1
|
Yadav CJ, Yadav U, Afrin S, Lee JY, Kamel J, Park KM. Heparin Immobilization Enhances Hemocompatibility, Re-Endothelization, and Angiogenesis of Decellularized Liver Scaffolds. Int J Mol Sci 2024; 25:12132. [PMID: 39596200 PMCID: PMC11595110 DOI: 10.3390/ijms252212132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/04/2024] [Accepted: 11/09/2024] [Indexed: 11/28/2024] Open
Abstract
Bioengineered livers are currently an acceptable alternative to orthotopic liver transplants to overcome the scarcity of donors. However, the challenge of using a bioengineered liver is the lack of an intact endothelial layer in the vascular network leading to thrombosis. Heparin-modified surfaces have been demonstrated to decrease thrombogenicity in earlier research. However, in our study, we aimed to apply heparin immobilization to enhance the hemocompatibility, endothelial cell (EC) adhesion, and angiogenesis of rat decellularized liver scaffolds (DLS). Heparin was immobilized on the DLS by the end-point attachment technique. The scaffold's hemocompatibility was assessed using ex vivo blood perfusion and platelet adhesion studies. The heparinized scaffold (HEP-DLS) showed a significantly reduced thrombogenicity and platelet aggregation. HEP-DLS was recellularized with EA.hy926 cells via the portal vein and maintained in the bioreactor for 7 days, showing increased EC adhesion and coverage within the blood vessels. The Resazurin reduction assay confirmed the presence of actively proliferating cells in the HEP-DLS. The scaffolds were implanted subcutaneously into the dorsum of mice for 21 days to evaluate cell migration and angiogenesis. The results showed significant increases in the number of blood vessels in the HEP-DLS group. Our results demonstrated that heparin immobilization reduces thrombosis, promotes re-endothelialization, and enhances angiogenesis in DLS. The research provides insight into the potential use of heparin in the formation of a functioning vasculature.
Collapse
Affiliation(s)
| | | | | | | | | | - Kyung-Mee Park
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (C.J.Y.); (U.Y.); (S.A.); (J.-Y.L.); (J.K.)
| |
Collapse
|
2
|
Martins B, Bister A, Dohmen RGJ, Gouveia MA, Hueber R, Melzener L, Messmer T, Papadopoulos J, Pimenta J, Raina D, Schaeken L, Shirley S, Bouchet BP, Flack JE. Advances and Challenges in Cell Biology for Cultured Meat. Annu Rev Anim Biosci 2024; 12:345-368. [PMID: 37963400 DOI: 10.1146/annurev-animal-021022-055132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
Cultured meat is an emerging biotechnology that aims to produce meat from animal cell culture, rather than from the raising and slaughtering of livestock, on environmental and animal welfare grounds. The detailed understanding and accurate manipulation of cell biology are critical to the design of cultured meat bioprocesses. Recent years have seen significant interest in this field, with numerous scientific and commercial breakthroughs. Nevertheless, these technologies remain at a nascent stage, and myriad challenges remain, spanning the entire bioprocess. From a cell biological perspective, these include the identification of suitable starting cell types, tuning of proliferation and differentiation conditions, and optimization of cell-biomaterial interactions to create nutritious, enticing foods. Here, we discuss the key advances and outstanding challenges in cultured meat, with a particular focus on cell biology, and argue that solving the remaining bottlenecks in a cost-effective, scalable fashion will require coordinated, concerted scientific efforts. Success will also require solutions to nonscientific challenges, including regulatory approval, consumer acceptance, and market feasibility. However, if these can be overcome, cultured meat technologies can revolutionize our approach to food.
Collapse
Affiliation(s)
- Beatriz Martins
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
| | - Arthur Bister
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
| | - Richard G J Dohmen
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
- Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | - Maria Ana Gouveia
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
| | - Rui Hueber
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
| | - Lea Melzener
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
- Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | - Tobias Messmer
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
- Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | - Joanna Papadopoulos
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
| | - Joana Pimenta
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
| | - Dhruv Raina
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
| | - Lieke Schaeken
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
| | - Sara Shirley
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
| | - Benjamin P Bouchet
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands;
| | - Joshua E Flack
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
| |
Collapse
|
3
|
Gupta S, Sharma A, Petrovski G, Verma RS. Vascular reconstruction of the decellularized biomatrix for whole-organ engineering-a critical perspective and future strategies. Front Bioeng Biotechnol 2023; 11:1221159. [PMID: 38026872 PMCID: PMC10680456 DOI: 10.3389/fbioe.2023.1221159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Whole-organ re-engineering is the most challenging goal yet to be achieved in tissue engineering and regenerative medicine. One essential factor in any transplantable and functional tissue engineering is fabricating a perfusable vascular network with macro- and micro-sized blood vessels. Whole-organ development has become more practical with the use of the decellularized organ biomatrix (DOB) as it provides a native biochemical and structural framework for a particular organ. However, reconstructing vasculature and re-endothelialization in the DOB is a highly challenging task and has not been achieved for constructing a clinically transplantable vascularized organ with an efficient perfusable capability. Here, we critically and articulately emphasized factors that have been studied for the vascular reconstruction in the DOB. Furthermore, we highlighted the factors used for vasculature development studies in general and their application in whole-organ vascular reconstruction. We also analyzed in detail the strategies explored so far for vascular reconstruction and angiogenesis in the DOB for functional and perfusable vasculature development. Finally, we discussed some of the crucial factors that have been largely ignored in the vascular reconstruction of the DOB and the future directions that should be addressed systematically.
Collapse
Affiliation(s)
- Santosh Gupta
- Stem Cell and Molecular Biology, Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences. Indian Institute of Technology Madras, Chennai, India
- Center for Eye Research and Innovative Diagnostics, Department of Ophthalmology, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Akriti Sharma
- Stem Cell and Molecular Biology, Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences. Indian Institute of Technology Madras, Chennai, India
| | - Goran Petrovski
- Center for Eye Research and Innovative Diagnostics, Department of Ophthalmology, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Ophthalmology, Oslo University Hospital, Oslo, Norway
- Department of Ophthalmology, University of Split School of Medicine and University Hospital Centre, Split, Croatia
| | - Rama Shanker Verma
- Stem Cell and Molecular Biology, Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences. Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
4
|
Uzarski JS, Beck EC, Russell EE, Vanderslice EJ, Holzner ML, Wadhera V, Adamson D, Shapiro R, Davidow DS, Ross JJ, Florman SS. Sustained in vivo perfusion of a re-endothelialized tissue engineered kidney graft in a human-scale animal model. Front Bioeng Biotechnol 2023; 11:1184408. [PMID: 37388767 PMCID: PMC10307518 DOI: 10.3389/fbioe.2023.1184408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 05/25/2023] [Indexed: 07/01/2023] Open
Abstract
Introduction: Despite progress in whole-organ decellularization and recellularization, maintaining long-term perfusion in vivo remains a hurdle to realizing clinical translation of bioengineered kidney grafts. The objectives for the present study were to define a threshold glucose consumption rate (GCR) that could be used to predict in vivo graft hemocompatibility and utilize this threshold to assess the in vivo performance of clinically relevant decellularized porcine kidney grafts recellularized with human umbilical vein endothelial cells (HUVECs). Materials and methods: Twenty-two porcine kidneys were decellularized and 19 were re-endothelialized using HUVECs. Functional revascularization of control decellularized (n = 3) and re-endothelialized porcine kidneys (n = 16) was tested using an ex vivo porcine blood flow model to define an appropriate metabolic glucose consumption rate (GCR) threshold above which would sustain patent blood flow. Re-endothelialized grafts (n = 9) were then transplanted into immunosuppressed pigs with perfusion measured using angiography post-implant and on days 3 and 7 with 3 native kidneys used as controls. Patent recellularized kidney grafts underwent histological analysis following explant. Results: The glucose consumption rate of recellularized kidney grafts reached a peak of 39.9 ± 9.7 mg/h at 21 ± 5 days, at which point the grafts were determined to have sufficient histological vascular coverage with endothelial cells. Based on these results, a minimum glucose consumption rate threshold of 20 mg/h was set. The revascularized kidneys had a mean perfusion percentage of 87.7% ± 10.3%, 80.9% ± 33.1%, and 68.5% ± 38.6% post-reperfusion on Days 0, 3 and 7, respectively. The 3 native kidneys had a mean post-perfusion percentage of 98.4% ± 1.6%. These results were not statistically significant. Conclusion: This study is the first to demonstrate that human-scale bioengineered porcine kidney grafts developed via perfusion decellularization and subsequent re-endothelialization using HUVEC can maintain patency with consistent blood flow for up to 7 days in vivo. These results lay the foundation for future research to produce human-scale recellularized kidney grafts for transplantation.
Collapse
Affiliation(s)
| | - Emily C. Beck
- Miromatrix Medical Inc., Eden Prairie, MN, United States
| | | | | | - Matthew L. Holzner
- Icahn School of Medicine at Mount Sinai, Recanati/Miller Transplantation Institute, New York, NY, United States
| | - Vikram Wadhera
- Icahn School of Medicine at Mount Sinai, Recanati/Miller Transplantation Institute, New York, NY, United States
| | - Dylan Adamson
- Icahn School of Medicine at Mount Sinai, Recanati/Miller Transplantation Institute, New York, NY, United States
| | - Ron Shapiro
- Icahn School of Medicine at Mount Sinai, Recanati/Miller Transplantation Institute, New York, NY, United States
| | | | - Jeff J. Ross
- Miromatrix Medical Inc., Eden Prairie, MN, United States
| | - Sander S. Florman
- Icahn School of Medicine at Mount Sinai, Recanati/Miller Transplantation Institute, New York, NY, United States
| |
Collapse
|
5
|
Sauter J, Degenhardt H, Tuebel J, Foehr P, Knoeckel P, Florian K, Charitou F, Burgkart R, Schmitt A. Effect of different decellularization protocols on reendothelialization with human cells for a perfused renal bioscaffold of the rat. BMC Biotechnol 2023; 23:8. [PMID: 36927344 PMCID: PMC10022115 DOI: 10.1186/s12896-022-00767-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 11/09/2022] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND Scaffolds for tissue engineering can be received by whole organ decellularization while maintaining the site-specific extracellular matrix and the vascular tree. One among other decellularization techniques is the perfusion-based method using specific agents e.g. SDS for the elimination of cellular components. While SDS can disrupt the composition of the extracellular matrix and impair the adherence and growth of site-specific cells there are indications that xenogeneic cell types may benefit from protein denaturation by using higher detergent concentrations. The aim of this work is to investigate the effect of two different SDS-concentrations (i.e. 0.66% and 3%) on the ability of human endothelial cells to adhere and proliferate in an acellular rat kidney scaffold. MATERIAL AND METHODS Acellular rat kidney scaffold was obtained by perfusion-based decellularization through the renal artery using a standardized protocol including SDS at concentrations of 0.66% or 3%. Subsequently cell seeding was performed with human immortalized endothelial cells EA.hy 926 via the renal artery. Recellularized kidneys were harvested after five days of pressure-controlled dynamic culture followed sectioning, histochemical and immunohistochemical staining as well as semiquantitative analysis. RESULTS Efficacy of decellularization was verified by absence of cellular components as well as preservation of ultrastructure and adhesive proteins of the extracellular matrix. In semiquantitative analysis of recellularization, cell count after five days of dynamic culture more than doubled when using the gentle decellularization protocol with a concentration of SDS at 0.66% compared to 3%. Detectable cells maintained their endothelial phenotype and presented proliferative behavior while only a negligible fraction underwent apoptosis. CONCLUSION Recellularization of acellular kidney scaffold with endothelial cells EA.hy 926 seeded through the renal artery benefits from gentle decellularization procedure. Because of that, decellularization with a SDS concentration at 0.66% should be preferred in further studies and coculture experiments.
Collapse
Affiliation(s)
- Johannes Sauter
- Department of Orthopedics and Sports Orthopedics, Klinikum Rechts der Isar der Technischen Universität München, Munich, Germany.,Department of Medicine II, LMU Klinikum München, Munich, Germany
| | - Hannes Degenhardt
- Division of Sports Orthopedics, Klinikum Rechts der Isar der Technischen Universität München, Munich, Germany
| | - Jutta Tuebel
- Department of Orthopedics and Sports Orthopedics, Klinikum Rechts der Isar der Technischen Universität München, Munich, Germany
| | - Peter Foehr
- Department of Orthopedics and Sports Orthopedics, Klinikum Rechts der Isar der Technischen Universität München, Munich, Germany
| | | | - Kira Florian
- Department of Orthopedics and Sports Orthopedics, Klinikum Rechts der Isar der Technischen Universität München, Munich, Germany
| | - Fiona Charitou
- Department of Orthopedics and Sports Orthopedics, Klinikum Rechts der Isar der Technischen Universität München, Munich, Germany
| | - Rainer Burgkart
- Department of Orthopedics and Sports Orthopedics, Klinikum Rechts der Isar der Technischen Universität München, Munich, Germany.
| | - Andreas Schmitt
- Division of Sports Orthopedics, Klinikum Rechts der Isar der Technischen Universität München, Munich, Germany.,Orthopädisches Fachzentrum Weilheim, Weilheim, Germany
| |
Collapse
|
6
|
Wu P, Asada H, Hakamada M, Mabuchi M. Bioengineering of High Cell Density Tissues with Hierarchical Vascular Networks for Ex Vivo Whole Organs. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2209149. [PMID: 36545785 DOI: 10.1002/adma.202209149] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/25/2022] [Indexed: 06/17/2023]
Abstract
The development of tissue-like structures such as cell sheets, spheroids, and organoids has contributed to progress in regenerative medicine. Simultaneous achievement of scale up and high cell density of these tissues is challenging because sufficient oxygen cannot be supplied to the inside of large, high cell density tissues. Here, in vitro fabrication of vessels to supply oxygen to the inside of millimeter-sized scaffold-free tissues whose cell density is ≈200 million cells mL-1 , corresponding to those of native tissues, is shown. Hierarchical vascular networks by anastomosis of capillaries and a large vessel are essential for oxygen supply, whereas a large vessel or capillary networks alone make negligible contributions to the supply. The hierarchical vascular networks are formed by a top-down approach, which offers a new option for ex vivo whole organs without decellularization and 3D-bioprinting.
Collapse
Affiliation(s)
- Peizheng Wu
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto, 606-8501, Japan
| | - Hiroki Asada
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto, 606-8501, Japan
| | - Masataka Hakamada
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto, 606-8501, Japan
| | - Mamoru Mabuchi
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto, 606-8501, Japan
| |
Collapse
|
7
|
Tissue engineering of decellularized pancreas scaffolds for regenerative medicine in diabetes. Acta Biomater 2023; 157:49-66. [PMID: 36427686 DOI: 10.1016/j.actbio.2022.11.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/12/2022] [Accepted: 11/16/2022] [Indexed: 11/27/2022]
Abstract
Diabetes mellitus is a global disease requiring long-term treatment and monitoring. At present, pancreas or islet transplantation is the only reliable treatment for achieving stable euglycemia in Type I diabetes patients. However, the shortage of viable pancreata for transplantation limits the use of this therapy for the majority of patients. Organ decellularization and recellularization is emerging as a promising solution to overcome the shortage of viable organs for transplantation by providing a potential alternative source of donor organs. Several studies on decellularization and recellularization of rodent, porcine, and human pancreata have been performed, and show promise for generating usable decellularized pancreas scaffolds for subsequent recellularization and transplantation. In this state-of-the-art review, we provide an overview of the latest advances in pancreas decellularization, recellularization, and revascularization. We also discuss clinical considerations such as potential transplantation sites, donor source, and immune considerations. We conclude with an outlook on the remaining work that needs to be done in order to realize the goal of using this technology to create bioengineered pancreata for transplantation in diabetes patients. STATEMENT OF SIGNIFICANCE: Pancreas or islet transplantation is a means of providing insulin-independence in diabetes patients. However, due to the shortage of viable pancreata, whole-organ decellularization and recellularization is emerging as a promising solution to overcome organ shortage for transplantation. Several studies on decellularization and recellularization of rodent, porcine, and human pancreata have shown promise for generating usable decellularized pancreas scaffolds for subsequent recellularization and transplantation. In this state-of-the-art review, we highlight the latest advances in pancreas decellularization, recellularization, and revascularization. We also discuss clinical considerations such as potential transplantation sites, donor source, and immune considerations. We conclude with future work that needs to be done in order to realize clinical translation of bioengineered pancreata for transplantation in diabetes patients.
Collapse
|
8
|
Whole-Heart Tissue Engineering and Cardiac Patches: Challenges and Promises. BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010106. [PMID: 36671678 PMCID: PMC9855348 DOI: 10.3390/bioengineering10010106] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/02/2023] [Accepted: 01/05/2023] [Indexed: 01/15/2023]
Abstract
Despite all the advances in preventing, diagnosing, and treating cardiovascular disorders, they still account for a significant part of mortality and morbidity worldwide. The advent of tissue engineering and regenerative medicine has provided novel therapeutic approaches for the treatment of various diseases. Tissue engineering relies on three pillars: scaffolds, stem cells, and growth factors. Gene and cell therapy methods have been introduced as primary approaches to cardiac tissue engineering. Although the application of gene and cell therapy has resulted in improved regeneration of damaged cardiac tissue, further studies are needed to resolve their limitations, enhance their effectiveness, and translate them into the clinical setting. Scaffolds from synthetic, natural, or decellularized sources have provided desirable characteristics for the repair of cardiac tissue. Decellularized scaffolds are widely studied in heart regeneration, either as cell-free constructs or cell-seeded platforms. The application of human- or animal-derived decellularized heart patches has promoted the regeneration of heart tissue through in vivo and in vitro studies. Due to the complexity of cardiac tissue engineering, there is still a long way to go before cardiac patches or decellularized whole-heart scaffolds can be routinely used in clinical practice. This paper aims to review the decellularized whole-heart scaffolds and cardiac patches utilized in the regeneration of damaged cardiac tissue. Moreover, various decellularization methods related to these scaffolds will be discussed.
Collapse
|
9
|
Jo Y, Hwang DG, Kim M, Yong U, Jang J. Bioprinting-assisted tissue assembly to generate organ substitutes at scale. Trends Biotechnol 2023; 41:93-105. [PMID: 35907704 DOI: 10.1016/j.tibtech.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/29/2022] [Accepted: 07/01/2022] [Indexed: 12/27/2022]
Abstract
Various external cues can guide cellular behavior and maturation during developmental processes. Recent studies on bioprinting-assisted tissue engineering have considered this a practical, versatile, and flexible way to provide external cues to developing engineered tissues. An ensemble of multiple external cues can improve the speed and capability of morphogenesis. In this review, we discuss how bioprinting and biomaterials provide multiple guidance to generate micro-sized building blocks with specific shapes and also highlight their applications in tissue assembly toward volumetric tissue and organ generation. Furthermore, we discuss our perspectives on the future translation of bioprinting technologies integrated with artificial intelligence (AI) and robot-assisted apparatus to promote automation, standardization, and clinical translation of bioprinted tissues.
Collapse
Affiliation(s)
- Yeonggwon Jo
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Dong Gyu Hwang
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Myungji Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Uijung Yong
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Jinah Jang
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea; Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea; Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea; Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
10
|
Chen Y, Ding BS. Comprehensive Review of the Vascular Niche in Regulating Organ Regeneration and Fibrosis. Stem Cells Transl Med 2022; 11:1135-1142. [PMID: 36169406 DOI: 10.1093/stcltm/szac070] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/28/2022] [Indexed: 11/14/2022] Open
Abstract
The vasculature occupies a large area of the body, and none of the physiological activities can be carried out without blood vessels. Blood vessels are not just passive conduits and barriers for delivering blood and nutrients. Meanwhile, endothelial cells covering the vascular lumen establish vascular niches by deploying some growth factors, known as angiocrine factors, and actively participate in the regulation of a variety of physiological processes, such as organ regeneration and fibrosis and the occurrence and development of cancer. After organ injury, vascular endothelial cells regulate the repair process by secreting various angiocrine factors, triggering the proliferation and differentiation process of stem cells. Therefore, analyzing the vascular niche and exploring the factors that maintain vascular homeostasis can provide strong theoretical support for clinical treatment targeting blood vessels. Here we mainly discuss the regulatory mechanisms of the vascular niche in organ regeneration and fibrosis.
Collapse
Affiliation(s)
- Yutian Chen
- The Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Bi-Sen Ding
- State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
11
|
Hannon E, Pellegrini M, Scottoni F, Durkin N, Shibuya S, Lutman R, Proctor TJ, Hutchinson JC, Arthurs OJ, Phylactopoulos DE, Maughan EF, Butler CR, Eaton S, Lowdell MW, Bonfanti P, Urbani L, De Coppi P. Lessons learned from pre-clinical testing of xenogeneic decellularized esophagi in a rabbit model. iScience 2022; 25:105174. [PMID: 36217545 PMCID: PMC9547295 DOI: 10.1016/j.isci.2022.105174] [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: 12/13/2021] [Revised: 06/21/2022] [Accepted: 09/19/2022] [Indexed: 11/04/2022] Open
Abstract
Decellularization of esophagi from several species for tissue engineering is well described, but successful implantation in animal models of esophageal replacement has been challenging. The purpose of this study was to assess feasibility and applicability of esophageal replacement using decellularized porcine esophageal scaffolds in a new pre-clinical model. Following surgical replacement in rabbits with a vascularizing muscle flap, we observed successful anastomoses of decellularized scaffolds, cues of early neovascularization, and prevention of luminal collapse by the use of biodegradable stents. However, despite the success of the surgical procedure, the long-term survival was limited by the fragility of the animal model. Our results indicate that transplantation of a decellularized porcine scaffold is possible and vascular flaps may be useful to provide a vascular supply, but long-term outcomes require further pre-clinical testing in a different large animal model.
Collapse
Affiliation(s)
- Edward Hannon
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK,Department of Paediatric Surgery, Leeds Children’s Hospital, Leeds Teaching Hospitals NHS Trust, Leeds LS1 3EX, UK
| | - Marco Pellegrini
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Federico Scottoni
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK,Department of Pediatric Surgery, Regina Margherita Children’s Hospital, Turin 10126, Italy
| | - Natalie Durkin
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Soichi Shibuya
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Roberto Lutman
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Toby J. Proctor
- Centre for Cell, Gene and Tissue Therapies, Royal Free Hospital & University College London, London NW3 2PF, UK
| | - J. Ciaran Hutchinson
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK,Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Owen J. Arthurs
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK,Department of Radiology, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Demetra-Ellie Phylactopoulos
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK,Epithelial Stem Cell Biology & Regenerative Medicine Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Elizabeth F. Maughan
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK,Charing Cross Airway Service, Department of Otolaryngology, Charing Cross Hospital, Imperial Healthcare NHS Trust, London W6 8RF, UK
| | - Colin R. Butler
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK,ENT Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Simon Eaton
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Mark W. Lowdell
- Centre for Cell, Gene and Tissue Therapies, Royal Free Hospital & University College London, London NW3 2PF, UK
| | - Paola Bonfanti
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK,Epithelial Stem Cell Biology & Regenerative Medicine Laboratory, The Francis Crick Institute, London NW1 1AT, UK,Institute of Immunity & Transplantation, University College London, London NW3 2PP, UK
| | - Luca Urbani
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London SE5 9NT, UK,Faculty of Life Sciences and Medicine, King’s College London, London SE5 8AF, UK
| | - Paolo De Coppi
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK,Specialist Neonatal and Paediatric Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK,Corresponding author
| |
Collapse
|
12
|
Tam PKH, Wong KKY, Atala A, Giobbe GG, Booth C, Gruber PJ, Monone M, Rafii S, Rando TA, Vacanti J, Comer CD, Elvassore N, Grikscheit T, de Coppi P. Regenerative medicine: postnatal approaches. THE LANCET. CHILD & ADOLESCENT HEALTH 2022; 6:654-666. [PMID: 35963270 DOI: 10.1016/s2352-4642(22)00193-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 05/20/2022] [Accepted: 05/27/2022] [Indexed: 06/15/2023]
Abstract
Paper 2 of the paediatric regenerative medicine Series focuses on recent advances in postnatal approaches. New gene, cell, and niche-based technologies and their combinations allow structural and functional reconstitution and simulation of complex postnatal cell, tissue, and organ hierarchies. Organoid and tissue engineering advances provide human disease models and novel treatments for both rare paediatric diseases and common diseases affecting all ages, such as COVID-19. Preclinical studies for gastrointestinal disorders are directed towards oesophageal replacement, short bowel syndrome, enteric neuropathy, biliary atresia, and chronic end-stage liver failure. For respiratory diseases, beside the first human tracheal replacement, more complex tissue engineering represents a promising solution to generate transplantable lungs. Genitourinary tissue replacement and expansion usually involve application of biocompatible scaffolds seeded with patient-derived cells. Gene and cell therapy approaches seem appropriate for rare paediatric diseases of the musculoskeletal system such as spinal muscular dystrophy, whereas congenital diseases of complex organs, such as the heart, continue to challenge new frontiers of regenerative medicine.
Collapse
Affiliation(s)
- Paul Kwong Hang Tam
- Faculty of Medicine, Macau University of Science and Technology, Macau Special Administrative Region, China; Division of Paediatric Surgery, Department of Surgery, Queen Mary Hospital, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Kenneth Kak Yuen Wong
- Division of Paediatric Surgery, Department of Surgery, Queen Mary Hospital, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| | - Giovanni Giuseppe Giobbe
- Stem Cell and Regenerative Medicine Section, Developmental Biology and Cancer Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Claire Booth
- Stem Cell and Regenerative Medicine Section, Developmental Biology and Cancer Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Peter J Gruber
- Department of Surgery, Yale University, New Haven, CT, USA
| | - Mimmi Monone
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Shahin Rafii
- Ansary Stem Cell Institute, Department of Medicine, Division of Regenerative Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Thomas A Rando
- Paul F Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph Vacanti
- Department of Pediatric Surgery, Laboratory for Tissue Engineering and Organ Fabrication, Harvard Medical School, Massachusetts General Hospital, Mass General Hospital for Children, Boston, MA, USA
| | - Carly D Comer
- Department of Pediatric Surgery, Laboratory for Tissue Engineering and Organ Fabrication, Harvard Medical School, Massachusetts General Hospital, Mass General Hospital for Children, Boston, MA, USA
| | - Nicola Elvassore
- Stem Cell and Regenerative Medicine Section, Developmental Biology and Cancer Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK; Department of Industrial Engineering, University of Padova, Padova, Italy
| | - Tracy Grikscheit
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Paolo de Coppi
- Stem Cell and Regenerative Medicine Section, Developmental Biology and Cancer Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK; Department of Specialist Neonatal and Paediatric Surgery, Great Ormond Street Hospital, London, UK.
| |
Collapse
|
13
|
Preethy S, Ranganathan N, Raghavan K, Dedeepiya VD, Ikewaki N, Abraham SJ. Integrating the Synergy of the Gut Microbiome into Regenerative Medicine: Relevance to Neurological Disorders. J Alzheimers Dis 2022; 87:1451-1460. [PMID: 35466942 PMCID: PMC9277691 DOI: 10.3233/jad-220313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2022] [Indexed: 11/29/2022]
Abstract
A new paradigm of cell therapy-based approaches as a solution to several diseases caused by damage or loss of cells/tissues leading to organ failure heralded the birth of a new branch in medicine called regenerative medicine (RM), which was further fueled by in vitro cell expansion and tissue engineering (TE) technologies, including the ability to grow embryonic stem cells, induce pluripotent stem cells, and so on. RM addresses organ failure by repair, regeneration, or restoration, rejuvenation using cells, stem cells, or progenitor cells as tools having added cell-derived products also as a tool, and extracellular matrix component-based support, either direct or indirect (e.g., matrix induced autologous chondrocyte implantation) using scaffolds. Now, the main objective of RM is to solve the functional loss of cells that have evolved from cells as tools to cell-derived factors and scaffolds per se as tools. In this context, an important yet indispensable group of cells that constitute the major portion of the human body in terms of the number of cells having several essential roles to play, both directly and indirectly, starting from digestion and the immune system to the growing evidence of influencing neuronal function, aging, and carcinogenesis has been ignored. We would like to focus on these in this review as they should essentially be considered as a tool of RM, especially for neurological disorders for their vital role. What we are indicating is the second genome or the gut microbiome.
Collapse
Affiliation(s)
- Senthilkumar Preethy
- Fujio-Eiji Academic Terrain (FEAT), Nichi-In Centre for Regenerative Medicine (NCRM), Chennai, India
| | | | - Kadalraja Raghavan
- Department of Paediatric Neurology, Jesuit Antonyraj memorial Inter-disciplinary Centre for Advanced Recovery and Education (JAICARE), Madurai, India
- Department of Paediatric Neurology, Sarvee Integra Private Limited, Chennai, India
| | | | - Nobunao Ikewaki
- Department of Medical Life Science, Kyushu University of Health and Welfare, Nobeoka, Japan
- Institute of Immunology, Junsei Educational Institute, Nobeoka, Japan
| | - Samuel J.K. Abraham
- Mary-Yoshio Translational Hexagon (MYTH), Nichi-In Centre for Regenerative Medicine (NCRM), Chennai, India
- Centre for Advancing Clinical Research (CACR), University of Yamanashi - School of Medicine, Chuo, Japan
- Antony- Xavier Interdisciplinary Scholastics (AXIS), GN Corporation Co. Ltd., Kofu, Japan
| |
Collapse
|
14
|
Wang WY, Kent RN, Huang SA, Jarman EH, Shikanov EH, Davidson CD, Hiraki HL, Lin D, Wall MA, Matera DL, Shin JW, Polacheck WJ, Shikanov A, Baker BM. Direct comparison of angiogenesis in natural and synthetic biomaterials reveals that matrix porosity regulates endothelial cell invasion speed and sprout diameter. Acta Biomater 2021; 135:260-273. [PMID: 34469789 PMCID: PMC8595798 DOI: 10.1016/j.actbio.2021.08.038] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/30/2021] [Accepted: 08/24/2021] [Indexed: 12/15/2022]
Abstract
Vascularization of large, diffusion-hindered biomaterial implants requires an understanding of how extracellular matrix (ECM) properties regulate angiogenesis. Sundry biomaterials assessed across many disparate angiogenesis assays have highlighted ECM determinants that influence this complex multicellular process. However, the abundance of material platforms, each with unique parameters to model endothelial cell (EC) sprouting presents additional challenges of interpretation and comparison between studies. In this work we directly compared the angiogenic potential of commonly utilized natural (collagen and fibrin) and synthetic dextran vinyl sulfone (DexVS) hydrogels in a multiplexed angiogenesis-on-a-chip platform. Modulating matrix density of collagen and fibrin hydrogels confirmed prior findings that increases in matrix density correspond to increased EC invasion as connected, multicellular sprouts, but with decreased invasion speeds. Angiogenesis in synthetic DexVS hydrogels, however, resulted in fewer multicellular sprouts. Characterizing hydrogel Young's modulus and permeability (a measure of matrix porosity), we identified matrix permeability to significantly correlate with EC invasion depth and sprout diameter. Although microporous collagen and fibrin hydrogels produced lumenized sprouts in vitro, they rapidly resorbed post-implantation into the murine epididymal fat pad. In contrast, DexVS hydrogels proved comparatively stable. To enhance angiogenesis within DexVS hydrogels, we incorporated sacrificial microgels to generate cell-scale pores throughout the hydrogel. Microporous DexVS hydrogels resulted in lumenized sprouts in vitro and enhanced cell invasion in vivo. Towards the design of vascularized biomaterials for long-term regenerative therapies, this work suggests that synthetic biomaterials offer improved size and shape control following implantation and that tuning matrix porosity may better support host angiogenesis. STATEMENT OF SIGNIFICANCE: Understanding how extracellular matrix properties govern angiogenesis will inform biomaterial design for engineering vascularized implantable grafts. Here, we utilized a multiplexed angiogenesis-on-a-chip platform to compare the angiogenic potential of natural (collagen and fibrin) and synthetic dextran vinyl sulfone (DexVS) hydrogels. Characterization of matrix properties and sprout morphometrics across these materials points to matrix porosity as a critical regulator of sprout invasion speed and diameter, supported by the observation that nanoporous DexVS hydrogels yielded endothelial cell sprouts that were not perfusable. To enhance angiogenesis into synthetic hydrogels, we incorporated sacrificial microgels to generate microporosity. We find that microporosity increased sprout diameter in vitro and cell invasion in vivo. This work establishes a composite materials approach to enhance the vascularization of synthetic hydrogels.
Collapse
Affiliation(s)
- William Y Wang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Robert N Kent
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Stephanie A Huang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC 27514, United States
| | - Evan H Jarman
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Eve H Shikanov
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Christopher D Davidson
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Harrison L Hiraki
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Daphne Lin
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Monica A Wall
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Daniel L Matera
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Jae-Won Shin
- Department of Pharmacology and Regenerative Medicine & Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 60607, United States
| | - William J Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC 27514, United States; McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, United States
| | - Ariella Shikanov
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States; Department of Macromolecular Science & Engineering, University of Michigan, Ann Arbor, MI, 48109, United States; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
15
|
Shen L, Song X, Xu Y, Tian R, Wang Y, Li P, Li J, Bai H, Zhu H, Wang D. Patterned vascularization in a directional ice-templated scaffold of decellularized matrix. Eng Life Sci 2021; 21:683-692. [PMID: 34690638 PMCID: PMC8518570 DOI: 10.1002/elsc.202100034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/02/2021] [Accepted: 06/21/2021] [Indexed: 11/22/2022] Open
Abstract
Vascularization is fundamental for large-scale tissue engineering. Most of the current vascularization strategies including microfluidics and three-dimensional (3D) printing aim to precisely fabricate microchannels for individual microvessels. However, few studies have examined the remodeling capacity of the microvessels in the engineered constructs, which is important for transplantation in vivo. Here we present a method for patterning microvessels in a directional ice-templated scaffold of decellularized porcine kidney extracellular matrix. The aligned microchannels made by directional ice templating allowed for fast and efficient cell seeding. The pure decellularized matrix without any fixatives or cross-linkers maximized the potential of tissue remodeling. Dramatical microvascular remodeling happened in the scaffold in 2 weeks, from small primary microvessel segments to long patterned microvessels. The majority of the microvessels were aligned in parallel and interconnected with each other to form a network. This method is compatible with other engineering techniques, such as microfluidics and 3D printing, and multiple cell types can be co-cultured to make complex vascularized tissue and organ models.
Collapse
Affiliation(s)
- Li Shen
- Institute for Translational MedicineThe Affiliated Hospital of Qingdao UniversityMedical CollegeQingdao UniversityQingdaoP. R. China
- School of Basic MedicineQingdao UniversityQingdaoP. R. China
| | - Xiuyue Song
- Institute for Translational MedicineThe Affiliated Hospital of Qingdao UniversityMedical CollegeQingdao UniversityQingdaoP. R. China
| | - Yalan Xu
- Institute for Translational MedicineThe Affiliated Hospital of Qingdao UniversityMedical CollegeQingdao UniversityQingdaoP. R. China
| | - Runhua Tian
- Department of Clinical LaboratoryThe Affiliated Hospital of Qingdao UniversityQingdaoP. R. China
| | - Yin Wang
- Institute for Translational MedicineThe Affiliated Hospital of Qingdao UniversityMedical CollegeQingdao UniversityQingdaoP. R. China
| | - Peifeng Li
- Institute for Translational MedicineThe Affiliated Hospital of Qingdao UniversityMedical CollegeQingdao UniversityQingdaoP. R. China
| | - Jing Li
- Institute for Translational MedicineThe Affiliated Hospital of Qingdao UniversityMedical CollegeQingdao UniversityQingdaoP. R. China
| | - Hao Bai
- State Key Laboratory of Chemical EngineeringCollege of Chemical and Biological EngineeringZhejiang UniversityHangzhouP. R. China
| | - Hai Zhu
- Department of UrologyQingdao Municipal Hospital Affiliated to Qingdao UniversityQingdaoP. R. China
| | - Dong Wang
- Institute for Translational MedicineThe Affiliated Hospital of Qingdao UniversityMedical CollegeQingdao UniversityQingdaoP. R. China
| |
Collapse
|
16
|
Lupon E, Lellouch AG, Acun A, Andrews AR, Oganesyan R, Goutard M, Taveau CB, Lantieri LA, Cetrulo CL, Uygun BE. Engineering Vascularized Composite Allografts Using Natural Scaffolds: A Systematic Review. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:677-693. [PMID: 34238047 DOI: 10.1089/ten.teb.2021.0102] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Vascularized Composite Allotransplantation refers to the transplantation of multiple tissues as a functional unit from a deceased donor to a recipient with a severe injury. These grafts serve as potential replacements for traumatic tissue losses. The main problems are the consequences of the long immunosuppressive drugs medications and the lake of compatible donor. To avoid these limitations, decellularization/recellularization constitute an attractive approach. The aim of decellularization/recellularization technology is to develop immunogenic free biological substitutes that will restore, maintain, or improve tissue and organ's function. METHODS A PubMed search was performed for articles on decellularization and recellularization of composite tissue allografts between March and February 2021, with no restrictions in publication year. The selected reports were evaluated in terms of decellularization protocols, assessment of decellularized grafts, and evaluation of their biocompatibility and repopulation with cells both in vitro and in vivo. RESULTS The search resulted in a total of 88 articles. Each article was reviewed, 77 were excluded and the remaining 11 articles reported decellularization of 12 different vascular composite allografts in humans (four), large animals (three), and small animals (rodents) (five). The decellularization protocol for vascularized composite allotransplantation varies slightly between studies, but majority of the reports employ 1% sodium dodecyl sulfate as the main reagent for decellularization. The immunological response of the decellularized scaffolds remains poorly evaluated. Few authors have been able to attempt the recellularization and transplantation of these scaffolds. Successful transplantation seems to require prior recellularization. CONCLUSION Decellularization/recellularization is a promising, growing, emerging developing research field in vascular composite allotransplantation.
Collapse
Affiliation(s)
- Elise Lupon
- University Toulouse III Paul Sabatier, Department of Plastic Surgery, Toulouse, Occitanie, France.,Massachusetts General Hospital, Harvard Medical School, Vascularized Composite Allotransplantation Laboratory, Center for Transplantation Sciences, Boston, Massachusetts, United States.,Shriners Hospitals for Children Boston, 24172, Boston, Massachusetts, United States.,Massachusetts General Hospital, 2348, Division of Plastic and Reconstructive Surgery, Boston, Massachusetts, United States;
| | - Alexandre G Lellouch
- Massachusetts General Hospital, Harvard Medical School, Vascularized Composite Allotransplantation Laboratory, Center for Transplantation Sciences, Boston, Massachusetts, United States.,Shriners Hospitals for Children Boston, 24172, Boston, Massachusetts, United States.,Hospital European George Pompidou, 55647, Department of Plastic Surgery, Paris, Île-de-France, France.,Massachusetts General Hospital, 2348, Division of Plastic and Reconstructive Surgery, Boston, Massachusetts, United States;
| | - Aylin Acun
- Shriners Hospitals for Children Boston, 24172, Boston, Massachusetts, United States.,Massachusetts General Hospital, Harvard Medical School, Center for Engineering in Medicine and Surgery, Boston, Massachusetts, United States;
| | - Alec R Andrews
- Massachusetts General Hospital, Harvard Medical School, Vascularized Composite Allotransplantation Laboratory, Center for Transplantation Sciences, Boston, Massachusetts, United States.,Shriners Hospitals for Children Boston, 24172, Boston, Massachusetts, United States.,Massachusetts General Hospital, 2348, Division of Plastic and Reconstructive Surgery, Boston, Massachusetts, United States;
| | - Ruben Oganesyan
- Shriners Hospitals for Children Boston, 24172, Boston, Massachusetts, United States.,Massachusetts General Hospital, Harvard Medical School, Center for Engineering in Medicine and Surgery, Boston, Massachusetts, United States;
| | - Marion Goutard
- Massachusetts General Hospital, Harvard Medical School, Vascularized Composite Allotransplantation Laboratory, Center for Transplantation Sciences, Boston, Massachusetts, United States.,Shriners Hospitals for Children Boston, 24172, Boston, Massachusetts, United States.,Hospital European George Pompidou, 55647, Department of Plastic Surgery, Paris, Île-de-France, France.,Massachusetts General Hospital, 2348, Division of Plastic and Reconstructive Surgery, Boston, Massachusetts, United States;
| | - Corentin B Taveau
- Hospital European George Pompidou, 55647, Department of Plastic Surgery, Paris, Île-de-France, France;
| | - Laurent A Lantieri
- Hospital European George Pompidou, 55647, Department of Plastic Surgery, Paris, Île-de-France, France;
| | - Curtis L Cetrulo
- Massachusetts General Hospital, Harvard Medical School, Vascularized Composite Allotransplantation Laboratory, Center for Transplantation Sciences, Boston, Massachusetts, United States.,Shriners Hospitals for Children Boston, 24172, Boston, Massachusetts, United States.,Massachusetts General Hospital, 2348, Division of Plastic and Reconstructive Surgery, Boston, Massachusetts, United States;
| | - Basak E Uygun
- Shriners Hospitals for Children Boston, 24172, Boston, Massachusetts, United States.,Massachusetts General Hospital, Harvard Medical School, Center for Engineering in Medicine and Surgery, Boston, Massachusetts, United States;
| |
Collapse
|
17
|
Mansouri M, Leipzig ND. Advances in removing mass transport limitations for more physiologically relevant in vitro 3D cell constructs. BIOPHYSICS REVIEWS 2021; 2:021305. [PMID: 38505119 PMCID: PMC10903443 DOI: 10.1063/5.0048837] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/31/2021] [Indexed: 03/21/2024]
Abstract
Spheroids and organoids are promising models for biomedical applications ranging from human disease modeling to drug discovery. A main goal of these 3D cell-based platforms is to recapitulate important physiological parameters of their in vivo organ counterparts. One way to achieve improved biomimetic architectures and functions is to culture cells at higher density and larger total numbers. However, poor nutrient and waste transport lead to low stability, survival, and functionality over extended periods of time, presenting outstanding challenges in this field. Fortunately, important improvements in culture strategies have enhanced the survival and function of cells within engineered microtissues/organs. Here, we first discuss the challenges of growing large spheroids/organoids with a focus on mass transport limitations, then highlight recent tools and methodologies that are available for producing and sustaining functional 3D in vitro models. This information points toward the fact that there is a critical need for the continued development of novel cell culture strategies that address mass transport in a physiologically relevant human setting to generate long-lasting and large-sized spheroids/organoids.
Collapse
Affiliation(s)
- Mona Mansouri
- Department of Chemical, Biomolecular, and Corrosion Engineering, University of Akron, Akron, Ohio 44325, USA
| | - Nic D. Leipzig
- Department of Chemical, Biomolecular, and Corrosion Engineering, University of Akron, Akron, Ohio 44325, USA
| |
Collapse
|
18
|
Hwang DG, Choi YM, Jang J. 3D Bioprinting-Based Vascularized Tissue Models Mimicking Tissue-Specific Architecture and Pathophysiology for in vitro Studies. Front Bioeng Biotechnol 2021; 9:685507. [PMID: 34136473 PMCID: PMC8201787 DOI: 10.3389/fbioe.2021.685507] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 04/30/2021] [Indexed: 12/11/2022] Open
Abstract
A wide variety of experimental models including 2D cell cultures, model organisms, and 3D in vitro models have been developed to understand pathophysiological phenomena and assess the safety and efficacy of potential therapeutics. In this sense, 3D in vitro models are an intermediate between 2D cell cultures and animal models, as they adequately reproduce 3D microenvironments and human physiology while also being controllable and reproducible. Particularly, recent advances in 3D in vitro biomimicry models, which can produce complex cell structures, shapes, and arrangements, can more similarly reflect in vivo conditions than 2D cell culture. Based on this, 3D bioprinting technology, which enables to place the desired materials in the desired locations, has been introduced to fabricate tissue models with high structural similarity to the native tissues. Therefore, this review discusses the recent developments in this field and the key features of various types of 3D-bioprinted tissues, particularly those associated with blood vessels or highly vascularized organs, such as the heart, liver, and kidney. Moreover, this review also summarizes the current state of the three categories: (1) chemical substance treatment, (2) 3D bioprinting of lesions, and (3) recapitulation of tumor microenvironments (TME) of 3D bioprinting-based disease models according to their disease modeling approach. Finally, we propose the future directions of 3D bioprinting approaches for the creation of more advanced in vitro biomimetic 3D tissues, as well as the translation of 3D bioprinted tissue models to clinical applications.
Collapse
Affiliation(s)
- Dong Gyu Hwang
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, South Korea
| | - Yoo-Mi Choi
- Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang, South Korea
| | - Jinah Jang
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, South Korea.,Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang, South Korea.,Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, South Korea.,Institute of Convergence Science, Yonsei University, Seoul, South Korea
| |
Collapse
|
19
|
Alghutaimel H, Yang X, Drummond B, Nazzal H, Duggal M, Raïf E. Investigating the vascularization capacity of a decellularized dental pulp matrix seeded with human dental pulp stem cells: in vitro and preliminary in vivo evaluations. Int Endod J 2021; 54:1300-1316. [PMID: 33709438 DOI: 10.1111/iej.13510] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 12/17/2022]
Abstract
AIM To investigate the vascularization capacity of a decellularized dental pulp matrix (DDP) of bovine origin seeded with human dental pulp stem cells (hDPSCs) in vitro and to present preliminary in vivo findings. METHODOLOGY Bovine dental pulps were decellularized and then analysed using histological staining and DNA quantification. The resultant DDPs were characterized using immunohistochemical staining for the retention of vascular endothelial growth factor A (VEGF-A) and fibroblast growth factor 2 (FGF-2). Furthermore, DDPs were recellularized with hDPSCs and analysed histologically. The expression of markers involved in angiogenesis by hDPSCs colonizing the DDPs was assessed in vitro. A preliminary in vivo study was then conducted in which hDPSCs-seeded and unseeded DDPs were inserted in debrided human premolars root slices and implanted subcutaneously in immunodeficient mice. Samples were retrieved after 30 days and analysed using histological and immunohistochemical staining. The independent samples t-test, analysis of variance and a Kruskal-Wallis test were used to analyse the quantitative data statistically depending on the group numbers and normality of data distribution. The difference between the groups was considered significant when the P-value was less than 0.05. RESULTS Acellular dental pulp matrices were generated following bovine dental pulp decellularization. Evaluation of the developed DDPs revealed a significant DNA reduction (P < 0.0001) with preservation of the native histoarchitecture and vasculature and retention of VEGF-A and FGF-2. Upon recellularization of the DDPs with hDPSCs, the in vitro analyses revealed cell engraftment with progressive repopulation of DDPs' matrices and vasculature and with enhanced expression of markers involved in angiogenesis. In vivo implantation of root slices with hDPSCs-seeded DDPs revealed apparent vascularization enhancement as compared to the unseeded DDP group (P < 0.0001). CONCLUSIONS The developed decellularized dental pulp matrix had pro-angiogenic properties characterized by the retention of native vasculature and angiogenic growth factors. Seeding of hDPSCs into the DDP led to progressive repopulation of the vasculature, enhanced expression of markers involved in angiogenesis in hDPSCs and improved in vivo vascularization capacity. The se suggest that a combination of DDP and hDPSCs have the potential to provide a promising vascularization promoting strategy for dental pulp regeneration.
Collapse
Affiliation(s)
- H Alghutaimel
- Department of Paediatric Dentistry, School of Dentistry, University of Leeds, Leeds, UK.,Department of Paediatric Dentistry, School of Dentistry, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - X Yang
- Department of Oral Biology, School of Dentistry, University of Leeds, Leeds, UK
| | - B Drummond
- Department of Paediatric Dentistry, School of Dentistry, University of Leeds, Leeds, UK
| | - H Nazzal
- Paediatric Dentistry Section, Hamad Dental Centre, Hamad Medical Corporation, Doha, Qatar
| | - M Duggal
- College of Dental Medicine, QU Health, Qatar University, Doha, Qatar
| | - E Raïf
- Department of Oral Biology, School of Dentistry, University of Leeds, Leeds, UK
| |
Collapse
|
20
|
Moriscot A, Miyabara EH, Langeani B, Belli A, Egginton S, Bowen TS. Firearms-related skeletal muscle trauma: pathophysiology and novel approaches for regeneration. NPJ Regen Med 2021; 6:17. [PMID: 33772028 PMCID: PMC7997931 DOI: 10.1038/s41536-021-00127-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
One major cause of traumatic injury is firearm-related wounds (i.e., ballistic trauma), common in both civilian and military populations, which is increasing in prevalence and has serious long-term health and socioeconomic consequences worldwide. Common primary injuries of ballistic trauma include soft-tissue damage and loss, haemorrhage, bone fracture, and pain. The majority of injuries are of musculoskeletal origin and located in the extremities, such that skeletal muscle offers a major therapeutic target to aid recovery and return to normal daily activities. However, the underlying pathophysiology of skeletal muscle ballistic trauma remains poorly understood, with limited evidence-based treatment options. As such, this review will address the topic of firearm-related skeletal muscle injury and regeneration. We first introduce trauma ballistics and the immediate injury of skeletal muscle, followed by detailed coverage of the underlying biological mechanisms involved in regulating skeletal muscle dysfunction following injury, with a specific focus on the processes of muscle regeneration, muscle wasting and vascular impairments. Finally, we evaluate novel approaches for minimising muscle damage and enhancing muscle regeneration after ballistic trauma, which may have important relevance for primary care in victims of violence.
Collapse
Affiliation(s)
- Anselmo Moriscot
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Elen H Miyabara
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Antonio Belli
- NIHR Surgical Reconstruction and Microbiology Research Centre, University of Birmingham, Birmingham, UK
| | - Stuart Egginton
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - T Scott Bowen
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK.
| |
Collapse
|
21
|
Asadi M, Khalili M, Lotfi H, Vaghefi Moghaddam S, Zarghami N, André H, Alizadeh E. Liver bioengineering: Recent trends/advances in decellularization and cell sheet technologies towards translation into the clinic. Life Sci 2021; 276:119373. [PMID: 33744324 DOI: 10.1016/j.lfs.2021.119373] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/03/2021] [Accepted: 03/08/2021] [Indexed: 02/07/2023]
Abstract
Development of novel technologies provides the best tissue constructs engineering and maximizes their therapeutic effects in regenerative therapy, especially for liver dysfunctions. Among the currently investigated approaches of tissue engineering, scaffold-based and scaffold-free tissues are widely suggested for liver regeneration. Analogs of liver acellular extracellular matrix (ECM) are utilized in native scaffolds to increase the self-repair and healing ability of organs. Native ECM analog could improve liver repairing through providing the supportive framework for cells and signaling molecules, exerting normal biomechanical, biochemical, and physiological signal complexes. Recently, innovative cell sheet technology is introduced as an alternative for conventional tissue engineering with the advantage of fewer scaffold restrictions and cell culture on a Thermo-Responsive Polymer Surface. These sheets release the layered cells through a temperature-controlled procedure without enzymatic digestion, while preserving the cell-ECM contacts and adhesive molecules on cell-cell junctions. In addition, several novelties have been introduced into the cell sheet and decellularization technologies to aid cell growth, instruct differentiation/angiogenesis, and promote cell migration. In this review, recent trends, advancements, and issues linked to translation into clinical practice are dissected and compared regarding the decellularization and cell sheet technologies for liver tissue engineering.
Collapse
Affiliation(s)
- Maryam Asadi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mostafa Khalili
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajie Lotfi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Physiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Nosratollah Zarghami
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Helder André
- Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institute, 11282 Stockholm, Sweden
| | - Effat Alizadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
22
|
Morrissey J, Mesquita FCP, Hochman-Mendez C, Taylor DA. Whole Heart Engineering: Advances and Challenges. Cells Tissues Organs 2021; 211:395-405. [PMID: 33640893 DOI: 10.1159/000511382] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/26/2020] [Indexed: 11/19/2022] Open
Abstract
Bioengineering a solid organ for organ replacement is a growing endeavor in regenerative medicine. Our approach - recellularization of a decellularized cadaveric organ scaffold with human cells - is currently the most promising approach to building a complex solid vascularized organ to be utilized in vivo, which remains the major unmet need and a key challenge. The 2008 publication of perfusion-based decellularization and partial recellularization of a rat heart revolutionized the tissue engineering field by showing that it was feasible to rebuild an organ using a decellularized extracellular matrix scaffold. Toward the goal of clinical translation of bioengineered tissues and organs, there is increasing recognition of the underlying need to better integrate basic science domains and industry. From the perspective of a research group focusing on whole heart engineering, we discuss the current approaches and advances in whole organ engineering research as they relate to this multidisciplinary field's 3 major pillars: organ scaffolds, large numbers of cells, and biomimetic bioreactor systems. The success of whole organ engineering will require optimization of protocols to produce biologically-active scaffolds for multiple organ systems, and further technological innovation both to produce the massive quantities of high-quality cells needed for recellularization and to engineer a bioreactor with physiologic stimuli to recapitulate organ function. Also discussed are the challenges to building an implantable vascularized solid organ.
Collapse
Affiliation(s)
- Jacquelynn Morrissey
- Regenerative Medicine Research Department, Texas Heart Institute, Houston, Texas, USA
| | - Fernanda C P Mesquita
- Regenerative Medicine Research Department, Texas Heart Institute, Houston, Texas, USA
| | - Camila Hochman-Mendez
- Regenerative Medicine Research Department, Texas Heart Institute, Houston, Texas, USA
| | | |
Collapse
|
23
|
Sant S, Wang D, Abidi M, Walker G, Ferrell N. Mechanical characterization of native and sugar-modified decellularized kidneys. J Mech Behav Biomed Mater 2021; 114:104220. [PMID: 33257205 PMCID: PMC7855467 DOI: 10.1016/j.jmbbm.2020.104220] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/28/2020] [Accepted: 11/20/2020] [Indexed: 10/22/2022]
Abstract
Decellularized organs have the potential to be used as scaffolds for tissue engineering organ replacements. The mechanical properties of the extracellular matrix (ECM) following decellularization are critical for structural integrity and for regulation of cell function upon recellularization. Advanced glycation end products (AGEs) accumulate in the ECM with age and their formation is accelerated by several pathological conditions including diabetes. Some AGEs span multiple amino acids to form crosslinks that may alter the mechanical properties of the ECM. The goal of this work was to evaluate how sugar-induced modifications to the ECM affect the mechanical behavior of decellularized kidney. The compressive and tensile properties of the kidney ECM were evaluated using an accelerated model of AGE formation by ribose. Results show that ribose modifications significantly alter the mechanical behavior of decellularized kidney. Increased resistance to deformation corresponds to increased ECM crosslinking, and mechanical changes can be partially mitigated by AGE inhibition. The degree of post-translational modification of the ECM is dependent on the age and health of the organ donor and may play a role in regulating the mechanical properties of decellularized organs.
Collapse
Affiliation(s)
- Snehal Sant
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, United States
| | - Dan Wang
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, United States
| | - Minhal Abidi
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, United States
| | - Gwyneth Walker
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, United States
| | - Nicholas Ferrell
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, United States; Department of Biomedical Engineering, Vanderbilt University, United States; Vanderbilt Center for Kidney Disease, United States.
| |
Collapse
|
24
|
A Perfusion Bioreactor for Longitudinal Monitoring of Bioengineered Liver Constructs. NANOMATERIALS 2021; 11:nano11020275. [PMID: 33494337 PMCID: PMC7912543 DOI: 10.3390/nano11020275] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/11/2021] [Accepted: 01/19/2021] [Indexed: 02/06/2023]
Abstract
In the field of in vitro liver disease models, decellularised organ scaffolds maintain the original biomechanical and biological properties of the extracellular matrix and are established supports for in vitro cell culture. However, tissue engineering approaches based on whole organ decellularized scaffolds are hampered by the scarcity of appropriate bioreactors that provide controlled 3D culture conditions. Novel specific bioreactors are needed to support long-term culture of bioengineered constructs allowing non-invasive longitudinal monitoring. Here, we designed and validated a specific bioreactor for long-term 3D culture of whole liver constructs. Whole liver scaffolds were generated by perfusion decellularisation of rat livers. Scaffolds were seeded with Luc+HepG2 and primary human hepatocytes and cultured in static or dynamic conditions using the custom-made bioreactor. The bioreactor included a syringe pump, for continuous unidirectional flow, and a circuit built to allow non-invasive monitoring of culture parameters and media sampling. The bioreactor allowed non-invasive analysis of cell viability, distribution, and function of Luc+HepG2-bioengineered livers cultured for up to 11 days. Constructs cultured in dynamic conditions in the bioreactor showed significantly higher cell viability, measured with bioluminescence, distribution, and functionality (determined by albumin production and expression of CYP enzymes) in comparison to static culture conditions. Finally, our bioreactor supports primary human hepatocyte viability and function for up to 30 days, when seeded in the whole liver scaffolds. Overall, our novel bioreactor is capable of supporting cell survival and metabolism and is suitable for liver tissue engineering for the development of 3D liver disease models.
Collapse
|
25
|
Meijer EM, van Dijk CGM, Kramann R, Verhaar MC, Cheng C. Implementation of Pericytes in Vascular Regeneration Strategies. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:1-21. [PMID: 33231500 DOI: 10.1089/ten.teb.2020.0229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
For the survival and integration of complex large-sized tissue-engineered (TE) organ constructs that exceed the maximal nutrients and oxygen diffusion distance required for cell survival, graft (pre)vascularization to ensure medium or blood supply is crucial. To achieve this, the morphology and functionality of the microcapillary bed should be mimicked by incorporating vascular cell populations, including endothelium and mural cells. Pericytes play a crucial role in microvascular function, blood vessel stability, angiogenesis, and blood pressure regulation. In addition, tissue-specific pericytes are important in maintaining specific functions in different organs, including vitamin A storage in the liver, renin production in the kidneys and maintenance of the blood-brain-barrier. Together with their multipotential differentiation capacity, this makes pericytes the preferred cell type for application in TE grafts. The use of a tissue-specific pericyte cell population that matches the TE organ may benefit organ function. In this review, we provide an overview of the literature for graft (pre)-vascularization strategies and highlight the possible advantages of using tissue-specific pericytes for specific TE organ grafts. Impact statement The use of a tissue-specific pericyte cell population that matches the tissue-engineered (TE) organ may benefit organ function. In this review, we provide an overview of the literature for graft (pre)vascularization strategies and highlight the possible advantages of using tissue-specific pericytes for specific TE organ grafts.
Collapse
Affiliation(s)
- Elana M Meijer
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Christian G M van Dijk
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rafael Kramann
- Division of Nephrology and Institute of Experimental Medicine and Systems Biology, University Hospital RWTH Aachen, Aachen, Germany.,Department of Internal Medicine, Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Caroline Cheng
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands.,Experimental Cardiology, Department of Cardiology, Thorax Center Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
26
|
Campo H, López-Martínez S, Cervelló I. Decellularization Methods of Uterus in Tissue Engineering. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1345:141-152. [PMID: 34582020 DOI: 10.1007/978-3-030-82735-9_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
A new field of investigation which aims to design tissues and organs similar to their native origin has been developed recently, named as regenerative medicine (tissue engineering and bio-engineering). Uterus is the main organ for regeneration and contributes in the fertility. At an ultimate level, the uterus plays a role in embryo implantation, sperm migration and fetal nutrition. Uterine congenital anomalies, attained uterine lesions and immune system disorders may affect such uterine functions preventing successful pregnancy. Due to following reasons, it is essential to consider regenerative medicine as a new approach for the treatment of uterine dysfunctions to overcome the failures that cannot be treated with clinical medication.
Collapse
Affiliation(s)
- Hannes Campo
- Fundación Instituto Valenciano de Infertilidad (FIVI), Instituto Universitario IVI/INCLIVA, Valencia, Spain
| | - Sara López-Martínez
- Fundación Instituto Valenciano de Infertilidad (FIVI), Instituto Universitario IVI/INCLIVA, Valencia, Spain
| | - Irene Cervelló
- Fundación Instituto Valenciano de Infertilidad (FIVI), Instituto Universitario IVI/INCLIVA, Valencia, Spain.
| |
Collapse
|
27
|
Composable microfluidic spinning platforms for facile production of biomimetic perfusable hydrogel microtubes. Nat Protoc 2020; 16:937-964. [PMID: 33318693 DOI: 10.1038/s41596-020-00442-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023]
Abstract
Microtissues with specific structures and integrated vessels play a key role in maintaining organ functions. To recapitulate the in vivo environment for tissue engineering and organ-on-a-chip purposes, it is essential to develop perfusable biomimetic microscaffolds. We developed facile all-aqueous microfluidic approaches for producing perfusable hydrogel microtubes with diverse biomimetic sizes and shapes. Here, we provide a detailed protocol describing the construction of the microtube spinning platforms, the assembly of microfluidic devices, and the fabrication and characterization of various perfusable hydrogel microtubes. The hydrogel microtubes can be continuously generated from microfluidic devices due to the crosslinking of alginate by calcium in the coaxial flows and collecting bath. Owing to the mild all-aqueous spinning process, cells can be loaded into the alginate prepolymer for microtube spinning, which enables the direct production of cell-laden hydrogel microtubes. By manipulating the fluid dynamics at the microscale, the composable microfluidic devices and platforms can be used for the facile generation of six types of biomimetic perfusable microtubes. The microfluidic platforms and devices can be set up within 3 h from commonly available and inexpensive materials. After 10-20 min required to adjust the platform and fluids, perfusable hydrogel microtubes can be generated continuously. We describe how to characterize the microtubes using scanning electron or confocal microscopy. As an example application, we describe how the microtubes can be used for the preparation of a vascular lumen and how to perform barrier permeability tests of the vascular lumen.
Collapse
|
28
|
Current and Future Perspectives of the Use of Organoids in Radiobiology. Cells 2020; 9:cells9122649. [PMID: 33317153 PMCID: PMC7764598 DOI: 10.3390/cells9122649] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
The majority of cancer patients will be treated with radiotherapy, either alone or together with chemotherapy and/or surgery. Optimising the balance between tumour control and the probability of normal tissue side effects is the primary goal of radiation treatment. Therefore, it is imperative to understand the effects that irradiation will have on both normal and cancer tissue. The more classical lab models of immortal cell lines and in vivo animal models have been fundamental to radiobiological studies to date. However, each of these comes with their own limitations and new complementary models are required to fill the gaps left by these traditional models. In this review, we discuss how organoids, three-dimensional tissue-resembling structures derived from tissue-resident, embryonic or induced pluripotent stem cells, overcome the limitations of these models and thus have a growing importance in the field of radiation biology research. The roles of organoids in understanding radiation-induced tissue responses and in moving towards precision medicine are examined. Finally, the limitations of organoids in radiobiology and the steps being made to overcome these limitations are considered.
Collapse
|
29
|
Shah Mohammadi M, Buchen JT, Pasquina PF, Niklason LE, Alvarez LM, Jariwala SH. Critical Considerations for Regeneration of Vascularized Composite Tissues. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:366-381. [PMID: 33115331 DOI: 10.1089/ten.teb.2020.0223] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Effective vascularization is vital for survival and functionality of complex tissue-engineered organs. The formation of the microvasculature, composed of endothelial cells (ECs) alone, has been mostly used to restore the vascular networks in organs. However, recent heterocellular studies demonstrate that co-culturing is a more effective approach in revascularization of engineered organs. This review presents key considerations for manufacturing of artificial vascularized composite tissues. We summarize the importance of co-cultures and the multicellular interactions with ECs, as well as design and use of bioreactors, as critical considerations for tissue vascularization. In addition, as an emerging scaffolding technique, this review also highlights the current caveats and hurdles associated with three-dimensional bioprinting and discusses recent developments in bioprinting strategies such as four-dimensional bioprinting and its future outlook for manufacturing of vascularized tissue constructs. Finally, the review concludes with addressing the critical challenges in the regulatory pathway and clinical translation of artificial composite tissue grafts. Impact statement Regeneration of composite tissues is critical as biophysical and biochemical characteristics differ between various types of tissues. Engineering a vascularized composite tissue has remained unresolved and requires additional evaluations along with optimization of methodologies and standard operating procedures. To this end, the main hurdle is creating a viable vascular endothelium that remains functional for a longer duration postimplantation, and can be manufactured using clinically appropriate source of cell lines that are scalable in vitro for the fabrication of human-scale organs. This review presents key considerations for regeneration and manufacturing of vascularized composite tissues as the field advances.
Collapse
Affiliation(s)
- Maziar Shah Mohammadi
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| | - Jack T Buchen
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| | - Paul F Pasquina
- Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA.,Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Laura E Niklason
- Department of Anesthesia and Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Luis M Alvarez
- Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA.,Lung Biotechnology PBC, Silver Spring, Maryland, USA
| | - Shailly H Jariwala
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
30
|
Palikuqi B, Nguyen DHT, Li G, Schreiner R, Pellegata AF, Liu Y, Redmond D, Geng F, Lin Y, Gómez-Salinero JM, Yokoyama M, Zumbo P, Zhang T, Kunar B, Witherspoon M, Han T, Tedeschi AM, Scottoni F, Lipkin SM, Dow L, Elemento O, Xiang JZ, Shido K, Spence JR, Zhou QJ, Schwartz RE, De Coppi P, Rabbany SY, Rafii S. Adaptable haemodynamic endothelial cells for organogenesis and tumorigenesis. Nature 2020; 585:426-432. [PMID: 32908310 PMCID: PMC7480005 DOI: 10.1038/s41586-020-2712-z] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 06/08/2020] [Indexed: 12/12/2022]
Abstract
Endothelial cells adopt tissue-specific characteristics to instruct organ development and regeneration1,2. This adaptability is lost in cultured adult endothelial cells, which do not vascularize tissues in an organotypic manner. Here, we show that transient reactivation of the embryonic-restricted ETS variant transcription factor 2 (ETV2)3 in mature human endothelial cells cultured in a serum-free three-dimensional matrix composed of a mixture of laminin, entactin and type-IV collagen (LEC matrix) ‘resets’ these endothelial cells to adaptable, vasculogenic cells, which form perfusable and plastic vascular plexi. Through chromatin remodelling, ETV2 induces tubulogenic pathways, including the activation of RAP1, which promotes the formation of durable lumens4,5. In three-dimensional matrices—which do not have the constraints of bioprinted scaffolds—the ‘reset’ vascular endothelial cells (R-VECs) self-assemble into stable, multilayered and branching vascular networks within scalable microfluidic chambers, which are capable of transporting human blood. In vivo, R-VECs implanted subcutaneously in mice self-organize into durable pericyte-coated vessels that functionally anastomose to the host circulation and exhibit long-lasting patterning, with no evidence of malformations or angiomas. R-VECs directly interact with cells within three-dimensional co-cultured organoids, removing the need for the restrictive synthetic semipermeable membranes that are required for organ-on-chip systems, therefore providing a physiological platform for vascularization, which we call ‘Organ-On-VascularNet’. R-VECs enable perfusion of glucose-responsive insulin-secreting human pancreatic islets, vascularize decellularized rat intestines and arborize healthy or cancerous human colon organoids. Using single-cell RNA sequencing and epigenetic profiling, we demonstrate that R-VECs establish an adaptive vascular niche that differentially adjusts and conforms to organoids and tumoroids in a tissue-specific manner. Our Organ-On-VascularNet model will permit metabolic, immunological and physiochemical studies and screens to decipher the crosstalk between organotypic endothelial cells and parenchymal cells for identification of determinants of endothelial cell heterogeneity, and could lead to advances in therapeutic organ repair and tumour targeting. The transient reactivation of ETV2 in adult human endothelial cells reprograms these cells to become adaptable vasculogenic endothelia that in three-dimensional matrices self-assemble into vascular networks that can transport blood and physiologically arborize organoids and decellularized tissues.
Collapse
Affiliation(s)
- Brisa Palikuqi
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Duc-Huy T Nguyen
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ge Li
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ryan Schreiner
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.,Department of Ophthalmology, Margaret Dyson Vision Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Alessandro F Pellegata
- Stem Cell and Regenerative Medicine Section, DBC Programme, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Ying Liu
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - David Redmond
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Fuqiang Geng
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Yang Lin
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jesus M Gómez-Salinero
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Masataka Yokoyama
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Paul Zumbo
- Applied Bioinformatics Core, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Tuo Zhang
- Genomics Resources Core Facility, Weill Cornell Medicine, New York, NY, USA
| | - Balvir Kunar
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Mavee Witherspoon
- Sandra and Edward Meyer Cancer Center, Weill Cornell Graduate School of Medical Sciences, Departments of Biochemistry and Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Teng Han
- Sandra and Edward Meyer Cancer Center, Weill Cornell Graduate School of Medical Sciences, Departments of Biochemistry and Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Alfonso M Tedeschi
- Stem Cell and Regenerative Medicine Section, DBC Programme, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Federico Scottoni
- Stem Cell and Regenerative Medicine Section, DBC Programme, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Steven M Lipkin
- Sandra and Edward Meyer Cancer Center, Weill Cornell Graduate School of Medical Sciences, Departments of Biochemistry and Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lukas Dow
- Sandra and Edward Meyer Cancer Center, Weill Cornell Graduate School of Medical Sciences, Departments of Biochemistry and Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Jenny Z Xiang
- Genomics Resources Core Facility, Weill Cornell Medicine, New York, NY, USA
| | - Koji Shido
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jason R Spence
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Qiao J Zhou
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Robert E Schwartz
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.,Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Paolo De Coppi
- Stem Cell and Regenerative Medicine Section, DBC Programme, Great Ormond Street Institute of Child Health, University College London, London, UK.,Specialist Neonatal and Paediatric Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Sina Y Rabbany
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.,Bioengineering Program, DeMatteis School of Engineering and Applied Science, Hofstra University, Hempstead, NY, USA
| | - Shahin Rafii
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
31
|
Wright ME, Yu JK, Jain D, Maeda A, Yeh SCA, DaCosta RS, Lin CP, Santerre JP. Engineering functional microvessels in synthetic polyurethane random-pore scaffolds by harnessing perfusion flow. Biomaterials 2020; 256:120183. [PMID: 32622017 DOI: 10.1016/j.biomaterials.2020.120183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/05/2020] [Accepted: 06/06/2020] [Indexed: 12/24/2022]
Abstract
Recently reported biomaterial-based approaches toward prevascularizing tissue constructs rely on biologically or structurally complex scaffolds that are complicated to manufacture and sterilize, and challenging to customize for clinical applications. In the current work, a prevascularization method for soft tissue engineering that uses a non-patterned and non-biological scaffold is proposed. Human fibroblasts and HUVECs were seeded on an ionomeric polyurethane-based hydrogel and cultured for 14 days under medium perfusion. A flow rate of 0.05 mL/min resulted in a greater lumen density in the constructs relative to 0.005 and 0.5 mL/min, indicating the critical importance of flow magnitude in establishing microvessels. Constructs generated at 0.05 mL/min perfusion flow were implanted in a mouse subcutaneous model and intravital imaging was used to characterize host blood perfusion through the construct after 2 weeks. Engineered microvessels were functional (i.e. perfused with host blood and non-leaky) and neovascularization of the construct by host vessels was enhanced relative to non-prevascularized constructs. We report on the first strategy toward engineering functional microvessels in a tissue construct using non-bioactive, non-patterned synthetic polyurethane materials.
Collapse
Affiliation(s)
- Meghan Ee Wright
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Jonathan K Yu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada; Faculty of Dentistry, University of Toronto, Toronto, Canada
| | - Devika Jain
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Azusa Maeda
- Princess Margaret Cancer Centre and Techna Institute, University Health Network, Toronto, Canada
| | - Shu-Chi A Yeh
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ralph S DaCosta
- Princess Margaret Cancer Centre and Techna Institute, University Health Network, Toronto, Canada
| | - Charles P Lin
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - J Paul Santerre
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada; Faculty of Dentistry, University of Toronto, Toronto, Canada.
| |
Collapse
|
32
|
Fabrication of a Microfluidic System Using Micromolded Alginate Gel as a Sacrificial Material for Tissues Engineering. J CHEM-NY 2020. [DOI: 10.1155/2020/3148652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
We described a sacrificial molding for the formation of microfluidic networks. In this molding, the micromolded calcium alginate (Ca-Alg) is introduced as a sacrificial template. The basis of this procedure is fabricating a micromolded Ca-Alg hydrogel and encapsulating this model within a second gel and removing it by ion-exchange to leave a microchannel in the remaining gel. This microfluidic system can readily deliver solutes into the channels and even control the transport of solutes from channels into the bulk of the gels. Furthermore, the perfused vascular channels can sustain the metabolic activity of encapsulated cells, indicating the feasibility of this microfluidic system in the field of tissue engineering.
Collapse
|
33
|
Allotransplantation of adult spinal cord tissues after complete transected spinal cord injury: Long-term survival and functional recovery in canines. SCIENCE CHINA-LIFE SCIENCES 2020; 63:1879-1886. [PMID: 32382980 DOI: 10.1007/s11427-019-1623-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 12/05/2019] [Indexed: 12/15/2022]
Abstract
Spinal cord injury (SCI), especially complete transected SCI, leads to loss of cells and extracellular matrix and functional impairments. In a previous study, we transplanted adult spinal cord tissues (aSCTs) to replace lost tissues and facilitate recovery in a rat SCI model. However, rodents display considerable differences from human patients in the scale, anatomy and functions of spinal cord systems, and responses after injury. Thus, use of a large animal SCI model is required to examine the repair efficiency of potential therapeutic approaches. In this study, we transplanted allogenic aSCTs from adult dogs to the lesion area of canines after complete transection of the thoracic spinal cord, and investigated the long-term cell survival and functional recovery. To enhance repair efficiency, a growth factor cocktail was added during aSCT transplantation, providing a favorable microenvironment. The results showed that transplantation of aSCTs, in particular with the addition of growth factors, significantly improves locomotor function restoration and increases the number of neurofilament-, microtubule-associated protein 2-, 5-hydroxytryptamine-, choline acetyltransferase- and tyrosine hydroxylase-positive neurons in the lesion area at 6 months post-surgery. In addition, we demonstrated that donor neurons in aSCTs can survive for a long period after transplantation. This study showed for the first time that transplanting aSCTs combined with growth factor supplementation facilitates reconstruction of injured spinal cords, and consequently promotes long lasting motor function recovery in a large animal complete transected SCI model, and therefore could be considered as a possible therapeutic strategy in humans.
Collapse
|
34
|
Moser PT, Gerli M, Diercks GR, Evangelista-Leite D, Charest JM, Gershlak JR, Ren X, Gilpin SE, Jank BJ, Gaudette GR, Hartnick CJ, Ott HC. Creation of Laryngeal Grafts from Primary Human Cells and Decellularized Laryngeal Scaffolds. Tissue Eng Part A 2020; 26:543-555. [DOI: 10.1089/ten.tea.2019.0128] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Philipp T. Moser
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Mattia Gerli
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Great Ormond Street Institute of Child Health, University College London Medical School, London, United Kingdom
| | - Gillian R. Diercks
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
| | | | - Jonathan M. Charest
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Joshua R. Gershlak
- Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts
| | - Xi Ren
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| | - Sarah E. Gilpin
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Bernhard J. Jank
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Otolaryngology, Medical University of Vienna, Vienna, Austria
| | - Glenn R. Gaudette
- Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts
| | - Christopher J. Hartnick
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
| | - Harald C. Ott
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Thoracic Surgery, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
35
|
Dorrello NV, Vunjak-Novakovic G. Bioengineering of Pulmonary Epithelium With Preservation of the Vascular Niche. Front Bioeng Biotechnol 2020; 8:269. [PMID: 32351946 PMCID: PMC7174601 DOI: 10.3389/fbioe.2020.00269] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 03/16/2020] [Indexed: 12/20/2022] Open
Abstract
The shortage of transplantable donor organs directly affects patients with end-stage lung disease, for which transplantation remains the only definitive treatment. With the current acceptance rate of donor lungs of only 20%, rescuing even one half of the rejected donor lungs would increase the number of transplantable lungs threefold, to 60%. We review recent advances in lung bioengineering that have potential to repair the epithelial and vascular compartments of the lung. Our focus is on the long-term support and recovery of the lung ex vivo, and the replacement of defective epithelium with healthy therapeutic cells. To this end, we first review the roles of the lung epithelium and vasculature, with focus on the alveolar-capillary membrane, and then discuss the available and emerging technologies for ex vivo bioengineering of the lung by decellularization and recellularization. While there have been many meritorious advances in these technologies for recovering marginal quality lungs to the levels needed to meet the standards for transplantation – many challenges remain, motivating further studies of the extended ex vivo support and interventions in the lung. We propose that the repair of injured epithelium with preservation of quiescent vasculature will be critical for the immediate blood supply to the lung and the lung survival and function following transplantation.
Collapse
Affiliation(s)
- N Valerio Dorrello
- Department of Pediatrics, Columbia University, New York, NY, United States
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY, United States.,Department of Medicine, Columbia University, New York, NY, United States
| |
Collapse
|
36
|
Tsuchiya T, Doi R, Obata T, Hatachi G, Nagayasu T. Lung Microvascular Niche, Repair, and Engineering. Front Bioeng Biotechnol 2020; 8:105. [PMID: 32154234 PMCID: PMC7047880 DOI: 10.3389/fbioe.2020.00105] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 02/03/2020] [Indexed: 12/28/2022] Open
Abstract
Biomaterials have been used for a long time in the field of medicine. Since the success of "tissue engineering" pioneered by Langer and Vacanti in 1993, tissue engineering studies have advanced from simple tissue generation to whole organ generation with three-dimensional reconstruction. Decellularized scaffolds have been widely used in the field of reconstructive surgery because the tissues used to generate decellularized scaffolds can be easily harvested from animals or humans. When a patient's own cells can be seeded onto decellularized biomaterials, theoretically this will create immunocompatible organs generated from allo- or xeno-organs. The most important aspect of lung tissue engineering is that the delicate three-dimensional structure of the organ is maintained during the tissue engineering process. Therefore, organ decellularization has special advantages for lung tissue engineering where it is essential to maintain the extremely thin basement membrane in the alveoli. Since 2010, there have been many methodological developments in the decellularization and recellularization of lung scaffolds, which includes improvements in the decellularization protocols and the selection and preparation of seeding cells. However, early transplanted engineered lungs terminated in organ failure in a short period. Immature vasculature reconstruction is considered to be the main cause of engineered organ failure. Immature vasculature causes thrombus formation in the engineered lung. Successful reconstruction of a mature vasculature network would be a major breakthrough in achieving success in lung engineering. In order to regenerate the mature vasculature network, we need to remodel the vascular niche, especially the microvasculature, in the organ scaffold. This review highlights the reconstruction of the vascular niche in a decellularized lung scaffold. Because the vascular niche consists of endothelial cells (ECs), pericytes, extracellular matrix (ECM), and the epithelial-endothelial interface, all of which might affect the vascular tight junction (TJ), we discuss ECM composition and reconstruction, the contribution of ECs and perivascular cells, the air-blood barrier (ABB) function, and the effects of physiological factors during the lung microvasculature repair and engineering process. The goal of the present review is to confirm the possibility of success in lung microvascular engineering in whole organ engineering and explore the future direction of the current methodology.
Collapse
Affiliation(s)
- Tomoshi Tsuchiya
- Department of Surgical Oncology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.,Division of Nucleic Acid Drug Development, Research Institute for Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Ryoichiro Doi
- Department of Surgical Oncology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Tomohiro Obata
- Department of Surgical Oncology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Go Hatachi
- Department of Surgical Oncology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Takeshi Nagayasu
- Department of Surgical Oncology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
37
|
Hussein KH, Park KM, Yu L, Song SH, Woo HM, Kwak HH. Vascular reconstruction: A major challenge in developing a functional whole solid organ graft from decellularized organs. Acta Biomater 2020; 103:68-80. [PMID: 31887454 DOI: 10.1016/j.actbio.2019.12.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 02/07/2023]
Abstract
Bioengineering a functional organ holds great potential to overcome the current gap between the organ need and shortage of available organs. Whole organ decellularization allows the removal of cells from large-scale organs, leaving behind extracellular matrices containing different growth factors, structural proteins, and a vascular network with a bare surface. Successful application of decellularized tissues as transplantable organs is hampered by the inability to completely reline the vasculature by endothelial cells (ECs), leading to blood coagulation, loss of vascular patency, and subsequent death of reseeded cells. Therefore, an intact, continuous layer of endothelium is essential to maintain proper functioning of the vascular system, which includes the transfer of nutrients to surrounding tissues and protecting other types of cells from shear stress. Here, we aimed to summarize the available cell sources that can be used for reendothelialization in addition to different trials performed by researchers to reconstruct vascularization of decellularized solid organs. Additionally, different techniques for enhancing reendothelialization and the methods used for evaluating reendothelialization efficiency along with the future prospective applications of this field are discussed. STATEMENT OF SIGNIFICANCE: Despite the great progress in whole organ decellularization, reconstruction of vasculature within the engineered constructs is still a major roadblock. Reconstructed endothelium acts as a multifunctional barrier of vessels, which can reduce thrombosis and help delivering of oxygen and nutrients throughout the whole organ. Successful reendothelialization can be achieved through reseeding of appropriate cell types on the naked vasculature with or without modification of its surface. Here, we present the current research milestones that so far established to reconstruct the vascular network in addition to the methods used for evaluating the efficiency of reendotheilization. Thus, this review is quite significant and will aid the researchers to know where we stand toward biofabricating a transplantable organ from decellularizd extracellular matrix.
Collapse
|
38
|
Ebrahimi Sadrabadi A, Baei P, Hosseini S, Baghaban Eslaminejad M. Decellularized Extracellular Matrix as a Potent Natural Biomaterial for Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1341:27-43. [PMID: 32166633 DOI: 10.1007/5584_2020_504] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Decellularization technique is a favorable method used to fabricate natural and tissue-like scaffolds. This technique is important because of its remarkable ability to perfectly mimic the natural extracellular matrix (ECM). ECM-based scaffolds/hydrogels provide structural support for cell differentiation and maturation. Therefore, novel natural-based bioinks, ECM-based hydrogels, and particulate forms of the ECM provide promising strategies for whole organ regeneration. Despite its efficacious characteristics, removal of residual detergent and the presence of various protocols make this technique challenging for scientists and regenerative medicine-related programs. This chapter reviews the most effective physical, chemical, and enzymatic protocols used to remove the cellular components and their challenges. We discuss the applications of decellularized ECM (dECM) in tissue engineering and regenerative medicine with an emphasis on hard tissues.
Collapse
Affiliation(s)
- Amin Ebrahimi Sadrabadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Payam Baei
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Samaneh Hosseini
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran. .,Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
39
|
Xie M, Wang Z, Wan X, Weng J, Tu M, Mei J, Wang Z, Du X, Wang L, Chen C. Crosslinking effects of branched PEG on decellularized lungs of rats for tissue engineering. J Biomater Appl 2019; 34:965-974. [PMID: 31690161 DOI: 10.1177/0885328219885068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Mengying Xie
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhiyi Wang
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinlong Wan
- School of basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jie Weng
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mengyun Tu
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jin Mei
- School of basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zhibin Wang
- School of basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiaohong Du
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liangxing Wang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chan Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
40
|
Wang Z, Mithieux SM, Weiss AS. Fabrication Techniques for Vascular and Vascularized Tissue Engineering. Adv Healthc Mater 2019; 8:e1900742. [PMID: 31402593 DOI: 10.1002/adhm.201900742] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/12/2019] [Indexed: 12/19/2022]
Abstract
Impaired or damaged blood vessels can occur at all levels in the hierarchy of vascular systems from large vasculatures such as arteries and veins to meso- and microvasculatures such as arterioles, venules, and capillary networks. Vascular tissue engineering has become a promising approach for fabricating small-diameter vascular grafts for occlusive arteries. Vascularized tissue engineering aims to fabricate meso- and microvasculatures for the prevascularization of engineered tissues and organs. The ideal small-diameter vascular graft is biocompatible, bridgeable, and mechanically robust to maintain patency while promoting tissue remodeling. The desirable fabricated meso- and microvasculatures should rapidly integrate with the host blood vessels and allow nutrient and waste exchange throughout the construct after implantation. A number of techniques used, including engineering-based and cell-based approaches, to fabricate these synthetic vasculatures are herein explored, as well as the techniques developed to fabricate hierarchical structures that comprise multiple levels of vasculature.
Collapse
Affiliation(s)
- Ziyu Wang
- School of Life and Environmental Sciences University of Sydney NSW 2006 Australia
- Charles Perkins Centre University of Sydney NSW 2006 Australia
| | - Suzanne M. Mithieux
- School of Life and Environmental Sciences University of Sydney NSW 2006 Australia
- Charles Perkins Centre University of Sydney NSW 2006 Australia
| | - Anthony S. Weiss
- School of Life and Environmental Sciences University of Sydney NSW 2006 Australia
- Charles Perkins Centre University of Sydney NSW 2006 Australia
- Bosch Institute University of Sydney NSW 2006 Australia
- Sydney Nano Institute University of Sydney NSW 2006 Australia
| |
Collapse
|
41
|
KC P, Hong Y, Zhang G. Cardiac tissue-derived extracellular matrix scaffolds for myocardial repair: advantages and challenges. Regen Biomater 2019; 6:185-199. [PMID: 31404421 PMCID: PMC6683951 DOI: 10.1093/rb/rbz017] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 03/04/2019] [Accepted: 03/14/2019] [Indexed: 12/12/2022] Open
Abstract
Decellularized extracellular matrix (dECM) derived from myocardium has been widely explored as a nature scaffold for cardiac tissue engineering applications. Cardiac dECM offers many unique advantages such as preservation of organ-specific ECM microstructure and composition, demonstration of tissue-mimetic mechanical properties and retention of biochemical cues in favor of subsequent recellularization. However, current processes of dECM decellularization and recellularization still face many challenges including the need for balance between cell removal and extracellular matrix preservation, efficient recellularization of dECM for obtaining homogenous cell distribution, tailoring material properties of dECM for enhancing bioactivity and prevascularization of thick dECM. This review summarizes the recent progresses of using dECM scaffold for cardiac repair and discusses its major advantages and challenges for producing biomimetic cardiac patch.
Collapse
Affiliation(s)
- Pawan KC
- Department of Biomedical Engineering, The University of Akron, Olson Research Center, Room 301L, 260 S Forge Street, Akron, OH, USA
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, 500 UTA Blvd, Room 240, Arlington, TX, USA
| | - Ge Zhang
- Department of Biomedical Engineering, The University of Akron, Olson Research Center, Room 301L, 260 S Forge Street, Akron, OH, USA
| |
Collapse
|
42
|
Affiliation(s)
- Jesus M Gomez-Salinero
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Shahin Rafii
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
43
|
Abstract
AIMS AND OBJECTIVES Children suffering from intestinal failure (IF) endure considerable morbidity and overall have poor survival rates, complicated by the shortage of organs available for transplantation. Therefore, new therapeutic approaches are pivotal if outcomes are to be improved. Over the past years, tissue engineering (TE) has emerged as a possible alternative treatment for many congenital and acquired conditions. TE aims at creating bioengineered organs by means of combining scaffolds with appropriate cell types, which in the intestine are organised within a multilayer structure. In order to generate functional intestine, this cellular diversity and organisation will need to be recreated. While the cells for the epithelial, neural and vascular compartments have been well defined, so far, less attention has been put on the muscular compartment. More recently, mesoangioblasts (MABs) have been identified as a novel source for tissue regeneration since they are able to give rise to vascular and other mesodermal derivatives. To date MABs have not been successfully isolated from intestinal tissue. Therefore, our aim was to demonstrate the possibility of isolating MABs from adult mouse small intestine. MATERIALS AND METHODS All experiments were carried out using small intestinal tissues from C57BL/6J mice. We applied an established protocol for MAB isolation from the isolated neuromuscular layer of the small intestine. Cultured cells were stained for Ki67 to assess proliferation rates as well as for a panel of pericyte markers to determine their phenotype. RESULTS Cells were successfully isolated from gut biopsies. Cultured cells showed good proliferative capacity and positivity for at least three pericytes markers found in vessels of the gut neuromuscular wall: neuron-glial antigen 2, alkaline phosphatase and platelet-derived growth factor β. CONCLUSION This proof-of-principle study lays the foundation for further characterization of MABs as a possible cell source for intestinal smooth muscle regeneration and TE.
Collapse
|
44
|
Zambaiti E, Scottoni F, Rizzi E, Russo S, Deguchi K, Eaton S, Pellegata AF, De Coppi P. Whole rat stomach decellularisation using a detergent-enzymatic protocol. Pediatr Surg Int 2019; 35:21-27. [PMID: 30443739 PMCID: PMC6326006 DOI: 10.1007/s00383-018-4372-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/18/2018] [Indexed: 02/03/2023]
Abstract
BACKGROUND Conditions leading to reduced gastric volume are difficult to manage and are associated to poor quality-of-life. Stomach augmentation using a tissue-engineered stomach is a potential solution to restore adequate physiology and food reservoir. Aim of this study was to evaluate the decellularisation of whole rat stomach using a detergent-enzymatic protocol. METHODS Stomachs harvested from rats were decellularised through luminal and vascular cannulation using 24-h detergent-enzymatic treatment and completely characterized by appropriate staining, DNA and Extracellular matrix -component quantifications. RESULTS The detergent-enzymatic protocol allows a complete decellularisation of the gastric tissue, with a complete removal of the DNA with two cycles as confirmed by both quantifications and histological analysis. Extracellular matrix components, collagen, fibronectin, laminin and elastin, were optimally preserved by the treatment, while glycosaminoglycans were reduced. CONCLUSION Gastric tissue can be efficiently decellularised. Scaffolds retained original structure and important components that could enhance integration with other tissues for in vivo transplant. The use of naturally derived material could be potentially considered for the treatment of both congenital and acquired conditions.
Collapse
Affiliation(s)
- Elisa Zambaiti
- Stem Cell and Regenerative Medicine Section, DBC, UCL, Great Ormond Street Institute of Child Health, University College of London, Surgery Offices, 30 Guilford Street, London, WC1N 1EH UK
| | - Federico Scottoni
- Stem Cell and Regenerative Medicine Section, DBC, UCL, Great Ormond Street Institute of Child Health, University College of London, Surgery Offices, 30 Guilford Street, London, WC1N 1EH UK
| | - Eleonora Rizzi
- Stem Cell and Regenerative Medicine Section, DBC, UCL, Great Ormond Street Institute of Child Health, University College of London, Surgery Offices, 30 Guilford Street, London, WC1N 1EH UK
| | - Simone Russo
- Stem Cell and Regenerative Medicine Section, DBC, UCL, Great Ormond Street Institute of Child Health, University College of London, Surgery Offices, 30 Guilford Street, London, WC1N 1EH UK
| | - Koichi Deguchi
- Stem Cell and Regenerative Medicine Section, DBC, UCL, Great Ormond Street Institute of Child Health, University College of London, Surgery Offices, 30 Guilford Street, London, WC1N 1EH UK
| | - Simon Eaton
- Stem Cell and Regenerative Medicine Section, DBC, UCL, Great Ormond Street Institute of Child Health, University College of London, Surgery Offices, 30 Guilford Street, London, WC1N 1EH UK
| | - Alessandro F. Pellegata
- Stem Cell and Regenerative Medicine Section, DBC, UCL, Great Ormond Street Institute of Child Health, University College of London, Surgery Offices, 30 Guilford Street, London, WC1N 1EH UK
| | - Paolo De Coppi
- Stem Cell and Regenerative Medicine Section, DBC, UCL, Great Ormond Street Institute of Child Health, University College of London, Surgery Offices, 30 Guilford Street, London, WC1N 1EH UK ,Specialist Neonatal and Paediatric Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| |
Collapse
|
45
|
Improving functional re-endothelialization of acellular liver scaffold using REDV cell-binding domain. Acta Biomater 2018; 78:151-164. [PMID: 30071351 DOI: 10.1016/j.actbio.2018.07.046] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 07/02/2018] [Accepted: 07/26/2018] [Indexed: 12/15/2022]
Abstract
Engineering of functional vascularized liver tissues holds great promise in addressing donor organ shortage for transplantation. Whole organ decellularization is a cell removal method that retains the native vascular structures of the organ such that it can be anastomosed with the recipient circulation after recellularization with healthy cells. However, a main hurdle to successful implantation of bioengineered organ is the inability to efficiently re-endothelialize the vasculature with a functional endothelium, resulting in blood clotting which is the primary cause of failure in early transplant studies. Here, we present an efficient approach for enhancing re-endothelialization of decellularized rat liver scaffolds by conjugating the REDV cell-binding domain to improve attachment of endothelial cells (EC) on vascular wall surfaces. In order to facilitate expression and purification of the peptide, REDV was fused with elastin-like peptide (ELP) that confers thermally triggered aggregation behavior to the fusion protein. After validating the adhesive properties of the REDV-ELP peptide, we covalently coupled REDV-ELP to the blood vasculature of decellularized rat livers and seeded EC using perfusion of the portal vein. We showed that REDV-ELP increased cell attachment, spreading and proliferation of EC within the construct resulting in uniform endothelial lining of the scaffold vasculature. We further observed that REDV-ELP conjugation dramatically reduced platelet adhesion and activation. Altogether, our results demonstrate that this method allowed functional re-endothelialization of liver scaffold and show great potential toward the generation of functional bioengineered liver for long-term transplantation. STATEMENT OF SIGNIFICANCE There is a critical need for novel organ replacement therapies as the grafts for transplantation fall short of demand. Recent advances in tissue engineering, through the use of decellularized scaffolds, have opened the possibility that engineered grafts could be used as substitutes for donor livers. However, successful implantation has been challenged by the inability to create a functional vasculature. Our research study reports a new strategy to increase efficiency of endothelialization by increasing the affinity of the vascular matrix for endothelial cells. We functionalized decellularized liver scaffold using elastin-like peptides grafted with REDV cell binding domain. We showed that REDV-ELP conjugation improve endothelial cell attachment and proliferation within the scaffold, demonstrating the feasibility of re-endothelializing a whole liver vasculature using our technique.
Collapse
|