: 36926238 PMCID: PMC10011915 DOI: 10.4254/wjh.v15.i2.151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/22/2022] [Accepted: 02/15/2023] [Indexed: 02/24/2023] Open
Abstract
Liver transplantation is the only curative therapy for end stage liver disease, but is limited by the organ shortage, and is associated with the adverse consequences of immunosuppression. Repopulation of decellularised whole organ scaffolds with appropriate cells of recipient origin offers a theoretically attractive solution, allowing reliable and timely organ sourcing without the need for immunosuppression. Decellularisation methodologies vary widely but seek to address the conflicting objectives of removing the cellular component of tissues whilst keeping the 3D structure of the extra-cellular matrix intact, as well as retaining the instructive cell fate determining biochemicals contained therein. Liver scaffold recellularisation has progressed from small rodent in vitro studies to large animal in vivo perfusion models, using a wide range of cell types including primary cells, cell lines, foetal stem cells, and induced pluripotent stem cells. Within these models, a limited but measurable degree of physiologically significant hepatocyte function has been reported with demonstrable ammonia metabolism in vivo. Biliary repopulation and function have been restricted by challenges relating to the culture and propagations of cholangiocytes, though advances in organoid culture may help address this. Hepatic vasculature repopulation has enabled sustainable blood perfusion in vivo, but with cell types that would limit clinical applications, and which have not been shown to have the specific functions of liver sinusoidal endothelial cells. Minority cell groups such as Kupffer cells and stellate cells have not been repopulated. Bioengineering by repopulation of decellularised scaffolds has significantly progressed, but there remain significant experimental challenges to be addressed before therapeutic applications may be envisaged.
Collapse
Affiliation(s)
- Zeeshan Afzal
- Department of Surgery, Addenbrookes Hospital, NIHR Comprehensive Biomedical Research and Academic Health Sciences Centre; Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| | - Emmanuel Laurent Huguet
- Department of Surgery, Addenbrookes Hospital, NIHR Comprehensive Biomedical Research and Academic Health Sciences Centre; Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
5
|
Jambar Nooshin B, Tayebi T, Babajani A, Khani MM, Niknejad H. Effects of Different Perfusing Routes through The Portal Vein, Hepatic Vein, and Biliary Duct on Whole Rat Liver Decellularization. CELL JOURNAL 2023; 25:35-44. [PMID: 36680482 PMCID: PMC9868438 DOI: 10.22074/cellj.2022.557600.1081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/11/2022] [Accepted: 10/04/2022] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Organ transplantation is the last therapeutic choice for end-stage liver failure, which is limited by the lack of sufficient donors. Decellularized liver can be used as a suitable matrix for liver tissue engineering with clinical application potential. Optimizing the decellularization procedure would obtain a biological matrix with completely removed cellular components and preserved 3-dimensional structure. This study aimed to evaluate the decellularization efficacy through three anatomical routes. MATERIALS AND METHODS In this experimental study, rat liver decellularization was performed through biliary duct (BD), portal vein (PV), and hepatic vein (HV); using chemical detergents and enzymes. The decellularization efficacy was evaluated by measurement of DNA content, extracellular matrix (ECM) total proteins, and glycosaminoglycans (GAGs). ECM preservation was examined by histological and immunohistochemical (IHC) staining and scanning electron microscopy (SEM). Scaffold biocompatibility was tested by the MTT assay for HepG2 and HUVEC cell lines. RESULTS Decellularization through HV and PV resulted in a transparent scaffold by complete cell removal, while the BD route produced an opaque scaffold with incomplete decellularization. H and E staining confirmed these results. Maximum DNA loss was obtained using 1% and 0.5% sodium dodecyl sulfate (SDS) in the PV and HV groups and the DNA content decreased faster in the HV group. At the final stages, the proteins excreted in the HV and PV groups were significantly less than the BD group. The GAGs level was diminished after decellularization, especially in the PV and HV groups. In the HV and PV groups the collagen amount was significantly more than the BD group. The IHC and SEM images showed that the ECM structure was preserved and cellular components were entirely removed. MTT assay showed the biocompatibility of the decellularized scaffold. CONCLUSION The results revealed that the HV is a more suitable route for liver decellularization than the PV and BD.
Collapse
Affiliation(s)
- Bahram Jambar Nooshin
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tahereh Tayebi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhesam Babajani
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Mehdi Khani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Xu J, Yang S, Su Y, Hu X, Xi Y, Cheng YY, Kang Y, Nie Y, Pan B, Song K. A 3D bioprinted tumor model fabricated with gelatin/sodium alginate/decellularized extracellular matrix bioink. Int J Bioprint 2022; 9:630. [PMID: 36844237 PMCID: PMC9947382 DOI: 10.18063/ijb.v9i1.630] [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: 05/29/2022] [Accepted: 09/02/2022] [Indexed: 11/05/2022] Open
Abstract
109Tissue-engineered scaffolds are more commonly used to construct three-dimensional (3D) tumor models for in vitro studies when compared to the conventional two-dimensional (2D) cell culture because the microenvironments provided by the 3D tumor models closely resemble the in vivo system and could achieve higher success rate when the scaffolds are translated for use in pre-clinical animal model. Physical properties, heterogeneity, and cell behaviors of the model could be regulated to simulate different tumors by changing the components and concentrations of materials. In this study, a novel 3D breast tumor model was fabricated by bioprinting using a bioink that consists of porcine liver-derived decellularized extracellular matrix (dECM) with different concentrations of gelatin and sodium alginate. Primary cells were removed while extracellular matrix components of porcine liver were preserved. The rheological properties of biomimetic bioinks and the physical properties of hybrid scaffolds were investigated, and we found that the addition of gelatin increased hydrophilia and viscoelasticity, while the addition of alginate increased mechanical properties and porosity. The swelling ratio, compression modulus, and porosity could reach 835.43 ± 130.61%, 9.64 ± 0.41 kPa, and 76.62 ± 4.43%, respectively. L929 cells and the mouse breast tumor cells 4T1 were subsequently inoculated to evaluate biocompatibility of the scaffolds and to form the 3D models. The results showed that all scaffolds exhibited good biocompatibility, and the average diameter of tumor spheres could reach 148.52 ± 8.02 μm on 7 d. These findings suggest that the 3D breast tumor model could serve as an effective platform for anticancer drug screening and cancer research in vitro.
Collapse
Affiliation(s)
- Jie Xu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Shuangjia Yang
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Ya Su
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Xueyan Hu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yue Xi
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yuen Yee Cheng
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, NSW 2007, Australia
| | - Yue Kang
- Department of Breast Surgery, Cancer Hospital of China Medical University, 44 Xiaoheyan Road, Dadong District, Shenyang 110042, China,Corresponding authors: Kedong Song () Yue Kang () Yi Nie (); Bo Pan ()
| | - Yi Nie
- Zhengzhou Institute of Emerging Industrial Technology, Zhengzhou 450000, China,Key Laboratory of Green Process and Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China,Corresponding authors: Kedong Song () Yue Kang () Yi Nie (); Bo Pan ()
| | - Bo Pan
- Department of Breast Surgery, The Second Hospital of Dalian Medical University, 467 Zhongshan Road, Shahekou District, Dalian, Liaoning 116023, China,Corresponding authors: Kedong Song () Yue Kang () Yi Nie (); Bo Pan ()
| | - Kedong Song
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China,Corresponding authors: Kedong Song () Yue Kang () Yi Nie (); Bo Pan ()
| |
Collapse
|
7
|
Nicholls DL, Rostami S, Karoubi G, Haykal S. Perfusion decellularization for vascularized composite allotransplantation. SAGE Open Med 2022; 10:20503121221123893. [PMID: 36120388 PMCID: PMC9478687 DOI: 10.1177/20503121221123893] [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: 01/20/2021] [Accepted: 08/12/2022] [Indexed: 11/01/2022] Open
Abstract
Vascularized composite allotransplantation is becoming the emerging standard for reconstructive surgery treatment for patients with limb trauma and facial injuries involving soft tissue loss. Due to the complex immunogenicity of composite grafts, patients who undergo vascularized composite allotransplantation are reliant on lifelong immunosuppressive therapy. Decellularization of donor grafts to create an extracellular matrix bio-scaffold provides an immunomodulatory graft that preserves the structural and bioactive function of the extracellular matrix. Retention of extracellular matrix proteins, growth factors, and signaling cascades allow for cell adhesion, migration, proliferation, and tissue regeneration. Perfusion decellularization of detergents through the graft vasculature allows for increased regent access to all tissue layers, and removal of cellular debris through the venous system. Grafts can subsequently be repopulated with appropriate cells through the vasculature to facilitate tissue regeneration. The present work reviews methods of decellularization, process parameters, evaluation of adequate cellular and nuclear removal, successful applications of perfusion decellularization for use in vascularized composite allotransplantation, and current limitations.
Collapse
Affiliation(s)
| | - Sara Rostami
- Latner Thoracic Surgery Laboratories, Toronto General Hospital Research Institute, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Golnaz Karoubi
- Latner Thoracic Surgery Laboratories, Toronto General Hospital Research Institute, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON, Canada.,Departments of Mechanical and Industrial Engineering and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Siba Haykal
- Latner Thoracic Surgery Laboratories, Toronto General Hospital Research Institute, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON, Canada.,Division of Plastic & Reconstructive Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
8
|
Rahmati S, Jalili A, Banitalebi Dehkordi M, Przedborski M. An Effective Method for Decellularization of Human Foreskin: Implications for Skin Regeneration in Small Wounds. CELL JOURNAL 2022; 24:506-514. [PMID: 36274203 PMCID: PMC9588162 DOI: 10.22074/cellj.2022.8005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Acellular matrices of different allogeneic or xenogeneic origins are widely used as structural scaffolds in regenerative medicine. The main goal of this research was to optimize a method for decellularization of foreskin for skin regeneration in small wounds. MATERIALS AND METHODS In this experimental study, the dermal layers of foreskin were divided into two sections and subjected to two different decellularization methods: the sodium dodecyl sulfate method (SDS-M), and our optimized foreskin decellularization method (OFD-M). A combination of non-ionic detergents and SDS were used to decellularize the foreskin in OFD-M. The histological, morphological, and biomechanical properties of both methods were compared. In addition, human umbilical cord mesenchymal stem cells (hucMSCs) were isolated, and the biocompatibility and recellularization of both scaffolds by hucMSC were subsequently determined. RESULTS We observed that OFD-M is an appropriate approach for successful removal of cellular components from the foreskin tissue, without physical disturbance to the acellular matrix. In comparison to SDS-M, this new bioscaffold possesses a fine network containing a high amount of collagen fibers and glycosaminoglycans (GAG) (P≤0.03), is biocompatible and harmless for hucMSC (viability 91.7%), and exhibits a relatively high tensile strength. CONCLUSION We found that the extracellular matrix (ECM) structural integrity, the main ECM components, and the mechanical properties of the foreskin are well maintained after applying the OFD-M decellularization technique, indicating that the resulting scaffold would be a suitable platform for culturing MSC for skin grafting in small wounds.
Collapse
Affiliation(s)
- Shima Rahmati
- Cancer and Immunology Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences,
Sanandaj, Iran
| | - Ali Jalili
- Cancer and Immunology Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences,
Sanandaj, Iran,P.O.Box: 6618634683Cancer and Immunology Research CenterResearch Institute for Health DevelopmentKurdistan University of Medical SciencesSanandajIranP.O.Box: 8815713471Department of Molecular MedicineSchool of Advanced TechnologiesShahrekord University of Medical SciencesShahrekordIran
Emails:,
| | - Mehdi Banitalebi Dehkordi
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran,P.O.Box: 6618634683Cancer and Immunology Research CenterResearch Institute for Health DevelopmentKurdistan University of Medical SciencesSanandajIranP.O.Box: 8815713471Department of Molecular MedicineSchool of Advanced TechnologiesShahrekord University of Medical SciencesShahrekordIran
Emails:,
| | | |
Collapse
|
9
|
Gao M, Zhu X, Peng W, He Y, Li Y, Wu Q, Zhou Y, Liao G, Yang G, Bao J, Bu H. Kidney ECM Pregel Nanoarchitectonics for Microarrays to Accelerate Harvesting Gene-Edited Porcine Primary Monoclonal Spheres. ACS OMEGA 2022; 7:23156-23169. [PMID: 35847249 PMCID: PMC9280780 DOI: 10.1021/acsomega.2c01074] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
![]()
One of the key steps
of using CRISPR/Cas9 to obtain gene-edited
cells used in generating gene-edited animals combined with somatic
cell nuclear transplantation (SCNT) is to harvest monoclonal cells
with genetic modifications. However, primary cells used as nuclear
donors always grow slowly and fragile after a series of gene-editing
operations. The extracellular matrix (ECM) formulated directly from
different organs comprises complex proteins and growth factors that
can improve and regulate the cellular functions of primary cells.
Herein, sodium lauryl ether sulfate (SLES) detergent was first used
to perfuse porcine kidney ECM, and the biological properties of the
kidney ECM were optimized. Then, we used a porcine kidney ECM pregel
to pattern the microarray and developed a novel strategy to shorten
the time of obtaining gene-edited monoclonal cell spheroids with low
damage in batches. Our results showed that the SLES-perfused porcine
kidney ECM pregel displayed superior biological activities in releasing
growth factors and promoting cell proliferation. Finally, combined
with microarray technology, we quickly obtained monoclonal cells in
good condition, and the cells used as nuclear donors to construct
recombinant embryos showed a significantly higher success rate than
those of the traditional method. We further successfully produced
genetically edited pigs.
Collapse
Affiliation(s)
- Mengyu Gao
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu 610041, China
| | - Xinglong Zhu
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu 610041, China
| | - Wanliu Peng
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu 610041, China
| | - Yuting He
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu 610041, China
| | - Yi Li
- Precision Medicine Key Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiong Wu
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu 610041, China
| | - Yanyan Zhou
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu 610041, China
| | - Guangneng Liao
- Experimental Animal Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Guang Yang
- Experimental Animal Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ji Bao
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu 610041, China
| | - Hong Bu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu 610041, China
| |
Collapse
|
10
|
Design by Nature: Emerging Applications of Native Liver Extracellular Matrix for Cholangiocyte Organoid-Based Regenerative Medicine. Bioengineering (Basel) 2022; 9:bioengineering9030110. [PMID: 35324799 PMCID: PMC8945468 DOI: 10.3390/bioengineering9030110] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/25/2022] [Accepted: 03/04/2022] [Indexed: 12/14/2022] Open
Abstract
Organoid technology holds great promise for regenerative medicine. Recent studies show feasibility for bile duct tissue repair in humans by successfully transplanting cholangiocyte organoids in liver grafts during perfusion. Large-scale expansion of cholangiocytes is essential for extending these regenerative medicine applications. Human cholangiocyte organoids have a high and stable proliferation capacity, making them an attractive source of cholangiocytes. Commercially available basement membrane extract (BME) is used to expand the organoids. BME allows the cells to self-organize into 3D structures and stimulates cell proliferation. However, the use of BME is limiting the clinical applications of the organoids. There is a need for alternative tissue-specific and clinically relevant culture substrates capable of supporting organoid proliferation. Hydrogels prepared from decellularized and solubilized native livers are an attractive alternative for BME. These hydrogels can be used for the culture and expansion of cholangiocyte organoids in a clinically relevant manner. Moreover, the liver-derived hydrogels retain tissue-specific aspects of the extracellular microenvironment. They are composed of a complex mixture of bioactive and biodegradable extracellular matrix (ECM) components and can support the growth of various hepatobiliary cells. In this review, we provide an overview of the clinical potential of native liver ECM-based hydrogels for applications with human cholangiocyte organoids. We discuss the current limitations of BME for the clinical applications of organoids and how native ECM hydrogels can potentially overcome these problems in an effort to unlock the full regenerative clinical potential of the organoids.
Collapse
|
11
|
Morales-Guerrero NA, Varela-Echavarría A, Lozano Flores C, Vázquez-Cuevas FG, Velázquez-Miranda E, Reyes-López JV, García-Solís P, Solís-S JC, Hernández-Montiel HL. A new strategy for the decellularization of whole organs by hydrostatic pressure. Biotechnol Prog 2022; 38:e3248. [PMID: 35201677 DOI: 10.1002/btpr.3248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 11/06/2022]
Abstract
Tissue engineering has been able to develop novel decellularization-recellularization techniques, which facilitates the research for the generation of functional organs. This is based in the initial obtention of the organ's extracellular matrix (ECM). Therefore, any improvement in the decellularization process would have a positive impact in the results of the recellularization process. Nevertheless, commonly the methods and equipment employed for this process are expensive and thus limit the access of this technique to various research groups globally. AIM To develop a decellularization technique with the exclusive use of hydrostatic pressure of detergent solutions, to have an easily accessible and low-cost technique that meets the basic requirements of acellularity and functionality of the ECM. METHODS This experimental study was performed in 10 male Wistar rats, obtaining the liver to carry out serial washes, with 1, 2 and 3% Triton X-100 solutions and 0.1% SDS. The washes were performed by using a Gravity Perfusion System (GPS), which assured us a continuous hydrostatic pressure of 7.5 mmHg. The obtained ECM was processed using stains and immunostaining to determine the residual cell content and preservation of its components. RESULTS The staining showed a removal of cellular and nuclear components of approximately 97% of the acellular ECM, with an adequate three-dimensional pattern of collagen and proteoglycans. Furthermore, the acellular ECM allowed the viability of a primary hepatocyte culture. CONCLUSIONS The use of the GPS decellularization technique allowed us to obtain an acellular and functional ECM, drastically reducing experimentation costs. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Nelly A Morales-Guerrero
- Department of Biomedical Research, School of Medicine, Autonomous University of Queretaro, Qro., Mexico
| | | | - Carlos Lozano Flores
- Institute of Neurobiology, National Autonomous University of Mexico, Qro., Mexico
| | | | | | - Julián V Reyes-López
- Laboratory of Neurobiology and Cellular Bioengineering, Neurodiagnostic and Rehabilitation Unit "Dr. Moisés López González ", Faculty of Natural Sciences, Autonomous University of Querétaro
| | - Pablo García-Solís
- Department of Biomedical Research, School of Medicine, Autonomous University of Queretaro, Qro., Mexico
| | - Juan Carlos Solís-S
- Department of Biomedical Research, School of Medicine, Autonomous University of Queretaro, Qro., Mexico
| | - Hebert Luis Hernández-Montiel
- Laboratory of Neurobiology and Cellular Bioengineering, Neurodiagnostic and Rehabilitation Unit "Dr. Moisés López González ", Faculty of Natural Sciences, Autonomous University of Querétaro
| |
Collapse
|
12
|
Zhu X, Wu Q, He Y, Gao M, Li Y, Peng W, Li S, Liu Y, Zhang R, Bao J. Fabrication of Size-Controllable and Arrangement-Orderly HepG2 Spheroids for Drug Screening via Decellularized Liver Matrix-Derived Micropattern Array Chips. ACS OMEGA 2022; 7:2364-2376. [PMID: 35071924 PMCID: PMC8772313 DOI: 10.1021/acsomega.1c06302] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/20/2021] [Indexed: 02/08/2023]
Abstract
![]()
Three-dimensional
(3D) culture via micropattern arrays to generate
cellular spheroids seems a promising in vitro biomimetic
system for liver tissue engineering applications, such as drug screening.
Recently, organ-derived decellularized extracellular matrix emerges
as arguably the most biomimetic bioink. Herein, decellularized liver
matrix (DLM)-derived micropattern array chips were developed to fabricate
size-controllable and arrangement-orderly HepG2 spheroids for drug
screening. The porcine DLM was obtained by the removal of cellular
components and then ground into powder, followed by enzymolysis. DLM
as a coating substrate was compared with collagen type I (Col I) and
Matrigel in terms of biological performance for enhancing cell adhesion,
proliferation, and functions. Subsequently, we used poly(dimethylsiloxane)
(PDMS) to adsorb DLM as the bioink to fabricate micropattern array
chips. The optimal shape and size of micropattern were determined
by evaluating the morphology, viability, and functions of HepG2 3D
cellular aggregates. In addition, drug-susceptibility testing (paclitaxel,
doxorubicin HCl, and disulfiram) was performed on this novel platform.
The DLM provided the tissue-specific microenvironment that provided
suitable supports for HepG2 cells, compared to Col I and Matrigel.
A circular micropattern with a diameter of 100 μm was the optimal
processing parameter to rapidly fabricate large-scale, size-controllable,
and arrangement-orderly HepG2 cellular aggregates with 3D spheroid’s
shape and high cell viability. Drug screening testing showed that
the effect of a drug could be directly demonstrated on-chip by confocal
microscopy measuring the viability of spheroids. We provide a novel
platform for the large-scale generation of HepG2 spheroids with uniform
size and arrangement, thus bringing convenience, reducing error, and
increasing reproducibility for a rapid drug discovery by fluorescence
quantitative analysis. This methodology may be possible to apply in
advancing personalized medicine and drug discovery.
Collapse
Affiliation(s)
- Xinglong Zhu
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Qiong Wu
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China.,Laboratory of Liver Transplantation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yuting He
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Mengyu Gao
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China.,Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yi Li
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China.,Precision Medicine Key Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wanliu Peng
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Shengfu Li
- Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yong Liu
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Rundong Zhang
- West China School of Medicine, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Ji Bao
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
13
|
Ebrahim N, Badr OAM, Yousef MM, Hassouna A, Sabry D, Farid AS, Mostafa O, Saihati HAA, Seleem Y, Abd El Aziz E, Khalil AH, Nawar A, Shoulah AA, Aljasir M, Mohamed AZ, El-Sherbiny M, Elsherbiny NM, Eladl MA, Forsyth NR, Salim RF. Functional Recellularization of Acellular Rat Liver Scaffold by Induced Pluripotent Stem Cells: Molecular Evidence for Wnt/B-Catenin Upregulation. Cells 2021; 10:cells10112819. [PMID: 34831042 PMCID: PMC8616374 DOI: 10.3390/cells10112819] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Liver transplantation remains the only viable therapy for liver failure but has a severely restricted utility. Here, we aimed to decellularize rat livers to form acellular 3D bio-scaffolds suitable for seeding with induced pluripotent cells (iPSCs) as a tool to investigate the role of Wnt/β-catenin signaling in liver development and generation. METHODS Dissected rat livers were randomly divided into three groups: I (control); II (decellularized scaffolds) and III (recellularized scaffolds). Liver decellularization was established via an adapted perfusion procedure and assessed through the measurement of extracellular matrix (ECM) proteins and DNA content. Liver recellularization was assessed through histological examination and measurement of transcript levels of Wnt/β-catenin pathway, hepatogenesis, liver-specific microRNAs and growth factors essential for liver development. Adult rat liver decellularization was confirmed by the maintenance of ECM proteins and persistence of growth factors essential for liver regeneration. RESULTS iPSCs seeded rat decellularized livers displayed upregulated transcript expression of Wnt/β-catenin pathway-related, growth factors, and liver specification genes. Further, recellularized livers displayed restored liver-specific functions including albumin secretion and urea synthesis. CONCLUSION This establishes proof-of-principle for the generation of three-dimensional liver organ scaffolds as grafts and functional re-establishment.
Collapse
Affiliation(s)
- Nesrine Ebrahim
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Banha 13511, Egypt; (N.E.); (M.M.Y.); (O.M.)
- Stem Cell Unit, Faculty of Medicine, Benha University, Banha 13511, Egypt
| | - Omnia A. M. Badr
- Department of Genetics and Genetic Engineering, Faculty of Agriculture, Benha University, Banha 13511, Egypt;
| | - Mohamed M. Yousef
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Banha 13511, Egypt; (N.E.); (M.M.Y.); (O.M.)
| | - Amira Hassouna
- School of Public Health and Interdisciplinary Studies, Faculty of Health and Environmental Sciences, AUT University, Auckland 1010, New Zealand;
| | - Dina Sabry
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo 12613, Egypt;
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Bader University in Cairo, Cairo 11562, Egypt
| | - Ayman Samir Farid
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Benha University, Banha 13511, Egypt;
| | - Ola Mostafa
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Banha 13511, Egypt; (N.E.); (M.M.Y.); (O.M.)
| | - Hajir A. Al Saihati
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hafr Albatin, Hafar Al Batin 39524, Saudi Arabia;
| | - Yasmin Seleem
- Department of Clinical Pharmacology, Faculty of Medicine, Benha University, Banha 13511, Egypt; (Y.S.); (E.A.E.A.)
| | - Eman Abd El Aziz
- Department of Clinical Pharmacology, Faculty of Medicine, Benha University, Banha 13511, Egypt; (Y.S.); (E.A.E.A.)
| | - Ahmed Hassan Khalil
- Department of Surgery & Radiology, Faculty of Veterinary Medicine, Benha University, Banha 13511, Egypt;
| | - Ahmed Nawar
- Department of General Surgery, Faculty of Medicine, Benha University, Banha 13511, Egypt; (A.N.); (A.A.S.)
| | - Ahmed A. Shoulah
- Department of General Surgery, Faculty of Medicine, Benha University, Banha 13511, Egypt; (A.N.); (A.A.S.)
| | - Mohammad Aljasir
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 52571, Saudi Arabia;
| | - Amira Zaki Mohamed
- Department of Microbiology, Faculty of Science, Tanta University, Tanta 31527, Egypt;
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh 71666, Saudi Arabia;
- Department of Anatomy, Mansoura Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Nehal M. Elsherbiny
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk 47512, Saudi Arabia
- Correspondence: or (N.M.E.); (M.A.E.); (R.F.S.)
| | - Mohamed Ahmed Eladl
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Correspondence: or (N.M.E.); (M.A.E.); (R.F.S.)
| | - Nicholas Robert Forsyth
- Guy Hilton Research Laboratories, School of Pharmacy and Bioengineering, Faculty of Medicine and Health Sciences, Keele University, Newcastle ST5 5BG, UK;
| | - Rabab F. Salim
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Banha 13511, Egypt
- Correspondence: or (N.M.E.); (M.A.E.); (R.F.S.)
| |
Collapse
|
14
|
Felgendreff P, Schindler C, Mussbach F, Xie C, Gremse F, Settmacher U, Dahmen U. Identification of tissue sections from decellularized liver scaffolds for repopulation experiments. Heliyon 2021; 7:e06129. [PMID: 33644446 PMCID: PMC7895725 DOI: 10.1016/j.heliyon.2021.e06129] [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: 10/26/2020] [Revised: 01/07/2021] [Accepted: 01/26/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Biological organ engineering is a novel experimental approach to generate functional liver grafts by decellularization and repopulation. Currently, healthy organs of small or large animals and human organs with preexisting liver diseases are used to optimize decellularization and repopulation.However, the effects of morphological changes on allo- and xenogeneic cell-scaffold interactions during repopulation procedure, e.g., using scaffold-sections, are unknown. We present a sequential morphological workflow to identify murine liver scaffold-sections with well-preserved microarchitecture. METHODS Native livers (CONT, n = 9) and livers with experimentally induced pathologies (hepatics steatosis: STEA, n = 7; hepatic fibrosis induced by bile duct ligation: BDL, n = 9; nodular regenerative hyperplasia induced by 90% partial hepatectomy: PH, n = 8) were decellularized using SDS and Triton X-100 to generate cell-free scaffolds. Scaffold-sections were assessed using a sequential morphological workflow consisting of macroscopic, microscopic and morphological evaluation: (1) The scaffold was evaluated by a macroscopic decellularization score. (2) Regions without visible tissue remnants were localized for sampling and histological processing. Subsequent microscopical examination served to identify tissue samples without cell remnants. (3) Only cell-free tissue sections were subjected to detailed liver-specific morphological assessment using a histological and immunohistochemical decellularization score. RESULTS Decellularization was feasible in 33 livers, which were subjected to the sequential morphological workflow. In 11 of 33 scaffolds we achieved a good macroscopic decellularization result (CONT: 3 scaffolds; STEA: 3 scaffolds; BDL: 3 scaffolds; PH: 2 scaffolds). The microscopic assessment resulted in the selection of 88 cell-free tissue sections (CONT: 15 sections; STEA: 38 sections; BDL: 30 sections; PH: 5 sections). In 27 of those sections we obtained a good histological decellularization result (CONT: 3 sections; STEA: 6 sections; BDL: 17 sections; PH: 1 section). All experimental groups contained sections with a good immunohistochemical decellularization result (CONT: 6 sections; STEA: 5 sections; BDL: 4 sections; PH: 1 section). DISCUSSION Decellularization was possible in all experimental groups, irrespectively of the underlying morphological alteration. Furthermore, our proposed sequential morphological workflow was suitable to detect tissue sections with well-preserved hepatic microarchitecture, as needed for further repopulation experiments.
Collapse
Affiliation(s)
- Philipp Felgendreff
- Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany
- Research Program “Else Kröner-Forschungskolleg AntiAge”, Jena University Hospital, Jena, Germany
| | - Claudia Schindler
- Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany
| | - Franziska Mussbach
- Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany
| | - Chichi Xie
- Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany
| | - Felix Gremse
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany
| | - Utz Settmacher
- Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany
| | - Uta Dahmen
- Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany
| |
Collapse
|
15
|
Pennarossa G, Arcuri S, De Iorio T, Gandolfi F, Brevini TAL. Current Advances in 3D Tissue and Organ Reconstruction. Int J Mol Sci 2021; 22:E830. [PMID: 33467648 PMCID: PMC7830719 DOI: 10.3390/ijms22020830] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/31/2020] [Accepted: 01/13/2021] [Indexed: 12/11/2022] Open
Abstract
Bi-dimensional culture systems have represented the most used method to study cell biology outside the body for over a century. Although they convey useful information, such systems may lose tissue-specific architecture, biomechanical effectors, and biochemical cues deriving from the native extracellular matrix, with significant alterations in several cellular functions and processes. Notably, the introduction of three-dimensional (3D) platforms that are able to re-create in vitro the structures of the native tissue, have overcome some of these issues, since they better mimic the in vivo milieu and reduce the gap between the cell culture ambient and the tissue environment. 3D culture systems are currently used in a broad range of studies, from cancer and stem cell biology, to drug testing and discovery. Here, we describe the mechanisms used by cells to perceive and respond to biomechanical cues and the main signaling pathways involved. We provide an overall perspective of the most recent 3D technologies. Given the breadth of the subject, we concentrate on the use of hydrogels, bioreactors, 3D printing and bioprinting, nanofiber-based scaffolds, and preparation of a decellularized bio-matrix. In addition, we report the possibility to combine the use of 3D cultures with functionalized nanoparticles to obtain highly predictive in vitro models for use in the nanomedicine field.
Collapse
Affiliation(s)
- Georgia Pennarossa
- Laboratory of Biomedical Embryology, Department of Health, Animal Science and Food Safety and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; (G.P.); (S.A.); (T.D.I.)
| | - Sharon Arcuri
- Laboratory of Biomedical Embryology, Department of Health, Animal Science and Food Safety and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; (G.P.); (S.A.); (T.D.I.)
| | - Teresina De Iorio
- Laboratory of Biomedical Embryology, Department of Health, Animal Science and Food Safety and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; (G.P.); (S.A.); (T.D.I.)
| | - Fulvio Gandolfi
- Department of Agricultural and Environmental Sciences—Production, Landscape, Agroenergy and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 2, 20133 Milan, Italy;
| | - Tiziana A. L. Brevini
- Laboratory of Biomedical Embryology, Department of Health, Animal Science and Food Safety and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; (G.P.); (S.A.); (T.D.I.)
| |
Collapse
|
16
|
He YT, Zhu XL, Li SF, Zhang BQ, Li Y, Wu Q, Zhang YL, Zhou YY, Li L, Qi YN, Bao J, Bu H. Creating rat hepatocyte organoid as an in vitro model for drug testing. World J Stem Cells 2020; 12:1184-1195. [PMID: 33178400 PMCID: PMC7596445 DOI: 10.4252/wjsc.v12.i10.1184] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/15/2020] [Accepted: 08/01/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Liver organoids have recently been applied as models for liver disease and drug screening, especially when combined with liver-on-a-chip technologies. Compared to hepatocyte-like cells, primary hepatocytes have high functionality but cannot maintain their function when cultured in vitro. Mesenchymal stem cells (MSCs) enhance hepatocyte function and maintain hepatocyte metabolism when co-cultured with hepatocytes. MSCs can help induced pluripotent stem cells to generate an organoid structure via the MSC-based traction force triggered by extracellular matrix (ECM) proteins. In this study, primary hepatocytes were co-cultured with MSCs on a liver-derived ECM to generate liver organoids within a short duration.
AIM To create hepatocyte organoids by co-culturing primary hepatocytes with MSCs on a porcine liver extracellular matrix (PLECM) gel.
METHODS Perfusion and enzymatic hydrolysis were used to form the PLECM gel. Rat hepatocytes and human MSCs were mixed and plated on pre-solidified PLECM gel in a 48-well plate for 48 h to generate organoids. Generated organoids were evaluated through hematoxylin and eosin, periodic acid-Schiff, immuno-histological, and immunofluorescence staining, and quantitative PCR for alb, CYP450 gene markers, and urea cycle genes. Culture medium was collected to detect albumin (ALB) and urea production on days 2, 4, 6, 8, 14, and 20.
RESULTS The whole porcine liver was perfused and enzymatically hydrolyzed to form a PLECM gel. The structural components and basement membrane composition of the ECM, such as collagen type I, collagen type IV, fibronectin, and laminin, were demonstrated to be retained. Through interaction of human MSCs with the liver-derived ECM, primary hepatocytes and human MSCs assembled together into a 3D construction and generated primary hepatocyte organoids for 48 h. The mRNAs of the gene alb, the CYP450 gene markers cyp1a1, cyp1a2, and cyp3a2 as well as urea cycle genes arg-1, asl, ass-1, cps-1, nags were highly expressed in hepatocyte organoids. Long-term survival of the primary hepatocyte organoids, as well as stable functionality, was demonstrated via ALB and urea production in vitro.
CONCLUSION Our new method of creating primary hepatocyte organoids by co-culturing hepatocytes with MSCs on liver-derived ECM hydrogels could be used to develop models for liver disease and for drug screening.
Collapse
Affiliation(s)
- Yu-Ting He
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Xing-Long Zhu
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Sheng-Fu Li
- Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Bing-Qi Zhang
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yi Li
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Qiong Wu
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yun-Lin Zhang
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yan-Yan Zhou
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Li Li
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Ya-Na Qi
- Chinese Evidence-based Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Ji Bao
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Hong Bu
- Department of Pathology, West China Hospital, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
17
|
Alaby Pinheiro Faccioli L, Suhett Dias G, Hoff V, Lemos Dias M, Ferreira Pimentel C, Hochman-Mendez C, Braz Parente D, Labrunie E, Souza Mourão PA, Rogério de Oliveira Salvalaggio P, Goldberg AC, Campos de Carvalho AC, Dos Santos Goldenberg RC. Optimizing the Decellularized Porcine Liver Scaffold Protocol. Cells Tissues Organs 2020; 211:385-394. [PMID: 33040059 DOI: 10.1159/000510297] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/20/2020] [Indexed: 02/05/2023] Open
Abstract
There are few existing methods for shortening the decellularization period for a human-sized whole-liver scaffold. Here, we describe a protocol that enables effective decellularization of the liver obtained from pigs weigh 120 ± 4.2 kg within 72 h. Porcine livers (approx. 1.5 kg) were decellularized for 3 days using a combination of chemical and enzymatic decellularization agents. After trypsin, sodium deoxycholate, and Triton X-100 perfusion, the porcine livers were completely translucent. Our protocol was efficient to promote cell removal, the preservation of extracellular matrix (ECM) components, and vascular tree integrity. In conclusion, our protocol is efficient to promote human-sized whole-liver scaffold decellularization and thus useful to generate bioengineered livers to overcome the shortage of organs.
Collapse
Affiliation(s)
- Lanuza Alaby Pinheiro Faccioli
- Cellular and Molecular Cardiology Laboratory, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Radiology Department, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Grazielle Suhett Dias
- Cellular and Molecular Cardiology Laboratory, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Research and Education Institute, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Victor Hoff
- Cellular and Molecular Cardiology Laboratory, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marlon Lemos Dias
- Cellular and Molecular Cardiology Laboratory, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cibele Ferreira Pimentel
- Cellular and Molecular Cardiology Laboratory, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Daniella Braz Parente
- Radiology Department, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- D'Or Institute for Research and Education, Botafogo, Rio de Janeiro, Brazil
| | - Ester Labrunie
- Radiology Department, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paulo Antonio Souza Mourão
- Connective Tissue Laboratory, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Anna Carla Goldberg
- Research and Education Institute, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Antonio Carlos Campos de Carvalho
- Cellular and Molecular Cardiology Laboratory, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Institute of Science and Technology for Regenerative Medicine - REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Center for Structural Biology and Bioimaging - CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Regina Coeli Dos Santos Goldenberg
- Cellular and Molecular Cardiology Laboratory, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil,
- Institute of Science and Technology for Regenerative Medicine - REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil,
| |
Collapse
|
18
|
Contessi Negrini N, Toffoletto N, Farè S, Altomare L. Plant Tissues as 3D Natural Scaffolds for Adipose, Bone and Tendon Tissue Regeneration. Front Bioeng Biotechnol 2020; 8:723. [PMID: 32714912 PMCID: PMC7344190 DOI: 10.3389/fbioe.2020.00723] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/09/2020] [Indexed: 01/06/2023] Open
Abstract
Decellularized tissues are a valid alternative as tissue engineering scaffolds, thanks to the three-dimensional structure that mimics native tissues to be regenerated and the biomimetic microenvironment for cells and tissues growth. Despite decellularized animal tissues have long been used, plant tissue decellularized scaffolds might overcome availability issues, high costs and ethical concerns related to the use of animal sources. The wide range of features covered by different plants offers a unique opportunity for the development of tissue-specific scaffolds, depending on the morphological, physical and mechanical peculiarities of each plant. Herein, three different plant tissues (i.e., apple, carrot, and celery) were decellularized and, according to their peculiar properties (i.e., porosity, mechanical properties), addressed to regeneration of adipose tissue, bone tissue and tendons, respectively. Decellularized apple, carrot and celery maintained their porous structure, with pores ranging from 70 to 420 μm, depending on the plant source, and were stable in PBS at 37°C up to 7 weeks. Different mechanical properties (i.e., Eapple = 4 kPa, Ecarrot = 43 kPa, Ecelery = 590 kPa) were measured and no indirect cytotoxic effects were demonstrated in vitro after plants decellularization. After coating with poly-L-lysine, apples supported 3T3-L1 preadipocytes adhesion, proliferation and adipogenic differentiation; carrots supported MC3T3-E1 pre-osteoblasts adhesion, proliferation and osteogenic differentiation; celery supported L929 cells adhesion, proliferation and guided anisotropic cells orientation. The versatile features of decellularized plant tissues and their potential for the regeneration of different tissues are proved in this work.
Collapse
Affiliation(s)
- Nicola Contessi Negrini
- Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Milan, Italy
- National Interuniversity Consortium of Materials Science and Technology, Local Unit Politecnico di Milano, Milan, Italy
| | - Nadia Toffoletto
- Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Milan, Italy
- National Interuniversity Consortium of Materials Science and Technology, Local Unit Politecnico di Milano, Milan, Italy
| | - Silvia Farè
- Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Milan, Italy
- National Interuniversity Consortium of Materials Science and Technology, Local Unit Politecnico di Milano, Milan, Italy
| | - Lina Altomare
- Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Milan, Italy
- National Interuniversity Consortium of Materials Science and Technology, Local Unit Politecnico di Milano, Milan, Italy
| |
Collapse
|
19
|
Verstegen MMA, Spee B, van der Laan LJW. Bioengineering Liver Transplantation. Bioengineering (Basel) 2019; 6:E96. [PMID: 31623066 PMCID: PMC6955917 DOI: 10.3390/bioengineering6040096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 10/14/2019] [Indexed: 11/17/2022] Open
Abstract
Since the first in-man liver transplantation was performed by Starzl et al [...].
Collapse
Affiliation(s)
- Monique M A Verstegen
- Department of Surgery, Erasmus MC-University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands.
| | - Bart Spee
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands.
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC-University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands.
| |
Collapse
|
20
|
Bizzaro D, Russo FP, Burra P. New Perspectives in Liver Transplantation: From Regeneration to Bioengineering. Bioengineering (Basel) 2019; 6:81. [PMID: 31514475 PMCID: PMC6783848 DOI: 10.3390/bioengineering6030081] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/09/2019] [Accepted: 09/10/2019] [Indexed: 12/18/2022] Open
Abstract
Advanced liver diseases have very high morbidity and mortality due to associated complications, and liver transplantation represents the only current therapeutic option. However, due to worldwide donor shortages, new alternative approaches are mandatory for such patients. Regenerative medicine could be the more appropriate answer to this need. Advances in knowledge of physiology of liver regeneration, stem cells, and 3D scaffolds for tissue engineering have accelerated the race towards efficient therapies for liver failure. In this review, we propose an update on liver regeneration, cell-based regenerative medicine and bioengineering alternatives to liver transplantation.
Collapse
Affiliation(s)
- Debora Bizzaro
- Department of Surgery, Oncology and Gastroenterology, Gastroenterology/Multivisceral Transplant Section, University/Hospital Padua, 35128 Padua, Italy.
| | - Francesco Paolo Russo
- Department of Surgery, Oncology and Gastroenterology, Gastroenterology/Multivisceral Transplant Section, University/Hospital Padua, 35128 Padua, Italy.
| | - Patrizia Burra
- Department of Surgery, Oncology and Gastroenterology, Gastroenterology/Multivisceral Transplant Section, University/Hospital Padua, 35128 Padua, Italy.
| |
Collapse
|
21
|
Advances in Hepatic Tissue Bioengineering with Decellularized Liver Bioscaffold. Stem Cells Int 2019; 2019:2693189. [PMID: 31198426 PMCID: PMC6526559 DOI: 10.1155/2019/2693189] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/08/2019] [Accepted: 03/17/2019] [Indexed: 12/28/2022] Open
Abstract
The burden of liver diseases continues to grow worldwide, and liver transplantation is the only option for patients with end-stage liver disease. This procedure is limited by critical issues, including the low availability of donor organs; thus, novel therapeutic strategies are greatly needed. Recently, bioengineering approaches using decellularized liver scaffolds have been proposed as a novel strategy to overcome these challenges. The aim of this systematic literature review was to identify the major advances in the field of bioengineering using decellularized liver scaffolds and to identify obstacles and challenges for clinical application. The main findings of the articles and each contribution for technique optimization were highlighted, including the protocols of perfusion and decellularization, duration, demonstration of quality control—scaffold acellularity, matrix composition, and preservation of growth factors—and tissue functionality after recellularization. In previous years, many advances have been made as this technique has evolved from studies in animal models to human livers. As the field develops and this promising technique has become much more feasible, many challenges remain, including the selection of appropriate cell types for recellularization, route of cell administration, cell-seeding protocol, and scalability that must be standardized prior to clinical application.
Collapse
|
22
|
Grant R, Hay D, Callanan A. From scaffold to structure: the synthetic production of cell derived extracellular matrix for liver tissue engineering. Biomed Phys Eng Express 2018. [DOI: 10.1088/2057-1976/aacbe1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
23
|
Wang A, Jank I, Wei W, Schindler C, Dahmen U. A Novel Surgical Technique As a Foundation for In Vivo Partial Liver Engineering in Rat. J Vis Exp 2018. [PMID: 30346385 DOI: 10.3791/57991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Organ engineering is a novel strategy to generate liver organ substitutes that can potentially be used in transplantation. Recently, in vivo liver engineering, including in vivo organ decellularization followed by repopulation, has emerged as a promising approach over ex vivo liver engineering. However, postoperative survival was not achieved. The aim of this study is to develop a novel surgical technique of in vivo selective liver lobe perfusion in rats as a prerequisite for in vivo liver engineering. We generate a circuit bypass only through the left lateral lobe. Then, the left lateral lobe is perfused with heparinized saline. The experiment is performed with 4 groups (n = 3 rats per group) based on different perfusion times of 20 min, 2 h, 3 h, and 4 h. Survival, as well as the macroscopically visible change of color and the histologically determined absence of blood cells in the portal triad and the sinusoids, is taken as an indicator for a successful model establishment. After selective perfusion of the left lateral lobe, we observe that the left lateral lobe, indeed, turned from red to faint yellow. In a histological assessment, no blood cells are visible in the branch of the portal vein, the central vein, and the sinusoids. The left lateral lobe turns red after reopening the blocked vessels. 12/12 rats survived the procedure for more than one week. We are the first to report a surgical model for in vivo single liver lobe perfusion with a long survival period of more than one week. In contrast to the previously published report, the most important advantage of the technique presented here is that perfusion of 70% of the liver is maintained throughout the whole procedure. The establishment of this technique provides a foundation for in vivo partial liver engineering in rats, including decellularization and recellularization.
Collapse
Affiliation(s)
- An Wang
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, University Hospital Jena
| | - Isabel Jank
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, University Hospital Jena
| | - Weiwei Wei
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, University Hospital Jena
| | - Claudia Schindler
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, University Hospital Jena
| | - Uta Dahmen
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, University Hospital Jena;
| |
Collapse
|
24
|
Hashemi J, Pasalar P, Soleimani M, Arefian E, Khorramirouz R, Akbarzadeh A, Ghorbani F, Enderami S, Kajbafzadeh A. Decellularized Pancreas Matrix Scaffolds for Tissue Engineering Using Ductal or Arterial Catheterization. Cells Tissues Organs 2018; 205:72-84. [DOI: 10.1159/000487230] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 01/29/2018] [Indexed: 12/21/2022] Open
Abstract
Introduction: Diabetes is known as a worldwide disease with a great burden on society. Since therapeutic options cover a limited number of target points, new therapeutic strategies in the field of regenerative medicine are considered. Bioscaffolds along with islet cells would provide bioengineered tissue as a substitute for β-cells. The perfusion-decellularization technique is considered to create such scaffolds since they mimic the compositional, architectural, and biomechanical nature of a native organ. In this study, we investigated 2 decellularization methods preserving tissue microarchitecture. Methods: Procured pancreas from Sprague-Dawley rats was exposed to different percentages of detergent for 2, 4, and 6 h after cannulation via the common bile duct or aorta. Results: High concentrations of sodium dodecyl sulfate (SDS), i.e., > 0.05%, resulted in tissue disruption or incomplete cell removal depending on the duration of exposure. In both methods, 6-h exposure to 0.05% SDS created a bioscaffold with intact extracellular matrices and proper biomechanical characteristics. Tissue-specific stainings revealed that elastic, reticular, and collagen fiber concentrations were well preserved. Quantitative findings showed that glycosaminoglycan content was slightly different, but hydroxyproline was in the range of native pancreas tissue. Dye infusion through ductal and vascular cannulation proved that the vascular network was intact, and scanning electron microscopy indicated a homogeneous porous structure. Conclusions: Using the detergent-based method, an effective and time-efficient procedure, a whole pancreas extracellular matrix bioscaffold can be developed that can be used as a 3D structure for pancreas tissue engineering-based studies and regenerative medicine applications.
Collapse
|
25
|
Ogoke O, Oluwole J, Parashurama N. Bioengineering considerations in liver regenerative medicine. J Biol Eng 2017; 11:46. [PMID: 29204185 PMCID: PMC5702480 DOI: 10.1186/s13036-017-0081-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 09/25/2017] [Indexed: 12/19/2022] Open
Abstract
Background Liver disease contributes significantly to global disease burden and is associated with rising incidence and escalating costs. It is likely that innovative approaches, arising from the emerging field of liver regenerative medicine, will counter these trends. Main body Liver regenerative medicine is a rapidly expanding field based on a rich history of basic investigations into the nature of liver structure, physiology, development, regeneration, and function. With a bioengineering perspective, we discuss all major subfields within liver regenerative medicine, focusing on the history, seminal publications, recent progress within these fields, and commercialization efforts. The areas reviewed include fundamental aspects of liver transplantation, liver regeneration, primary hepatocyte cell culture, bioartificial liver, hepatocyte transplantation and liver cell therapies, mouse liver repopulation, adult liver stem cell/progenitor cells, pluripotent stem cells, hepatic microdevices, and decellularized liver grafts. Conclusion These studies highlight the creative directions of liver regenerative medicine, the collective efforts of scientists, engineers, and doctors, and the bright outlook for a wide range of approaches and applications which will impact patients with liver disease.
Collapse
Affiliation(s)
- Ogechi Ogoke
- Department of Chemical and Biological Engineering, University at Buffalo (State University of New York), Furnas Hall, Buffalo, NY 14260 USA.,Clinical and Translation Research Center (CTRC), University at Buffalo (State University of New York), 875 Ellicott St., Buffalo, NY 14203 USA
| | - Janet Oluwole
- Clinical and Translation Research Center (CTRC), University at Buffalo (State University of New York), 875 Ellicott St., Buffalo, NY 14203 USA.,Department of Biomedical Engineering, University at Buffalo (State University of New York), Furnas Hall, 907 Furnas Hall, Buffalo, NY 14260 USA
| | - Natesh Parashurama
- Department of Chemical and Biological Engineering, University at Buffalo (State University of New York), Furnas Hall, Buffalo, NY 14260 USA.,Clinical and Translation Research Center (CTRC), University at Buffalo (State University of New York), 875 Ellicott St., Buffalo, NY 14203 USA.,Department of Biomedical Engineering, University at Buffalo (State University of New York), Furnas Hall, 907 Furnas Hall, Buffalo, NY 14260 USA
| |
Collapse
|
26
|
Sachs PC, Mollica PA, Bruno RD. Tissue specific microenvironments: a key tool for tissue engineering and regenerative medicine. J Biol Eng 2017; 11:34. [PMID: 29177006 PMCID: PMC5688702 DOI: 10.1186/s13036-017-0077-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 08/24/2017] [Indexed: 12/12/2022] Open
Abstract
The accumulated evidence points to the microenvironment as the primary mediator of cellular fate determination. Comprised of parenchymal cells, stromal cells, structural extracellular matrix proteins, and signaling molecules, the microenvironment is a complex and synergistic edifice that varies tissue to tissue. Furthermore, it has become increasingly clear that the microenvironment plays crucial roles in the establishment and progression of diseases such as cardiovascular disease, neurodegeneration, cancer, and ageing. Here we review the historical perspectives on the microenvironment, and how it has directed current explorations in tissue engineering. By thoroughly understanding the role of the microenvironment, we can begin to correctly manipulate it to prevent and cure diseases through regenerative medicine techniques.
Collapse
Affiliation(s)
- Patrick C Sachs
- Medical Diagnostic and Translational Sciences, College of Health Science, Old Dominion University, Norfolk, VA 23529 USA
| | - Peter A Mollica
- Medical Diagnostic and Translational Sciences, College of Health Science, Old Dominion University, Norfolk, VA 23529 USA
| | - Robert D Bruno
- Medical Diagnostic and Translational Sciences, College of Health Science, Old Dominion University, Norfolk, VA 23529 USA
| |
Collapse
|
27
|
Wu Q, Tang J, Li Y, Li L, Wang Y, Bao J, Bu H. Hepatic differentiation of mouse bone marrow‑derived mesenchymal stem cells using a novel 3D culture system. Mol Med Rep 2017; 16:9473-9479. [PMID: 29152658 PMCID: PMC5780005 DOI: 10.3892/mmr.2017.7818] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/16/2017] [Indexed: 02/05/2023] Open
Abstract
The development of novel culture systems that mimic the in vivo microenvironment may be beneficial for inducing the differentiation of stem cells and promoting liver function. In the present study, spheroid cultures and decellularized liver scaffolds (DLSs) were utilized to obtain differentiated hepatocyte-like cells. Mouse bone marrow (BM)-derived mesenchymal stem cells (MSCs) self-aggregated into spheroids under low-attachment conditions and implanted into the DLSs via a negative pressure suction device. The Albp-ZsGreen adenoviral vector was utilized for real-time monitoring of hepatocyte-like cell differentiation. To detect the differentiation stages of the MSCs, immunostaining of hepatocyte markers and functional analysis was performed. Compared with traditional 2D monolayer induction, mouse BM-MSCs spheroids and DLSs in 3D culture generated greater yields of mature, differentiated hepatocytes. In conclusion, this 3D culture system may provide a strategy for generating hepatocyte-like cells for portable liver micro-organs, and aid clinical hepatocyte transplantation and liver tissue engineering.
Collapse
Affiliation(s)
- Qiong Wu
- Laboratory of Pathology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jing Tang
- Laboratory of Pathology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yi Li
- Laboratory of Pathology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Li Li
- Laboratory of Pathology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yujia Wang
- Laboratory of Pathology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ji Bao
- Laboratory of Pathology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hong Bu
- Laboratory of Pathology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
28
|
Mandrycky C, Phong K, Zheng Y. Tissue engineering toward organ-specific regeneration and disease modeling. MRS COMMUNICATIONS 2017; 7:332-347. [PMID: 29750131 PMCID: PMC5939579 DOI: 10.1557/mrc.2017.58] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/17/2017] [Indexed: 05/17/2023]
Abstract
Tissue engineering has been recognized as a translational approach to replace damaged tissue or whole organs. Engineering tissue, however, faces an outstanding knowledge gap in the challenge to fully recapitulate complex organ-specific features. Major components, such as cells, matrix, and architecture, must each be carefully controlled to engineer tissue-specific structure and function that mimics what is found in vivo. Here we review different methods to engineer tissue, and discuss critical challenges in recapitulating the unique features and functional units in four major organs-the kidney, liver, heart, and lung, which are also the top four candidates for organ transplantation in the USA. We highlight advances in tissue engineering approaches to enable the regeneration of complex tissue and organ substitutes, and provide tissue-specific models for drug testing and disease modeling. We discuss the current challenges and future perspectives toward engineering human tissue models.
Collapse
Affiliation(s)
- Christian Mandrycky
- Departments of Bioengineering, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Kiet Phong
- Departments of Bioengineering, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Ying Zheng
- Departments of Bioengineering, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
29
|
Tang J, Wu Q, Li Y, Wu X, Wang Y, Zhu L, Shi Y, Bu H, Bao J, Xie M. Construction of a general albumin promoter reporter system for real-time monitoring of the differentiation status of functional hepatocytes from stem cells in mouse, rat and human. Biomed Rep 2017; 6:627-632. [PMID: 28584633 PMCID: PMC5449956 DOI: 10.3892/br.2017.905] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 04/06/2017] [Indexed: 02/05/2023] Open
Abstract
Genetic constructs with promoters fused to reporter genes for simultaneous monitoring of cellular events have been the focus of attention in recent years. Adenoviral vectors, which have distinctive characteristics, have been used to monitor the differentiation of stem cells in vitro. In the present study, a modified adenoviral vector was constructed, containing a mouse, rat, and human general albumin promoter sequence fused to a ZsGreen reporter gene, and evaluated its efficiency in different cell types. Two hepatocyte cell lines (Hepa1-6 and HepG2), rat primary hepatocytes, rat bone marrow mesenchymal stem cells (BM-MSCs) and rat BM-MSCs-derived hepatocyte-like cells were transduced with this vector, and the transfection efficiency and functional capabilities of the promoter were evaluated by fluorescent microscopy. The results demonstrated efficient expression of ZsGreen in Hepa1-6 cells, HepG2 cells, rat primary hepatocytes, and rat BM-MSCs-derived hepatocyte-like cells, but not in rat BM-MSCs. In conclusion, the current study demonstrates a simple, high-efficiency, general tool for real-time monitoring of the differentiation status of hepatocytes from stem cells in mice, rats, and humans. This tool may be useful for evaluating different protocols to generate functional hepatocytes from stem cells in multiple species.
Collapse
Affiliation(s)
- Jing Tang
- Department of Clinical Medicine, Sichuan Medical University, Luzhou, Sichuan 646000, P.R. China.,Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China.,Department of General Surgery, Yibin City First People's Hospital, Yibin, Sichuan 644000, P.R. China
| | - Qiong Wu
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yi Li
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiujuan Wu
- Department of General Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Yujia Wang
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Lihua Zhu
- Department of General Surgery, Yibin City First People's Hospital, Yibin, Sichuan 644000, P.R. China
| | - Yujun Shi
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hong Bu
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ji Bao
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Mingjun Xie
- Department of Clinical Medicine, Sichuan Medical University, Luzhou, Sichuan 646000, P.R. China.,Department of General Surgery, Yibin City First People's Hospital, Yibin, Sichuan 644000, P.R. China
| |
Collapse
|
30
|
Simões IN, Vale P, Soker S, Atala A, Keller D, Noiva R, Carvalho S, Peleteiro C, Cabral JMS, Eberli D, da Silva CL, Baptista PM. Acellular Urethra Bioscaffold: Decellularization of Whole Urethras for Tissue Engineering Applications. Sci Rep 2017; 7:41934. [PMID: 28165009 PMCID: PMC5292742 DOI: 10.1038/srep41934] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 01/03/2017] [Indexed: 11/16/2022] Open
Abstract
Patients with stress urinary incontinence mainly suffer from malfunction of the urethra closure mechanism. We established the decellularization of porcine urethras to produce acellular urethra bioscaffolds for future tissue engineering applications, using bioscaffolds or bioscaffold-derived soluble products. Cellular removal was evaluated by H&E, DAPI and DNA quantification. The presence of specific ECM proteins was assessed through immunofluorescence staining and colorimetric assay kits. Human skeletal muscle myoblasts, muscle progenitor cells and adipose-derived stromal vascular fractions were used to evaluate the recellularization of the acellular urethra bioscaffolds. The mechanochemical decellularization system removed ~93% of tissue's DNA, generally preserving ECM's components and microarchitecture. Recellularization was achieved, though methodological advances are required regarding cell seeding strategies and functional assessment. Through microdissection and partial digestion, different urethra ECM-derived coating substrates were formulated (i.e. containing smooth or skeletal muscle ECM) and used to culture MPCs in vitro. The skeletal muscle ECM substrates enhanced fiber formation leading to the expression of the main skeletal muscle-related proteins and genes, as confirmed by immunofluorescence and RT-qPCR. The described methodology produced a urethra bioscaffold that retained vital ECM proteins and was liable to cell repopulation, a crucial first step towards the generation of urethra bioscaffold-based Tissue Engineering products.
Collapse
Affiliation(s)
- Irina N. Simões
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Paulo Vale
- Serviço Urologia, Hospital Garcia de Orta, Almada, Portugal
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | - Daniel Keller
- Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Rute Noiva
- Faculdade de Medicina Veterinária, The Interdisciplinary Centre of Research in Animal Health (CIISA), Universidade de Lisboa, Lisboa, Portugal
| | - Sandra Carvalho
- Faculdade de Medicina Veterinária, The Interdisciplinary Centre of Research in Animal Health (CIISA), Universidade de Lisboa, Lisboa, Portugal
| | - Conceição Peleteiro
- Faculdade de Medicina Veterinária, The Interdisciplinary Centre of Research in Animal Health (CIISA), Universidade de Lisboa, Lisboa, Portugal
| | - Joaquim M. S. Cabral
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Daniel Eberli
- Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Cláudia L. da Silva
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Pedro M. Baptista
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Instituto de Investigacion Sanitaria de Aragón (IIS Aragon), Zaragoza, Spain
- CIBERehd, Zaragoza, Spain
| |
Collapse
|
31
|
Zhang W, Choi JK, He X. Engineering Microvascularized 3D Tissue Using Alginate-Chitosan Microcapsules. J BIOMATER TISS ENG 2017; 7:170-173. [PMID: 29399384 PMCID: PMC5794023 DOI: 10.1166/jbt.2017.1547] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Construction of vascularized tissues is one of the major challenges of tissue engineering. The goal of this study was to engineer 3D microvascular tissues by incorporating the HUVEC-CS cells with a collagen/alginate-chitosan (AC) microcapsule scaffold. In the presence of AC microcapsules, a 3D vascular-like network was clearly observable. The results indicated the importance of AC microcapsules in engineering microvascular tissues -- providing support and guiding alignment of HUVEC-CS cells. This approach provides an alternative and promising method for constructing vascularized tissues.
Collapse
Affiliation(s)
- Wujie Zhang
- Biomolecular Engineering Program, Department of Physics and Chemistry, Milwaukee School of Engineering, Milwaukee, WI 53202, USA
| | - Jung K. Choi
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Xiaoming He
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
32
|
Junatas KL, Tonar Z, Kubíková T, Liška V, Pálek R, Mik P, Králíčková M, Witter K. Stereological analysis of size and density of hepatocytes in the porcine liver. J Anat 2016; 230:575-588. [PMID: 28032348 DOI: 10.1111/joa.12585] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2016] [Indexed: 02/06/2023] Open
Abstract
The porcine liver is frequently used as a large animal model for verification of surgical techniques, as well as experimental therapies. Often, a histological evaluation is required that include measurements of the size, nuclearity or density of hepatocytes. Our aims were to assess the mean number-weighted volume of hepatocytes, the numerical density of hepatocytes, and the fraction of binuclear hepatocytes (BnHEP) in the porcine liver, and compare the distribution of these parameters among hepatic lobes and macroscopic regions of interest (ROIs) with different positions related to the liver vasculature. Using disector and nucleator as design-based stereological methods, the morphometry of hepatocytes was quantified in seven healthy piglets. The samples were obtained from all six hepatic lobes and three ROIs (peripheral, paracaval and paraportal) within each lobe. Histological sections (thickness 16 μm) of formalin-fixed paraffin-embedded material were stained with the periodic acid-Schiff reaction to indicate the cell outlines and were assessed in a series of 3-μm-thick optical sections. The mean number-weighted volume of mononuclear hepatocytes (MnHEP) in all samples was 3670 ± 805 μm3 (mean ± SD). The mean number-weighted volume of BnHEP was 7050 ± 2550 μm3 . The fraction of BnHEP was 4 ± 2%. The numerical density of all hepatocytes was 146 997 ± 15 738 cells mm-3 of liver parenchyma. The porcine hepatic lobes contained hepatocytes of a comparable size, nuclearity and density. No significant differences were identified between the lobes. The peripheral ROIs of the hepatic lobes contained the largest MnHEP with the smallest numerical density. The distribution of a larger MnHEP was correlated with a larger volume of BnHEP and a smaller numerical density of all hepatocytes. Practical recommendations for designing studies that involve stereological evaluations of the size, nuclearity and density of hepatocytes in porcine liver are provided.
Collapse
Affiliation(s)
- Khan L Junatas
- Department for Pathobiology, Institute of Anatomy, Histology and Embryology, University of Veterinary Medicine Vienna, Vienna, Austria.,College of Veterinary Medicine, University of Southern Mindanao, Cotabato, Philippines
| | - Zbyněk Tonar
- Department of Histology and Embryology and Biomedical Center, Faculty of Medicine in Pilsen, Charles University in Prague, Pilsen, Czech Republic
| | - Tereza Kubíková
- NTIS, European Centre of Excellence, Faculty of Applied Sciences, University of West Bohemia, Pilsen, Czech Republic
| | - Václav Liška
- Department of Surgery and Biomedical Center, Faculty of Medicine in Pilsen, Charles University in Prague, Pilsen, Czech Republic
| | - Richard Pálek
- Department of Surgery and Biomedical Center, Faculty of Medicine in Pilsen, Charles University in Prague, Pilsen, Czech Republic
| | - Patrik Mik
- Department of Anatomy, Faculty of Medicine in Pilsen, Charles University in Prague, Pilsen, Czech Republic
| | - Milena Králíčková
- Department of Histology and Embryology and Biomedical Center, Faculty of Medicine in Pilsen, Charles University in Prague, Pilsen, Czech Republic
| | - Kirsti Witter
- Department for Pathobiology, Institute of Anatomy, Histology and Embryology, University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|
33
|
Wu Q, Li Y, Wang Y, Li L, Jiang X, Tang J, Yang H, Zhang J, Bao J, Bu H. The effect of heparinized decellularized scaffolds on angiogenic capability. J Biomed Mater Res A 2016; 104:3021-3030. [PMID: 27459086 DOI: 10.1002/jbm.a.35843] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 07/13/2016] [Accepted: 07/25/2016] [Indexed: 02/05/2023]
Affiliation(s)
- Qiong Wu
- Laboratory of Pathology; West China Hospital, Sichuan University; Chengdu 610041 China
- Department of Transplantation Pathology, Key Laboratory of Transplant Engineering and Immunology, Ministry of Health; West China Hospital, Sichuan University; Chengdu 610041 China
| | - Yi Li
- Laboratory of Pathology; West China Hospital, Sichuan University; Chengdu 610041 China
- Department of Transplantation Pathology, Key Laboratory of Transplant Engineering and Immunology, Ministry of Health; West China Hospital, Sichuan University; Chengdu 610041 China
| | - Yujia Wang
- Laboratory of Pathology; West China Hospital, Sichuan University; Chengdu 610041 China
- Department of Transplantation Pathology, Key Laboratory of Transplant Engineering and Immunology, Ministry of Health; West China Hospital, Sichuan University; Chengdu 610041 China
| | - Li Li
- Laboratory of Pathology; West China Hospital, Sichuan University; Chengdu 610041 China
- Department of Transplantation Pathology, Key Laboratory of Transplant Engineering and Immunology, Ministry of Health; West China Hospital, Sichuan University; Chengdu 610041 China
| | - Xin Jiang
- Department of Biomedical Engineering, College of Polymer Science and Engineering; Sichuan University; Chengdu 610041 China
| | - Jing Tang
- Department of General Surgery; the First People's Hospital of Yibin; Yibin 644000 China
| | - Hao Yang
- Department of Transplantation Pathology, Key Laboratory of Transplant Engineering and Immunology, Ministry of Health; West China Hospital, Sichuan University; Chengdu 610041 China
| | - Jie Zhang
- Department of Transplantation Pathology, Key Laboratory of Transplant Engineering and Immunology, Ministry of Health; West China Hospital, Sichuan University; Chengdu 610041 China
| | - Ji Bao
- Laboratory of Pathology; West China Hospital, Sichuan University; Chengdu 610041 China
- Department of Transplantation Pathology, Key Laboratory of Transplant Engineering and Immunology, Ministry of Health; West China Hospital, Sichuan University; Chengdu 610041 China
| | - Hong Bu
- Laboratory of Pathology; West China Hospital, Sichuan University; Chengdu 610041 China
- Department of Transplantation Pathology, Key Laboratory of Transplant Engineering and Immunology, Ministry of Health; West China Hospital, Sichuan University; Chengdu 610041 China
- Department of Pathology; West China Hospital, Sichuan University; Chengdu 610041 China
| |
Collapse
|
34
|
Bao J, Wu Q, Wang Y, Li Y, Li L, Chen F, Wu X, Xie M, Bu H. Enhanced hepatic differentiation of rat bone marrow-derived mesenchymal stem cells in spheroidal aggregate culture on a decellularized liver scaffold. Int J Mol Med 2016; 38:457-65. [PMID: 27314916 PMCID: PMC4935452 DOI: 10.3892/ijmm.2016.2638] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 06/01/2016] [Indexed: 02/05/2023] Open
Abstract
In the present study, we aimed to determine whether the combination of aggregate culture and decellularized liver scaffolds (DLSs) promoted the hepatic differentiation of murine bone marrow-derived mesenchymal stem cells (BM-MSCs) into high yields of mature hepatocytes in vitro. Four culturing methods for differentiation [single cell (2D), spheroids (3D), 2D + DLS and 3D + DLS] were studied. To determine the differentiation stages of the MSCs, RT-qPCR of the hepatocyte genes, immunostaining of hepatocyte markers, and functional analyses were all performed. Compared with the other groups, hepatocyte-like cells which differentiated from BM-MSC spheroids on extracellular matrix (ECM) exhibited more intensive staining of stored glycogen, an elevated level of urea biosynthesis and albumin secretion as well as the higher expression of hepatocyte-specific genes. Our results indicated that DLSs combined with spheroidal aggregate culture may be used as an effective method to facilitate the hepatic maturation of BM-MSCs and may have future applications in stem cell-based liver regenerative medicine.
Collapse
Affiliation(s)
- Ji Bao
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Qiong Wu
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yujia Wang
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yi Li
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Li Li
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Fei Chen
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiujuan Wu
- Department of General Surgery, Yibin City First People's Hospital, Yibin, Sichuan 644000, P.R. China
| | - Mingjun Xie
- Department of General Surgery, Yibin City First People's Hospital, Yibin, Sichuan 644000, P.R. China
| | - Hong Bu
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|