1
|
Gomez-Salinero JM, Redmond D, Rafii S. Microenvironmental determinants of endothelial cell heterogeneity. Nat Rev Mol Cell Biol 2025; 26:476-495. [PMID: 39875728 DOI: 10.1038/s41580-024-00825-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2024] [Indexed: 01/30/2025]
Abstract
During development, endothelial cells (ECs) undergo an extraordinary specialization by which generic capillary microcirculatory networks spanning from arteries to veins transform into patterned organotypic zonated blood vessels. These capillary ECs become specialized to support the cellular and metabolic demands of each specific organ, including supplying tissue-specific angiocrine factors that orchestrate organ development, maintenance of organ-specific functions and regeneration of injured adult organs. Here, we illustrate the mechanisms by which microenvironmental signals emanating from non-vascular niche cells induce generic ECs to acquire specific inter-organ and intra-organ functional attributes. We describe how perivascular, parenchymal and immune cells dictate vascular heterogeneity and capillary zonation, and how this system is maintained through tissue-specific signalling activated by vasculogenic and angiogenic factors and deposition of matrix components. We also discuss how perturbation of organotypic vascular niche cues lead to erasure of EC signatures, contributing to the pathogenesis of disease processes. We also describe approaches that use reconstitution of tissue-specific signatures of ECs to promote regeneration of damaged organs.
Collapse
Affiliation(s)
- Jesus M Gomez-Salinero
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - David Redmond
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Shahin Rafii
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
2
|
Safi K, Pawlicka AJ, Pradhan B, Sobieraj J, Zhylko A, Struga M, Grąt M, Chrzanowska A. Perspectives and Tools in Liver Graft Assessment: A Transformative Era in Liver Transplantation. Biomedicines 2025; 13:494. [PMID: 40002907 PMCID: PMC11852418 DOI: 10.3390/biomedicines13020494] [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: 12/16/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Liver transplantation is a critical and evolving field in modern medicine, offering life-saving treatment for patients with end-stage liver disease and other hepatic conditions. Despite its transformative potential, transplantation faces persistent challenges, including a global organ shortage, increasing liver disease prevalence, and significant waitlist mortality rates. Current donor evaluation practices often discard potentially viable livers, underscoring the need for refined graft assessment tools. This review explores advancements in graft evaluation and utilization aimed at expanding the donor pool and optimizing outcomes. Emerging technologies, such as imaging techniques, dynamic functional tests, and biomarkers, are increasingly critical for donor assessment, especially for marginal grafts. Machine learning and artificial intelligence, exemplified by tools like LiverColor, promise to revolutionize donor-recipient matching and liver viability predictions, while bioengineered liver grafts offer a future solution to the organ shortage. Advances in perfusion techniques are improving graft preservation and function, particularly for donation after circulatory death (DCD) grafts. While challenges remain-such as graft rejection, ischemia-reperfusion injury, and recurrence of liver disease-technological and procedural advancements are driving significant improvements in graft allocation, preservation, and post-transplant outcomes. This review highlights the transformative potential of integrating modern technologies and multidisciplinary approaches to expand the donor pool and improve equity and survival rates in liver transplantation.
Collapse
Affiliation(s)
- Kawthar Safi
- Department of Biochemistry, Medical University of Warsaw, 02-097 Warsaw, Poland; (K.S.)
| | | | - Bhaskar Pradhan
- Department of Biochemistry, Medical University of Warsaw, 02-097 Warsaw, Poland; (K.S.)
| | - Jan Sobieraj
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Andriy Zhylko
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland
| | - Marta Struga
- Department of Biochemistry, Medical University of Warsaw, 02-097 Warsaw, Poland; (K.S.)
| | - Michał Grąt
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland
| | - Alicja Chrzanowska
- Department of Biochemistry, Medical University of Warsaw, 02-097 Warsaw, Poland; (K.S.)
| |
Collapse
|
3
|
Wani SI, Mir TA, Nakamura M, Tsuchiya T, Alzhrani A, Iwanaga S, Arai K, Alshehri EA, Shamma T, Obeid DA, Chinnappan R, Assiri AM, Yaqinuddin A, Vashist YK, Broering DC. A review of current state-of-the-art materiobiology and technological approaches for liver tissue engineering. BIOPRINTING 2024; 42:e00355. [DOI: 10.1016/j.bprint.2024.e00355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
|
4
|
Capella-Monsonís H, Crum RJ, Hussey GS, Badylak SF. Advances, challenges, and future directions in the clinical translation of ECM biomaterials for regenerative medicine applications. Adv Drug Deliv Rev 2024; 211:115347. [PMID: 38844005 DOI: 10.1016/j.addr.2024.115347] [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: 03/26/2024] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 06/11/2024]
Abstract
Extracellular Matrix (ECM) scaffolds and biomaterials have been widely used for decades across a variety of diverse clinical applications and have been implanted in millions of patients worldwide. ECM-based biomaterials have been especially successful in soft tissue repair applications but their utility in other clinical applications such as for regeneration of bone or neural tissue is less well understood. The beneficial healing outcome with the use of ECM biomaterials is the result of their biocompatibility, their biophysical properties and their ability to modify cell behavior after injury. As a consequence of successful clinical outcomes, there has been motivation for the development of next-generation formulations of ECM materials ranging from hydrogels, bioinks, powders, to whole organ or tissue scaffolds. The continued development of novel ECM formulations as well as active research interest in these materials ensures a wealth of possibilities for future clinical translation and innovation in regenerative medicine. The clinical translation of next generation formulations ECM scaffolds faces predictable challenges such as manufacturing, manageable regulatory pathways, surgical implantation, and the cost required to address these challenges. The current status of ECM-based biomaterials, including clinical translation, novel formulations and therapies currently under development, and the challenges that limit clinical translation of ECM biomaterials are reviewed herein.
Collapse
Affiliation(s)
- Héctor Capella-Monsonís
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, USA; Department of Surgery, School of Medicine, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Viscus Biologics LLC, 2603 Miles Road, Cleveland, OH 44128, USA
| | - Raphael J Crum
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, USA; Department of Surgery, School of Medicine, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - George S Hussey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, USA; Department of Pathology, School of Medicine, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Stephen F Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, USA; Department of Surgery, School of Medicine, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Department of Bioengineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA 15261, USA.
| |
Collapse
|
5
|
Cavallini C, Olivi E, Tassinari R, Zannini C, Ragazzini G, Marcuzzi M, Taglioli V, Ventura C. Deer antler stem cell niche: An interesting perspective. World J Stem Cells 2024; 16:479-485. [PMID: 38817324 PMCID: PMC11135255 DOI: 10.4252/wjsc.v16.i5.479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/09/2024] [Accepted: 04/25/2024] [Indexed: 05/24/2024] Open
Abstract
In recent years, there has been considerable exploration into methods aimed at enhancing the regenerative capacity of transplanted and/or tissue-resident cells. Biomaterials, in particular, have garnered significant interest for their potential to serve as natural scaffolds for cells. In this editorial, we provide commentary on the study by Wang et al, in a recently published issue of World J Stem Cells, which investigates the use of a decellularized xenogeneic extracellular matrix (ECM) derived from antler stem cells for repairing osteochondral defects in rat knee joints. Our focus lies specifically on the crucial role of biological scaffolds as a strategy for augmenting stem cell potential and regenerative capabilities, thanks to the establishment of a favorable microenvironment (niche). Stem cell differentiation heavily depends on exposure to intrinsic properties of the ECM, including its chemical and protein composition, as well as the mechanical forces it can generate. Collectively, these physicochemical cues contribute to a bio-instructive signaling environment that offers tissue-specific guidance for achieving effective repair and regeneration. The interest in mechanobiology, often conceptualized as a form of "structural memory", is steadily gaining more validation and momentum, especially in light of findings such as these.
Collapse
Affiliation(s)
- Claudia Cavallini
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems - Eldor Lab, Bologna 40128, Italy
- Eldor Lab, Bologna 40128, Italy
| | | | | | | | | | - Martina Marcuzzi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna 40138, Italy
| | | | - Carlo Ventura
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems - Eldor Lab, Bologna 40128, Italy.
| |
Collapse
|
6
|
Im P, Shin H, Kim J. Tilapia-Derived Granular Hydrogel as a 3D Scaffold Promoting Rapid Wound Healing. Biomacromolecules 2024; 25:1153-1161. [PMID: 38290478 DOI: 10.1021/acs.biomac.3c01137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
The skin, a crucial organ that protects the body, is vulnerable to external damage. Traditional tissue regeneration methods, including bulk hydrogels, aim to facilitate wound healing by interacting with host cells and providing a conducive environment. However, the nanoscale porosity of conventional hydrogels limits cell penetration and tissue regeneration. To overcome this, hydrogels composed of microgels have emerged as promising alternatives. In this study, we propose a granular hydrogel using decellularized tilapia skin. The tilapia skin-based microgels are cost-effective, immune-friendly, and have a high collagen content. Microgels based on the decellularized extracellular matrix of tilapia were successfully fabricated by using microfluidics. Through the assembly of these microgels using adhesive hyaluronic acid-catechol, the resulting 3D granular hydrogel scaffold facilitated enhanced cell growth, accelerated cell differentiation, and successful healing of full-thickness wounds in a mouse model. This study reveals the potential of tilapia skin-based granular hydrogel assembly in wound healing, overcoming conventional hydrogel limits.
Collapse
Affiliation(s)
- Pilseon Im
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Hyunsu Shin
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Jaeyun Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Science & Technology (SAIHST), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Quantum Biophysics (IQB), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| |
Collapse
|
7
|
De S, Vasudevan A, Tripathi DM, Kaur S, Singh N. A decellularized matrix enriched collagen microscaffold for a 3D in vitro liver model. J Mater Chem B 2024; 12:772-783. [PMID: 38167699 DOI: 10.1039/d3tb01652h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
The development of liver scaffolds retaining their three-dimensional (3D) structure and extra-cellular matrix (ECM) composition is essential for the advancement of liver tissue engineering. We report the design and validation of an alginate-based platform using a combination of decellularized matrices and collagen to preserve the functionality of liver cells. The scaffolds were characterized using SEM and fluorescence microscopy techniques. The proliferation and functional behaviours of hepatocellular carcinoma HuH7 cells were observed. It was found that the decellularized skin scaffold with collagen was better for maintaining the growth of cells in comparison to other decellularized matrices. In addition, we observed a significant increase in the functional profile once exogenous collagen was added to the liver matrix. Our study also suggests that a cirrhotic liver model should have a different matrix composition as compared to a healthy liver model. When primary rat hepatocytes were used for developing a healthy liver model, the proliferation studies with hepatocytes showed a decellularized skin matrix as the better option, but the functionality was only maintained in a decellularized liver matrix with addition of exogenous collagen. We further checked if these platforms can be used for studying drug induced toxicity observed in the liver by studying the activation of cytochrome P450 upon drug exposure of the cells growing in our model. We observed a significant induction of the CYP1A1 gene on administering the drugs for 6 days. Thus, this platform could be used for drug-toxicity screening studies using primary hepatocytes in a short span of time. Being a microscaffold based system, this platform offers some advantages, such as smaller volumes of samples, analysing multiple samples simultaneously and a minimal amount of decellularized matrix in the matrix composition, making it an economical option compared to a completely dECM based platform.
Collapse
Affiliation(s)
- Shreemoyee De
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi-110016, India.
| | - Ashwini Vasudevan
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, D1, Vasant Kunj Marg, New Delhi, Delhi 110070, India.
| | - Dinesh M Tripathi
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, D1, Vasant Kunj Marg, New Delhi, Delhi 110070, India.
| | - Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, D1, Vasant Kunj Marg, New Delhi, Delhi 110070, India.
| | - Neetu Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi-110016, India.
- Biomedical Engineering Unit, All India Institute of Medical Sciences, Ansari Nagar, New Delhi-110029, India
| |
Collapse
|
8
|
Bhatt S S, Krishna Kumar J, Laya S, Thakur G, Nune M. Scaffold-mediated liver regeneration: A comprehensive exploration of current advances. J Tissue Eng 2024; 15:20417314241286092. [PMID: 39411269 PMCID: PMC11475092 DOI: 10.1177/20417314241286092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/08/2024] [Indexed: 10/19/2024] Open
Abstract
The liver coordinates over 500 biochemical processes crucial for maintaining homeostasis, detoxification, and metabolism. Its specialized cells, arranged in hexagonal lobules, enable it to function as a highly efficient metabolic engine. However, diseases such as cirrhosis, fatty liver disease, and hepatitis present significant global health challenges. Traditional drug development is expensive and often ineffective at predicting human responses, driving interest in advanced in vitro liver models utilizing 3D bioprinting and microfluidics. These models strive to mimic the liver's complex microenvironment, improving drug screening and disease research. Despite its resilience, the liver is vulnerable to chronic illnesses, injuries, and cancers, leading to millions of deaths annually. Organ shortages hinder liver transplantation, highlighting the need for alternative treatments. Tissue engineering, employing polymer-based scaffolds and 3D bioprinting, shows promise. This review examines these innovative strategies, including liver organoids and liver tissue-on-chip technologies, to address the challenges of liver diseases.
Collapse
Affiliation(s)
- Supriya Bhatt S
- Manipal Institute of Regenerative Medicine, Bengaluru, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Jayanthi Krishna Kumar
- Manipal Institute of Regenerative Medicine, Bengaluru, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Shurthi Laya
- Manipal Institute of Regenerative Medicine, Bengaluru, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
- Department of Biomedical Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Goutam Thakur
- Department of Biomedical Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Manasa Nune
- Manipal Institute of Regenerative Medicine, Bengaluru, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
9
|
Wang F, Wang H, Shan X, Mei J, Wei P, Song Q, Chen W. High-strength and high-toughness ECM films with the potential for peripheral nerve repair. Biomed Mater 2023; 19:015010. [PMID: 38048625 DOI: 10.1088/1748-605x/ad11fa] [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: 06/27/2023] [Accepted: 12/04/2023] [Indexed: 12/06/2023]
Abstract
Extracellular matrix (ECM) scaffolds are widely applied in the field of regeneration as the result of their irreplaceable biological advantages, and the preparation of ECM scaffolds into ECM hydrogels expands the applications to some extent. However, weak mechanical properties of current ECM materials limit the complete exploitation of ECM's biological advantages. To enable ECM materials to be utilized in applications requiring high strength, herein, we created a kind of new ECM material, ECM film, and evaluated its mechanical properties. ECM films exhibited outstanding toughness with no cracks after arbitrarily folding and crumpling, and dramatically high strength levels of 86 ± 17.25 MPa, the maximum of which was 115 MPa. Such spectacular high-strength and high-toughness films, containing only pure ECM without any crosslinking agents and other materials, far exceed current pure natural polymer gel films and even many composite gel films and synthetic polymer gel films. In addition, both PC12 cells and Schwann cells cultured on the surface of ECM films, especially Schwann cells, showed good proliferation, and the neurite outgrowth of the PC12 cells was promoted, indicating the application potential of ECM film in peripheral nerve repair.
Collapse
Affiliation(s)
- Fangfang Wang
- Medical Research Center, The First Affiliated Hospital of Ningbo University; Ningbo University, Ningbo 315010, People's Republic of China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo 315010, People's Republic of China
- Department of Plastic and Reconstructive Surgery, The First Affiliated Hospital of Ningbo University, Ningbo University, Ningbo 315010, People's Republic of China
| | - Haiyang Wang
- Institute of Bioscaffold Transplantation and Immunology, Wenzhou Medical University, Wenzhou 325035, People's Republic of China
| | - Xiaotong Shan
- Department of Nephrology, The First Affiliated Hospital of Ningbo University, Ningbo University, Ningbo 315010, People's Republic of China
| | - Jin Mei
- Medical Research Center, The First Affiliated Hospital of Ningbo University; Ningbo University, Ningbo 315010, People's Republic of China
- Institute of Bioscaffold Transplantation and Immunology, Wenzhou Medical University, Wenzhou 325035, People's Republic of China
- Department of Plastic and Reconstructive Surgery, The First Affiliated Hospital of Ningbo University, Ningbo University, Ningbo 315010, People's Republic of China
| | - Peng Wei
- Department of Plastic and Reconstructive Surgery, The First Affiliated Hospital of Ningbo University, Ningbo University, Ningbo 315010, People's Republic of China
| | - Qinghua Song
- Department of Plastic and Reconstructive Surgery, The First Affiliated Hospital of Ningbo University, Ningbo University, Ningbo 315010, People's Republic of China
| | - Weiwei Chen
- Department of Plastic and Reconstructive Surgery, The First Affiliated Hospital of Ningbo University, Ningbo University, Ningbo 315010, People's Republic of China
| |
Collapse
|
10
|
Septiana WL, Ayudyasari W, Gunardi H, Pawitan JA, Balachander GM, Yu H, Antarianto RD. Liver organoids cocultured on decellularized native liver scaffolds as a bridging therapy improves survival from liver failure in rabbits. In Vitro Cell Dev Biol Anim 2023; 59:747-763. [PMID: 38110841 DOI: 10.1007/s11626-023-00817-8] [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: 07/18/2023] [Accepted: 09/28/2023] [Indexed: 12/20/2023]
Abstract
The present study aimed to develop viable liver organoids using decellularized native liver scaffolds and evaluate the efficacy of human liver organoid transplantation in a rabbit model of cirrhosis. Liver organoids were formed by coculture of hepatocyte-like cells derived from the human-induced pluripotent stem cells with three other cell types. Twelve 3-mo-old New Zealand White Rabbits underwent a sham operation, bile duct ligation, or biliary duct ligation followed by liver organoid transplantation. Liver organoid structure and function before and after transplantation were evaluated using histological and molecular analyses. A survival analysis using the Kaplan-Meier method was performed to determine the cumulative probability of survival according to liver organoid transplantation with significantly greater overall survival observed in rabbits that underwent liver organoid transplantation (P = 0.003, log-rank test). The short-term group had higher hepatic expression levels of ALB and CYP3A mRNA and lower expression levels of AST mRNA compared to the long-term group. The short-term group also had lower collagen deposition in liver tissues. Transplantation of human liver organoids cocultured in decellularized native liver scaffold into rabbits that had undergone bile duct ligation improved short-term survival and hepatic function. The results of the present study highlight the potential of liver organoid transplantation as a bridging therapy in liver failure; however, rejection and poor liver organoid function may limit the long-term efficacy of this therapeutic approach.
Collapse
Affiliation(s)
- Wahyunia Likhayati Septiana
- Program Doktor Ilmu Biomedik, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Department of Histology, Faculty of Medicine, Universitas Gunadarma, Depok, Indonesia
| | - Wulan Ayudyasari
- Department of Surgery, Fakultas Kedokteran Universitas Indonesia, Jakarta, Indonesia
| | - Hardian Gunardi
- Department of Surgery, Fakultas Kedokteran Universitas Indonesia, Jakarta, Indonesia
| | - Jeanne Adiwinata Pawitan
- Department of Histology, Fakultas Kedokteran Universitas Indonesia, Jl Salemba Raya No 6. Jakarta Pusat 10430, Jakarta, Indonesia
- Stem Cell and Tissue Engineering Research Cluster, (IMERI) Indonesian Medical Education and Research Institute, Jakarta, Indonesia
- Integrated Service Unit of Stem Cell Medical Technology (IPT TK Sel Punca), Dr. Cipto Mangunkusumo General Hospital (RSCM), Jakarta, Indonesia
| | - Gowri Manohari Balachander
- Department of Physiology, The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, MD9-04-11, 2 Medical Drive, Singapore, 117593, Singapore
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, India, 221005
| | - Hanry Yu
- Department of Physiology, The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, MD9-04-11, 2 Medical Drive, Singapore, 117593, Singapore
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, India, 221005
- Institute of Bioengineering & Bioimaging, A*STAR, 31 Biopolis Way, #07-01, Singapore, 138669, Singapore
- CAMP, Singapore-MIT Alliance for Research and Technology, 1 CREATE Way, Level 4 Enterprise Wing, Singapore, 138602, Singapore
- Mechanobiology Institute, National University of Singapore, T-Lab, #05-01, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - Radiana Dhewayani Antarianto
- Department of Histology, Fakultas Kedokteran Universitas Indonesia, Jl Salemba Raya No 6. Jakarta Pusat 10430, Jakarta, Indonesia.
- Stem Cell and Tissue Engineering Research Cluster, (IMERI) Indonesian Medical Education and Research Institute, Jakarta, Indonesia.
| |
Collapse
|
11
|
Khaleghi S, Eivazkhani F, Tavana S, Moini A, Novin MG, Stoyan P, Nazarian H, Fathi R. Follicular reconstruction and neo-oogenesis in xenotransplantation of human ovarian isolated cells derived from chemotherapy-induced POF patients. J Biol Eng 2023; 17:70. [PMID: 37986177 PMCID: PMC10662631 DOI: 10.1186/s13036-023-00384-2] [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: 03/24/2023] [Accepted: 10/17/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND Developing new strategies to restore fertility in patients with chemotherapy-induced Premature Ovarian Failure (Chemo-POF) is important. We aimed to construct an Artificial Ovary (AO) by seeding Human Ovarian Cortical Cells (HOCCs) into Human ovarian Decellularized Cortical Tissue (DCT). We assessed the AO's ability to produce new ovarian follicles following xenotransplantation to NMRI mice. MATERIAL AND METHODS The DCTs were prepared, and cell removal was confirmed through DNA content, MTT assay, DAPI and H&E staining. Next, HOCCs were isolated from both Chemo-POF and Trans (as a control group) ovarian patients. The HOCCs were characterized using immunostaining (FRAGILIS, Vimentin, and Inhibin α) and real time PCR (DDX4, STELLA, FRAGILIS, Vimentin, FSH-R, KI67) assays. The HOCCs were then seeded into the DCTs and cultured for one week to construct an AO, which was subsequently xenotransplanted into the mice. The existence of active follicles within the AO was studied with H&E and immunofluorescence (GDF9) staining, Real-time PCR (GDF9, ZP3) and hormone analysis (Estradiol, FSH and AMH). RESULTS The results of gene expression and immunostaining showed that 85-86% of the isolated cells from both Trans and Chemo-POF groups were positive for vimentin, while 2-5% were granulosa cells and OSCs were less than 3%. After xenotransplantation, histological study confirmed the presence of morphologically healthy reconstructed human ovarian follicles. Additionally, immunofluorescence staining of GDF9 and hormonal assay confirmed the presence of secretory-active follicles on the AO. CONCLUSION Our findings demonstrate that an artificial ovary produced by seeding HOCCs on DCT can support HOCCs proliferation as well as neo-oogenesis, and enable sex hormone secretion following xenotransplantation.
Collapse
Affiliation(s)
- Sara Khaleghi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farideh Eivazkhani
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Somayeh Tavana
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Ashraf Moini
- Department of Endocrinology and Female Infertility, Royan Institute of Reproductive Biomedicine, ACECR, Tehran, Iran
- Breast Disease Research Center (BDRC), Tehran University of Medical Science, Tehran, Iran
| | - Marefat Ghaffari Novin
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Petkov Stoyan
- Platform Degenerative Diseases, German Primate Center, GmbH, Leibniz Institute for Primate Research, Göttingen, 37077, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, 37077, Germany
| | - Hamid Nazarian
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Rouhollah Fathi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
| |
Collapse
|
12
|
Allu I, Sahi AK, Koppadi M, Gundu S, Sionkowska A. Decellularization Techniques for Tissue Engineering: Towards Replicating Native Extracellular Matrix Architecture in Liver Regeneration. J Funct Biomater 2023; 14:518. [PMID: 37888183 PMCID: PMC10607724 DOI: 10.3390/jfb14100518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
The process of tissue regeneration requires the utilization of a scaffold, which serves as a structural framework facilitating cellular adhesion, proliferation, and migration within a physical environment. The primary aim of scaffolds in tissue engineering is to mimic the structural and functional properties of the extracellular matrix (ECM) in the target tissue. The construction of scaffolds that accurately mimic the architecture of the extracellular matrix (ECM) is a challenging task, primarily due to the intricate structural nature and complex composition of the ECM. The technique of decellularization has gained significant attention in the field of tissue regeneration because of its ability to produce natural scaffolds by removing cellular and genetic components from the extracellular matrix (ECM) while preserving its structural integrity. The present study aims to investigate the various decellularization techniques employed for the purpose of isolating the extracellular matrix (ECM) from its native tissue. Additionally, a comprehensive comparison of these methods will be presented, highlighting their respective advantages and disadvantages. The primary objective of this study is to gain a comprehensive understanding of the anatomical and functional features of the native liver, as well as the prevalence and impact of liver diseases. Additionally, this study aims to identify the limitations and difficulties associated with existing therapeutic methods for liver diseases. Furthermore, the study explores the potential of tissue engineering techniques in addressing these challenges and enhancing liver performance. By investigating these aspects, this research field aims to contribute to the advancement of liver disease treatment and management.
Collapse
Affiliation(s)
- Ishita Allu
- Department of Biomedical Engineering, University College of Engineering (UCE), Osmania University, Hyderabad 500007, India; (I.A.); (M.K.)
| | - Ajay Kumar Sahi
- School of Medicine, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA;
| | - Meghana Koppadi
- Department of Biomedical Engineering, University College of Engineering (UCE), Osmania University, Hyderabad 500007, India; (I.A.); (M.K.)
| | - Shravanya Gundu
- Department of Biomedical Engineering, University College of Engineering (UCE), Osmania University, Hyderabad 500007, India; (I.A.); (M.K.)
| | - Alina Sionkowska
- Faculty of Chemistry, Nicolaus Copernicus University in Torun, Jurija Gagarina 11, 87-100 Torun, Poland
- Faculty of Health Sciences, Calisia University, Nowy Świat 4, 62-800 Kalisz, Poland
| |
Collapse
|
13
|
Nishi K, Yagi H, Ohtomo M, Nagata S, Udagawa D, Tsuchida T, Morisaku T, Kitagawa Y. A thioacetamide-induced liver fibrosis model for pre-clinical studies in microminipig. Sci Rep 2023; 13:14996. [PMID: 37696857 PMCID: PMC10495379 DOI: 10.1038/s41598-023-42144-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/06/2023] [Indexed: 09/13/2023] Open
Abstract
Drug-induced liver fibrosis models are used in normal and immunosuppressed small animals for transplantation and regenerative medicine to improve liver fibrosis. Although large animal models are needed for pre-clinical studies, they are yet to be established owing to drug sensitivity in animal species and difficulty in setting doses. In this study, we evaluated liver fibrosis by administering thioacetamide (TA) to normal microminipig and thymectomized microminipig; 3 times for 1 week (total duration: 8 weeks). The pigs treated with TA showed elevated blood cytokine levels and a continuous liver injury at 8 weeks. RNA-seq of the liver showed increased expression of fibrosis-related genes after TA treatment. Histopathological examination showed degenerative necrosis of hepatocytes around the central vein, and revealed fibrogenesis and hepatocyte proliferation. TA treatment caused CD3-positive T cells and macrophages scattered within the hepatic lobule to congregate near the center of the lobule and increased αSMA-positive cells. Thymectomized pigs showed liver fibrosis similar to that of normal pigs, although the clinical signs tended to be milder. This model is similar to pathogenesis of liver fibrosis reported in other animal models. Therefore, it is expected to contribute to research as a drug discovery and pre-clinical transplantation models.
Collapse
Affiliation(s)
- Kotaro Nishi
- Department of Surgery, Keio University School of Medicine, 35, Shinano-machi, Shinjuku-ku, Tokyo, Japan
| | - Hiroshi Yagi
- Department of Surgery, Keio University School of Medicine, 35, Shinano-machi, Shinjuku-ku, Tokyo, Japan.
| | - Mana Ohtomo
- Department of Surgery, Keio University School of Medicine, 35, Shinano-machi, Shinjuku-ku, Tokyo, Japan
| | - Shogo Nagata
- Department of Surgery, Keio University School of Medicine, 35, Shinano-machi, Shinjuku-ku, Tokyo, Japan
| | - Daisuke Udagawa
- Department of Surgery, Keio University School of Medicine, 35, Shinano-machi, Shinjuku-ku, Tokyo, Japan
| | - Tomonori Tsuchida
- Department of Surgery, Keio University School of Medicine, 35, Shinano-machi, Shinjuku-ku, Tokyo, Japan
| | - Toshinori Morisaku
- Department of Surgery, Keio University School of Medicine, 35, Shinano-machi, Shinjuku-ku, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, 35, Shinano-machi, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
14
|
Ibi Y, Nishinakamura R. Kidney Bioengineering for Transplantation. Transplantation 2023; 107:1883-1894. [PMID: 36717963 DOI: 10.1097/tp.0000000000004526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The kidney is an important organ for maintenance of homeostasis in the human body. As renal failure progresses, renal replacement therapy becomes necessary. However, there is a chronic shortage of kidney donors, creating a major problem for transplantation. To solve this problem, many strategies for the generation of transplantable kidneys are under investigation. Since the first reports describing that nephron progenitors could be induced from human induced pluripotent stem cells, kidney organoids have been attracting attention as tools for studying human kidney development and diseases. Because the kidney is formed through the interactions of multiple renal progenitors, current studies are investigating ways to combine these progenitors derived from human induced pluripotent stem cells for the generation of transplantable kidney organoids. Other bioengineering strategies, such as decellularization and recellularization of scaffolds, 3-dimensional bioprinting, interspecies blastocyst complementation and progenitor replacement, and xenotransplantation, also have the potential to generate whole kidneys, although each of these strategies has its own challenges. Combinations of these approaches will lead to the generation of bioengineered kidneys that are transplantable into humans.
Collapse
Affiliation(s)
- Yutaro Ibi
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | | |
Collapse
|
15
|
Schlegel A, Mergental H, Fondevila C, Porte RJ, Friend PJ, Dutkowski P. Machine perfusion of the liver and bioengineering. J Hepatol 2023; 78:1181-1198. [PMID: 37208105 DOI: 10.1016/j.jhep.2023.02.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/03/2023] [Accepted: 02/09/2023] [Indexed: 05/21/2023]
Abstract
With the increasing number of accepted candidates on waiting lists worldwide, there is an urgent need to expand the number and the quality of donor livers. Dynamic preservation approaches have demonstrated various benefits, including improving liver function and graft survival, and reducing liver injury and post-transplant complications. Consequently, organ perfusion techniques are being used in clinical practice in many countries. Despite this success, a proportion of livers do not meet current viability tests required for transplantation, even with the use of modern perfusion techniques. Therefore, devices are needed to further optimise machine liver perfusion - one promising option is to prolong machine liver perfusion for several days, with ex situ treatment of perfused livers. For example, stem cells, senolytics, or molecules targeting mitochondria or downstream signalling can be administered during long-term liver perfusion to modulate repair mechanisms and regeneration. Besides, today's perfusion equipment is also designed to enable the use of various liver bioengineering techniques, to develop scaffolds or for their re-cellularisation. Cells or entire livers can also undergo gene modulation to modify animal livers for xenotransplantation, to directly treat injured organs or to repopulate such scaffolds with "repaired" autologous cells. This review first discusses current strategies to improve the quality of donor livers, and secondly reports on bioengineering techniques to design optimised organs during machine perfusion. Current practice, as well as the benefits and challenges associated with these different perfusion strategies are discussed.
Collapse
Affiliation(s)
- Andrea Schlegel
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Centre of Preclinical Research, Milan, 20122, Italy; Department of Surgery and Transplantation, Swiss HPB Center, University Hospital Zurich, Switzerland
| | - Hynek Mergental
- The Liver Unit, Queen Elizabeth University Hospital Birmingham, United Kingdom
| | - Constantino Fondevila
- Hepatopancreatobiliary Surgery & Transplantation, General & Digestive Surgery Service, Hospital Universitario La Paz, IdiPAZ, CIBERehd, Madrid, Spain
| | - Robert J Porte
- Erasmus MC Transplant Institute, Department of Surgery, Division of HPB & Transplant Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Peter J Friend
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Philipp Dutkowski
- Department of Surgery and Transplantation, Swiss HPB Center, University Hospital Zurich, Switzerland.
| |
Collapse
|
16
|
Dias ML, Wajsenzon IJR, Alves GBN, Paranhos BA, Andrade CBV, Siqueira Monteiro VR, de Sousa RMR, da Silva Pereira ENG, Rodrigues KL, Daliry A, Mello DB, Coeli dos Santos Goldenberg R. Cirrhotic Liver Sustains In Situ Regeneration of Acellular Liver Scaffolds after Transplantation into G-CSF-Treated Animals. Cells 2023; 12:cells12070976. [PMID: 37048049 PMCID: PMC10093225 DOI: 10.3390/cells12070976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 03/29/2023] Open
Abstract
Acellular liver scaffolds (ALS) produced by decellularization have been successfully explored for distinct regenerative purposes. To date, it is unknown whether transplanted ALSs are affected by cirrhotic livers, either becoming cirrhotic themselves or instead remaining as a robust template for healthy cell growth after transplantation into cirrhotic rats. Moreover, little is known about the clinical course of recipient cirrhotic livers after ALS transplantation. To address these questions, we transplanted ALSs into cirrhotic rats previously treated with the granulocyte colony-stimulating factor. Here, we report successful cellular engraftment within the transplanted ALSs at 7, 15, and 30 days after transplantation. Recellularization was orchestrated by liver tissue cell activation, resident hepatocytes and bile duct proliferation, and an immune response mediated by the granulocyte components. Furthermore, we showed that transplanted ALSs ensured a pro-regenerative and anti-inflammatory microenvironment, attracted vessels from the host cirrhotic tissue, and promoted progenitor cell recruitment. ALS transplantation induced cirrhotic liver regeneration and extracellular matrix remodeling. Moreover, the transplanted ALS sustained blood circulation and attenuated alterations in the ultrasonographic and biochemical parameters in cirrhotic rats. Taken together, our results confirm that transplanted ALSs are not affected by cirrhotic livers and remain a robust template for healthy cell growth and stimulated cirrhotic liver regeneration.
Collapse
|
17
|
Mir TA, Nakamura M, Sakai S, Iwanaga S, Wani SI, Alzhrani A, Arai K, Mir BA, Kazmi S, Assiri AM, Broering DC. Mammalian-specific decellularized matrices derived bioink for bioengineering of liver tissue analogues: A review. Int J Bioprint 2023; 9:714. [PMID: 37273993 PMCID: PMC10236352 DOI: 10.18063/ijb.714] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/18/2023] [Indexed: 06/06/2023] Open
Abstract
The absolute shortage of compatible liver donors and the growing number of potential recipients have led scientists to explore alternative approaches to providing tissue/ organ substitutes from bioengineered sources. Bioartificial regeneration of a fully functional tissue/organ replacement is highly dependent on the right combination of engineering tools, biological principles, and materiobiology horizons. Over the past two decades, remarkable achievements have been made in hepatic tissue engineering by converging various advanced interdisciplinary research approaches. Three-dimensional (3D) bioprinting has arisen as a promising state-of-the-art tool with strong potential to fabricate volumetric liver tissue/organ equivalents using viscosity- and degradation-controlled printable bioinks composed of hydrous microenvironments, and formulations containing living cells and associated supplements. Source of origin, biophysiochemical, or thermomechanical properties and crosslinking reaction kinetics are prerequisites for ideal bioink formulation and realizing the bioprinting process. In this review, we delve into the forecast of the potential future utility of bioprinting technology and the promise of tissue/organ- specific decellularized biomaterials as bioink substrates. Afterward, we outline various methods of decellularization, and the most relevant studies applying decellularized bioinks toward the bioengineering of in vitro liver models. Finally, the challenges and future prospects of decellularized material-based bioprinting in the direction of clinical regenerative medicine are presented to motivate further developments.
Collapse
Affiliation(s)
- Tanveer Ahmad Mir
- Transplant Research and Innovation Department, Tissue/Organ Bioengineering & BioMEMS Laboratory, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, KSA
| | - Makoto Nakamura
- Division of Biomedical System Engineering, Graduate School of Science and Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Shinji Sakai
- Division of Chemical Engineering, Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka 560-8531, Japan
| | - Shintaroh Iwanaga
- Division of Biomedical System Engineering, Graduate School of Science and Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Shadil Ibrahim Wani
- Division of Biomedical System Engineering, Graduate School of Science and Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Alaa Alzhrani
- Transplant Research and Innovation Department, Tissue/Organ Bioengineering & BioMEMS Laboratory, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, KSA
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, KSA
- College of Medicine, Alfaisal University, Riyadh 11211, KSA
| | - Kenichi Arai
- Department of Clinical Biomaterial Applied Science, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Bilal Ahmed Mir
- Division of Intellectual Information Engineering, Graduate School of Science and Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Shadab Kazmi
- Transplant Research and Innovation Department, Tissue/Organ Bioengineering & BioMEMS Laboratory, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, KSA
- Department of Child Health, School of Medicine, University of Missouri, Columbia, USA
| | - Abdullah M. Assiri
- Transplant Research and Innovation Department, Tissue/Organ Bioengineering & BioMEMS Laboratory, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, KSA
- College of Medicine, Alfaisal University, Riyadh 11211, KSA
| | - Dieter C. Broering
- Transplant Research and Innovation Department, Tissue/Organ Bioengineering & BioMEMS Laboratory, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, KSA
- College of Medicine, Alfaisal University, Riyadh 11211, KSA
| |
Collapse
|
18
|
Martinez-Castillo M, Altamirano-Mendoza I, Zielinski R, Priebe W, Piña-Barba C, Gutierrez-Reyes G. Collagen matrix scaffolds: Future perspectives for the management of chronic liver diseases. World J Clin Cases 2023; 11:1224-1235. [PMID: 36926129 PMCID: PMC10013111 DOI: 10.12998/wjcc.v11.i6.1224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/21/2022] [Accepted: 02/02/2023] [Indexed: 02/23/2023] Open
Abstract
Approximately 1.5 billion chronic liver disease (CLD) cases have been estimated worldwide, encompassing a wide range of liver damage severities. Moreover, liver disease causes approximately 1.75 million deaths per year. CLD is typically characterized by the silent and progressive deterioration of liver parenchyma due to an incessant inflammatory process, cell death, over deposition of extracellular matrix proteins, and dysregulated regeneration. Overall, these processes impair the correct function of this vital organ. Cirrhosis and liver cancer are the main complications of CLD, which accounts for 3.5% of all deaths worldwide. Liver transplantation is the optimal therapeutic option for advanced liver damage. The liver is one of the most common organs transplanted; however, only 10% of liver transplants are successful. In this context, regenerative medicine has made significant progress in the design of biomaterials, such as collagen matrix scaffolds, to address the limitations of organ transplantation (e.g., low donation rates and biocompatibility). Thus, it remains crucial to continue with experimental and clinical studies to validate the use of collagen matrix scaffolds in liver disease.
Collapse
Affiliation(s)
- Moises Martinez-Castillo
- Liver, Pancreas and Motility Laboratory, Unit of Experimental Medicine, School of Medicine, Universidad Nacional Autonoma de Mexico, Mexico City 06726, Mexico City, Mexico
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, United States
| | - Itzel Altamirano-Mendoza
- Liver, Pancreas and Motility Laboratory, Unit of Experimental Medicine, School of Medicine, Universidad Nacional Autonoma de Mexico, Mexico City 06726, Mexico City, Mexico
| | - Rafal Zielinski
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, United States
| | - Waldemar Priebe
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, United States
| | - Cristina Piña-Barba
- Materials Research Institute, Universidad Nacional Autónoma de México, Mexico City 06726, Mexico City, Mexico
| | - Gabriela Gutierrez-Reyes
- Liver, Pancreas and Motility Laboratory, Unit of Experimental Medicine, School of Medicine, Universidad Nacional Autonoma de Mexico, Mexico City 06726, Mexico City, Mexico
| |
Collapse
|
19
|
Cinelli L, Muttillo EM, Felli E, Baiocchini A, Giannone F, Marescaux J, Mutter D, De Mathelin M, Gioux S, Felli E, Diana M. Surgical Models of Liver Regeneration in Pigs: A Practical Review of the Literature for Researchers. Cells 2023; 12:603. [PMID: 36831271 PMCID: PMC9954688 DOI: 10.3390/cells12040603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/01/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
The remarkable capacity of regeneration of the liver is well known, although the involved mechanisms are far from being understood. Furthermore, limits concerning the residual functional mass of the liver remain critical in both fields of hepatic resection and transplantation. The aim of the present study was to review the surgical experiments regarding liver regeneration in pigs to promote experimental methodological standardization. The Pubmed, Medline, Scopus, and Cochrane Library databases were searched. Studies evaluating liver regeneration through surgical experiments performed on pigs were included. A total of 139 titles were screened, and 41 articles were included in the study, with 689 pigs in total. A total of 29 studies (71% of all) had a survival design, with an average study duration of 13 days. Overall, 36 studies (88%) considered partial hepatectomy, of which four were an associating liver partition and portal vein ligation for staged hepatectomy (ALPPS). Remnant liver volume ranged from 10% to 60%. Only 2 studies considered a hepatotoxic pre-treatment, while 25 studies evaluated additional liver procedures, such as stem cell application, ischemia/reperfusion injury, portal vein modulation, liver scaffold application, bio-artificial, and pharmacological liver treatment. Only nine authors analysed how cytokines and growth factors changed in response to liver resection. The most used imaging system to evaluate liver volume was CT-scan volumetry, even if performed only by nine authors. The pig represents one of the best animal models for the study of liver regeneration. However, it remains a mostly unexplored field due to the lack of experiments reproducing the chronic pathological aspects of the liver and the heterogeneity of existing studies.
Collapse
Affiliation(s)
- Lorenzo Cinelli
- Department of Gastrointestinal Surgery, San Raffaele Hospital IRCCS, 20132 Milan, Italy
- Research Institute against Digestive Cancer (IRCAD), 67000 Strasbourg, France
| | - Edoardo Maria Muttillo
- Division of General Surgery, Department of Medical and Surgical Sciences and Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, Via di Grottarossa 1035, 00189 Rome, Italy
| | - Emanuele Felli
- Service Chirurgie Digestive et Transplantation Hépatique, Hôpital Trousseau CHU, 37170 Tours, France
| | - Andrea Baiocchini
- Department of Pathology, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Fabio Giannone
- Digestive and Endocrine Surgery, Nouvel Hopital Civil, University of Strasbourg, 67000 Strasbourg, France
| | - Jacques Marescaux
- Research Institute against Digestive Cancer (IRCAD), 67000 Strasbourg, France
| | - Didier Mutter
- Digestive and Endocrine Surgery, Nouvel Hopital Civil, University of Strasbourg, 67000 Strasbourg, France
- Institut de Chirurgie Guidée par L’image, University Hospital Institute (IHU), University of Strasbourg, 67000 Strasbourg, France
| | - Michel De Mathelin
- ICube Laboratory, Photonics Instrumentation for Health, 67400 Strasbourg, France
| | - Sylvain Gioux
- ICube Laboratory, Photonics Instrumentation for Health, 67400 Strasbourg, France
| | - Eric Felli
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, 3012 Bern, Switzerland
| | - Michele Diana
- Research Institute against Digestive Cancer (IRCAD), 67000 Strasbourg, France
- Digestive and Endocrine Surgery, Nouvel Hopital Civil, University of Strasbourg, 67000 Strasbourg, France
- ICube Laboratory, Photonics Instrumentation for Health, 67400 Strasbourg, France
| |
Collapse
|
20
|
Zhu Y, Zhang L, Duan W, Martin-Saldaña S, Li C, Yu H, Feng L, Zhang X, Du B, Li G, Zheng X, Bu Y. Succinic Ester-Based Shape Memory Gelatin Sponge for Noncompressible Hemorrhage without Hindering Tissue Regeneration. Adv Healthc Mater 2023; 12:e2202122. [PMID: 36399015 DOI: 10.1002/adhm.202202122] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/18/2022] [Indexed: 11/19/2022]
Abstract
Shape memory sponges are very promising in stopping the bleeding from noncompressible and narrow entrance wounds. However, few shape memory sponges have fast degradable properties in order to not hinder tissue healing. In this work, based on cryopolymerization, a succinic ester-based sponge (Ssponge) is fabricated using gelatin and bi-polyethylene glycol-succinimidyl succinate (Bi-PEG-SS). Compared with the commercially available gelatin sponge (Csponge), Ssponge possesses better water/blood absorption ability and higher mechanical pressure over the surrounding tissues. Moreover, in the models of massive liver hemorrhage after transection and noncompressive liver wounds by penetration, Ssponge exhibits a better hemostasis performance than Csponge. Furthermore, in a liver regeneration model, Ssponge-treated livers shows higher regeneration speed compared with Csponge, including a lower injury score, more cavity-like tissues, less fibrosis and enhanced tissue regeneration. Overall, it is shown that Ssponge, with a fast degradation behavior, is not only highly efficient in stopping bleeding but also not detrimental for tissue healing, possessing promising clinical translational potential.
Collapse
Affiliation(s)
- Ye Zhu
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China.,Department of Orthopedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, P. R. China
| | - Lining Zhang
- Department of Rehabilitation Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, P. R. China
| | - Wanglin Duan
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Sergio Martin-Saldaña
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián, 20018, Spain
| | - Chaowei Li
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Hongwen Yu
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Luyao Feng
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Xianpeng Zhang
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Baoji Du
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Guanying Li
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Xifu Zheng
- Department of Orthopedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, P. R. China
| | - Yazhong Bu
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| |
Collapse
|
21
|
Yang Z, Liu X, Cribbin EM, Kim AM, Li JJ, Yong KT. Liver-on-a-chip: Considerations, advances, and beyond. BIOMICROFLUIDICS 2022; 16:061502. [PMID: 36389273 PMCID: PMC9646254 DOI: 10.1063/5.0106855] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/25/2022] [Indexed: 05/14/2023]
Abstract
The liver is the largest internal organ in the human body with largest mass of glandular tissue. Modeling the liver has been challenging due to its variety of major functions, including processing nutrients and vitamins, detoxification, and regulating body metabolism. The intrinsic shortfalls of conventional two-dimensional (2D) cell culture methods for studying pharmacokinetics in parenchymal cells (hepatocytes) have contributed to suboptimal outcomes in clinical trials and drug development. This prompts the development of highly automated, biomimetic liver-on-a-chip (LOC) devices to simulate native liver structure and function, with the aid of recent progress in microfluidics. LOC offers a cost-effective and accurate model for pharmacokinetics, pharmacodynamics, and toxicity studies. This review provides a critical update on recent developments in designing LOCs and fabrication strategies. We highlight biomimetic design approaches for LOCs, including mimicking liver structure and function, and their diverse applications in areas such as drug screening, toxicity assessment, and real-time biosensing. We capture the newest ideas in the field to advance the field of LOCs and address current challenges.
Collapse
Affiliation(s)
| | | | - Elise M. Cribbin
- School of Biomedical Engineering, University of Technology Sydney, New South Wales 2007, Australia
| | - Alice M. Kim
- School of Biomedical Engineering, University of Technology Sydney, New South Wales 2007, Australia
| | - Jiao Jiao Li
- Authors to whom correspondence should be addressed: and
| | - Ken-Tye Yong
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
22
|
Lascaris B, de Meijer VE, Porte RJ. Normothermic liver machine perfusion as a dynamic platform for regenerative purposes: What does the future have in store for us? J Hepatol 2022; 77:825-836. [PMID: 35533801 DOI: 10.1016/j.jhep.2022.04.033] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 04/19/2022] [Accepted: 04/27/2022] [Indexed: 01/06/2023]
Abstract
Liver transplantation has become an immense success; nevertheless, far more recipients are registered on waiting lists than there are available donor livers for transplantation. High-risk, extended criteria donor livers are increasingly used to reduce the discrepancy between organ demand and supply. Especially for high-risk livers, dynamic preservation using machine perfusion can decrease post-transplantation complications and may increase donor liver utilisation by improving graft quality and enabling viability testing before transplantation. To further increase the availability of donor livers suitable for transplantation, new strategies are required that make it possible to use organs that are initially too damaged to be transplanted. With the current progress in experimental liver transplantation research, (long-term) normothermic machine perfusion may be used in the future as a dynamic platform for regenerative medicine approaches, enabling repair and regeneration of injured donor livers. Currently explored therapeutics such as defatting cocktails, RNA interference, senolytics, and stem cell therapy may assist in the repair and/or regeneration of injured livers before transplantation. This review will provide a forecast of the future utility of normothermic machine perfusion in decreasing the imbalance between donor liver demand and supply by enabling the repair and regeneration of damaged donor livers.
Collapse
Affiliation(s)
- Bianca Lascaris
- Section of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vincent E de Meijer
- Section of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Robert J Porte
- Section of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
23
|
Talaei-Khozani T, Yaghoubi A. An overview of post transplantation events of decellularized scaffolds. Transpl Immunol 2022; 74:101640. [PMID: 35667545 DOI: 10.1016/j.trim.2022.101640] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/29/2022] [Accepted: 05/31/2022] [Indexed: 12/19/2022]
Abstract
Regenerative medicine and tissue engineering are reasonable techniques for repairing failed tissues and could be a suitable alternative to organ transplantation. One of the most widely used methods for preparing bioscaffolds is the decellularization procedure. Although cell debris and DNA are removed from the decellularized tissues, important compositions of the extracellular matrix including proteins, proteoglycans, and glycoproteins are nearly preserved. Moreover, the obtained scaffolds have a 3-dimensional (3D) structure, appropriate naïve mechanical properties, and good biocompatibility. After transplantation, different types of host cells migrate to the decellularized tissues. Histological and immunohistochemical assessment of the different bioscaffolds after implantation reveals the migration of parenchymal cells, angiogenesis, as well as the invasion of inflammatory and giant foreign cells. In this review, the events after transplantation including angiogenesis, scaffold degradation, and the presence of immune and tissue-specific progenitor cells in the decellularized scaffolds in various hosts, are discussed.
Collapse
Affiliation(s)
- Tahereh Talaei-Khozani
- Histotomorphometry and stereology research center, Shiraz University of Medical Sciences, Shiraz, Iran; Tissue engineering lab, Anatomy Department, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Atefeh Yaghoubi
- Tissue engineering lab, Anatomy Department, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
24
|
Bongolan T, Whiteley J, Castillo-Prado J, Fantin A, Larsen B, Wong CJ, Mazilescu L, Kawamura M, Urbanellis P, Jonebring A, Salter E, Collingridge G, Gladdy R, Hicks R, Gingras AC, Selzner M, Rogers IM. Decellularization of porcine kidney with submicellar concentrations of SDS results in the retention of ECM proteins required for the adhesion and maintenance of human adult renal epithelial cells. Biomater Sci 2022; 10:2972-2990. [PMID: 35521809 DOI: 10.1039/d1bm01017d] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
When decellularizing kidneys, it is important to maintain the integrity of the acellular extracellular matrix (ECM), including associated adhesion proteins and growth factors that allow recellularized cells to adhere and migrate according to ECM specificity. Kidney decellularization requires the ionic detergent sodium dodecyl sulfate (SDS); however, this results in a loss of ECM proteins important for cell adherence, migration, and growth, particularly glycosaminoglycan (GAG)-associated proteins. Here, we demonstrate that using submicellar concentrations of SDS results in a greater retention of structural proteins, GAGs, growth factors, and cytokines. When porcine kidney ECM scaffolds were recellularized using human adult primary renal epithelial cells (RECs), the ECM promoted cell survival and the uniform distribution of cells throughout the ECM. Cells maintained the expression of mature renal epithelial markers but did not organize on the ECM, indicating that mature cells are unable to migrate to specific locations on ECM scaffolds.
Collapse
Affiliation(s)
- Tonya Bongolan
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada.
| | - Jennifer Whiteley
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada.
| | - Jorge Castillo-Prado
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada.
| | - Amanda Fantin
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada.
| | - Brett Larsen
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada.
| | - Cassandra J Wong
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada.
| | - Laura Mazilescu
- Toronto General Hospital Research Institute, Toronto, ON, M5G 2C4, Canada.,Soham & Shaila Ajmera Family Transplant Centre, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Masataka Kawamura
- Toronto General Hospital Research Institute, Toronto, ON, M5G 2C4, Canada.,Soham & Shaila Ajmera Family Transplant Centre, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Peter Urbanellis
- Toronto General Hospital Research Institute, Toronto, ON, M5G 2C4, Canada.,Soham & Shaila Ajmera Family Transplant Centre, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Anna Jonebring
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, 431 83, Sweden
| | - Eric Salter
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada.
| | - Graham Collingridge
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada.
| | - Rebecca Gladdy
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Ryan Hicks
- BioPharmaceuticals R&D Cell Therapy Department, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, 431 83, Sweden
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada. .,Department of Molecular Genetics, University of Toronto, Toronto, ON, M5G1E2, Canada
| | - Markus Selzner
- Toronto General Hospital Research Institute, Toronto, ON, M5G 2C4, Canada.,Soham & Shaila Ajmera Family Transplant Centre, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Ian M Rogers
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada. .,Soham & Shaila Ajmera Family Transplant Centre, University Health Network, Toronto, ON, M5G 2C4, Canada.,Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, M5G1E2, Canada
| |
Collapse
|
25
|
Najar-Asl M, Bahadoran H, Asadi MH, Saheli M, Asghari MH, Sodeifi N, Ashtiani MK, Vosough M, Baharvand H, Piryaei A. Transplantation of SDF-1α-loaded liver extracellular matrix repopulated with autologous cells attenuated liver fibrosis in a rat model. EXCLI JOURNAL 2022; 21:704-721. [PMID: 35721572 PMCID: PMC9203988 DOI: 10.17179/excli2022-4761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/20/2022] [Indexed: 11/10/2022]
Abstract
Cell-based therapy and tissue engineering are promising substitutes for liver transplantation to cure end-stage liver disorders. However, the limited sources for healthy and functional cells and poor engraftment rate are main challenges to the cell-based therapy approach. On the other hand, feasibility of production and size of bioengineered tissues are primary bottlenecks in tissue engineering. Here, we induce regeneration in a rat fibrotic liver model by transplanting a natural bioengineered scaffold with a native microenvironment repopulated with autologous stem/progenitor cells. In the main experimental group, a 1 mm3 stromal derived factor-1α (SDF-1α; S) loaded scaffold from decellularized liver extracellular matrix (LEM) was transplanted (Tx) into a fibrotic liver and the endogenous stem/progenitor cells were mobilized via granulocyte colony stimulating factor (G-CSF; G) therapy. Four weeks after transplantation, changes in liver fibrosis and necrosis, efficacy of cell engraftment and differentiation, vasculogenesis, and liver function recovery were assessed in this (LEM-TxSG) group and compared to the other groups. We found significant reduction in liver fibrosis stage in the LEM-TxSG, LEM-TxS and LEM-TxG groups compared to the control (fibrotic) group. Liver necrosis grade, and alanine transaminase (ALT) and aspartate transaminase (AST) levels dramatically reduced in all experimental groups compared to the control group. However, the number of engrafted cells into the transplanted scaffold and ratio of albumin (Alb) positive cells per total incorporated cells were considerably higher in the LEM-TxSG group compared to the LEM-Tx, LEM-TxS and LEM-TxG groups. Serum Alb levels increased in the LEM-Tx, LEM-TxS, and LEM-TxG groups, and was highest in the LEM-TxSG group, which was significantly more than the fibrotic group. Small vessel formation in the LEM-TxSG group was significantly higher than the LEM-Tx and LEM-TxS groups. Totally, these findings support application of the in vivo tissue engineering approach as a possible novel therapeutic strategy for liver fibrosis.
Collapse
Affiliation(s)
- Mostafa Najar-Asl
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran,Department of Anatomical Sciences, School of Medical Sciences, Baqiyatallah University, Tehran, Iran
| | - Hossein Bahadoran
- Department of Anatomical Sciences, School of Medical Sciences, Baqiyatallah University, Tehran, Iran,*To whom correspondence should be addressed: Hossein Bahadoran, Department of Anatomical Sciences, School of Medical Sciences, Baqiyatallah University, Tehran, Iran; Tel: +98 9124276200, E-mail:
| | - Mohammad-Hossein Asadi
- Department of Anatomical Sciences, School of Medical Sciences, Baqiyatallah University, Tehran, Iran
| | - Mona Saheli
- Department of Anatomical Sciences, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad-Hassan Asghari
- Animal Core Facility, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Tehran, Iran
| | - Niloofar Sodeifi
- Department of Andrology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mohammad Kazemi Ashtiani
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran,Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Abbas Piryaei
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran,Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technology in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
Higashi H, Yagi H, Kuroda K, Tajima K, Kojima H, Nishi K, Morisaku T, Hirukawa K, Fukuda K, Matsubara K, Kitago M, Shinoda M, Obara H, Adachi S, Nishimura K, Natsume T, Tomi M, Soto-Gutierrez A, Kitagawa Y. Transplantation of bioengineered liver capable of extended function in a preclinical liver failure model. Am J Transplant 2022; 22:731-744. [PMID: 34932270 PMCID: PMC9008767 DOI: 10.1111/ajt.16928] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 11/26/2021] [Accepted: 11/26/2021] [Indexed: 01/25/2023]
Abstract
Unlimited organ availability would represent a paradigm shift in transplantation. Long-term in vivo engraftment and function of scaled-up bioengineered liver grafts have not been previously reported. In this study, we describe a human-scale transplantable liver graft engineered on a porcine liver-derived scaffold. We repopulated the scaffold parenchyma with primary hepatocytes and the vascular system with endothelial cells. For in vivo functional testing, we performed auxiliary transplantation of the repopulated scaffold in pigs with induced liver failure. It was observed that the auxiliary bioengineered liver graft improved liver function for 28 days and exhibited upregulation of liver-specific genes. This study is the first of its kind to present 28 days of posttransplant evaluation of a bioengineered liver graft using a preclinical large animal model. Furthermore, it provides definitive evidence for the feasibility of engineering human-scale transplantable liver grafts for clinical applications.
Collapse
Affiliation(s)
- Hisanobu Higashi
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Hiroshi Yagi
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Kohei Kuroda
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Kazuki Tajima
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan,Department of Small Animal Internal Medicine, Kitasato University School of Veterinary Medicine, Towada, Aomori, Japan
| | - Hideaki Kojima
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Kotaro Nishi
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Toshinori Morisaku
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Kazuya Hirukawa
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Kazumasa Fukuda
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Kentaro Matsubara
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Minoru Kitago
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Masahiro Shinoda
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Hideaki Obara
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Shungo Adachi
- Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Koto, Tokyo, Japan
| | - Kumiko Nishimura
- Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Koto, Tokyo, Japan
| | - Tohru Natsume
- Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Koto, Tokyo, Japan
| | - Masatoshi Tomi
- Division of Pharmaceutics, Faculty of Pharmacy, Keio University, Minato, Tokyo, Japan
| | - Alejandro Soto-Gutierrez
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA,McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, USA
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| |
Collapse
|
27
|
New Perspectives to Improve Mesenchymal Stem Cell Therapies for Drug-Induced Liver Injury. Int J Mol Sci 2022; 23:ijms23052669. [PMID: 35269830 PMCID: PMC8910533 DOI: 10.3390/ijms23052669] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 02/06/2023] Open
Abstract
Drug-induced liver injury (DILI) is one of the leading causes of acute liver injury. Many factors may contribute to the susceptibility of patients to this condition, making DILI a global medical problem that has an impact on public health and the pharmaceutical industry. The use of mesenchymal stem cells (MSCs) has been at the forefront of regenerative medicine therapies for many years, including MSCs for the treatment of liver diseases. However, there is currently a huge gap between these experimental approaches and their application in clinical practice. In this concise review, we focus on the pathophysiology of DILI and highlight new experimental approaches conceived to improve cell-based therapy by the in vitro preconditioning of MSCs and/or the use of cell-free products as treatment for this liver condition. Finally, we discuss the advantages of new approaches, but also the current challenges that must be addressed in order to develop safer and more effective procedures that will allow cell-based therapies to reach clinical practice, enhancing the quality of life and prolonging the survival time of patients with DILI.
Collapse
|
28
|
Wei X, Cui C, Fan C, Wu T, Li Y, Zhang X, Wang K, Pang Y, Yao P, Yang J. Injectable hydrogel based on dodecyl-modified N-carboxyethyl chitosan/oxidized konjac glucomannan effectively prevents bleeding and postoperative adhesions after partial hepatectomy. Int J Biol Macromol 2022; 199:401-412. [PMID: 34999041 DOI: 10.1016/j.ijbiomac.2021.12.193] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 12/18/2021] [Accepted: 12/30/2021] [Indexed: 11/27/2022]
Abstract
Hemostasis and prevention of postoperative adhesions after hepatectomy are still challenges. In this work, we chose chitosan, a competitive candidate hemostatic material, as the backbone, and konjac glucomannan as the functional moieties, to form an injectable hydrogel. The hydrogel was prepared by the Schiff base reaction of dodecyl-modified N-carboxyethyl chitosan (DCEC) and oxidized konjac glucomannan (OKGM), which could effectively prevent bleeding and postoperative adhesions. The resultant hydrogel possessed self-healing and tissue adhesive capability, and combined the unique bioactivities of two polysaccharides: DCEC endowed the hydrogel with excellent antibacterial and hemostatic ability by the electrostatic and hydrophobic interactions between the cell membrane and amine/dodecyl groups, and OKGM imparted hydrogel anti-inflammatory action by activating macrophages. Moreover, the notable hemostatic efficacy of the hydrogel was confirmed in a rat hepatectomy model. The hydrogel could prevent postoperative adhesions and down-regulate the inflammatory factor TNF-α and the pro-fibrotic factor TGF-β1 in situ, which might be caused by the combination of the barrier function of hydrogel and instinct bioactivities of DCEC and OKGM. Thus, this multifunctional injectable hydrogel is potentially valuable for preventing bleeding and postoperative adhesions after hepatectomy.
Collapse
Affiliation(s)
- Xiangyu Wei
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300350, China
| | - Chunyan Cui
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300350, China
| | - Chuanchuan Fan
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300350, China
| | - Tengling Wu
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300350, China
| | - Yuan Li
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300350, China
| | - Xiaoping Zhang
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300350, China
| | - Kuan Wang
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300350, China
| | - Yudi Pang
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300350, China
| | - Puqing Yao
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300350, China
| | - Jianhai Yang
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300350, China.
| |
Collapse
|
29
|
Fan W, Li Y, Kunimoto K, Török NJ. 3D Imaging of the Liver Extracellular Matrix in a Mouse Model of Non-Alcoholic Steatohepatitis. J Vis Exp 2022:10.3791/63106. [PMID: 35285832 PMCID: PMC9836498 DOI: 10.3791/63106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is the most common chronic liver disease in the United States, affecting more than 70 million Americans. NASH can progress to fibrosis and eventually to cirrhosis, a significant risk factor for hepatocellular carcinoma. The extracellular matrix (ECM) provides structural support and maintains liver homeostasis via matricellular signals. Liver fibrosis results from an imbalance in the dynamic ECM remodeling process and is characterized by excessive accumulation of structural elements and associated changes in glycosaminoglycans. The typical fibrosis pattern of NASH is called "chicken wire," which usually consists of zone 3 perisinusoidal/pericellular fibrosis, based on features observed by Masson's trichrome stain and Picrosirius Red stains. However, these traditional thin two-dimensional (2D) tissue slide-based imaging techniques cannot demonstrate the detailed three-dimensional (3D) ECM structural changes, limiting the understanding of the dynamic ECM remodeling in liver fibrosis. The current work optimized a fast and efficient protocol to image the native ECM structure in the liver via decellularization to address the above challenges. Mice were fed either with chow or fast-food diet for 14 weeks. Decellularization was performed after in situ portal vein perfusion, and the two-photon microscopy techniques were applied to image and analyze changes in the native ECM. The 3D images of the normal and NASH livers were reconstituted and analyzed. Performing in situ perfusion decellularization and analyzing the scaffold by two-photon microscopy provided a practical and reliable platform to visualize the dynamic ECM remodeling in the liver.
Collapse
Affiliation(s)
- Weiguo Fan
- Gastroenterology and Hepatology, Stanford University;
| | - Yuan Li
- Gastroenterology and Hepatology, Stanford University
| | | | | |
Collapse
|
30
|
Dai Q, Jiang W, Huang F, Song F, Zhang J, Zhao H. Recent Advances in Liver Engineering With Decellularized Scaffold. Front Bioeng Biotechnol 2022; 10:831477. [PMID: 35223793 PMCID: PMC8866951 DOI: 10.3389/fbioe.2022.831477] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/24/2022] [Indexed: 12/02/2022] Open
Abstract
Liver transplantation is currently the only effective treatment for patients with end-stage liver disease; however, donor liver scarcity is a notable concern. As a result, extensive endeavors have been made to diversify the source of donor livers. For example, the use of a decellularized scaffold in liver engineering has gained considerable attention in recent years. The decellularized scaffold preserves the original orchestral structure and bioactive chemicals of the liver, and has the potential to create a de novo liver that is fit for transplantation after recellularization. The structure of the liver and hepatic extracellular matrix, decellularization, recellularization, and recent developments are discussed in this review. Additionally, the criteria for assessment and major obstacles in using a decellularized scaffold are covered in detail.
Collapse
Affiliation(s)
- Qingqing Dai
- Department of Hepatopancreatobiliary Surgery and Organ Transplantation Center, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena, Germany
| | - Wei Jiang
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fan Huang
- Department of Hepatopancreatobiliary Surgery and Organ Transplantation Center, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fei Song
- Department of Urology, Jena University Hospital, Jena, Germany
| | - Jiqian Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Jiqian Zhang, ; Hongchuan Zhao,
| | - Hongchuan Zhao
- Department of Hepatopancreatobiliary Surgery and Organ Transplantation Center, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Jiqian Zhang, ; Hongchuan Zhao,
| |
Collapse
|
31
|
Decellularised extracellular matrix-based biomaterials for repair and regeneration of central nervous system. Expert Rev Mol Med 2022; 23:e25. [PMID: 34994341 PMCID: PMC9884794 DOI: 10.1017/erm.2021.22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The central nervous system (CNS), consisting of the brain and spinal cord, regulates the mind and functions of the organs. CNS diseases, leading to changes in neurological functions in corresponding sites and causing long-term disability, represent one of the major public health issues with significant clinical and economic burdens worldwide. In particular, the abnormal changes in the extracellular matrix under various disease conditions have been demonstrated as one of the main factors that can alter normal cell function and reduce the neuroregeneration potential in damaged tissue. Decellularised extracellular matrix (dECM)-based biomaterials have been recently utilised for CNS applications, closely mimicking the native tissue. dECM retains tissue-specific components, including proteoglycan as well as structural and functional proteins. Due to their unique composition, these biomaterials can stimulate sensitive repair mechanisms associated with CNS damages. Herein, we discuss the decellularisation of the brain and spinal cord as well as recellularisation of acellular matrix and the recent progress in the utilisation of brain and spinal cord dECM.
Collapse
|
32
|
Dias ML, Paranhos BA, Goldenberg RCDS. Liver scaffolds obtained by decellularization: A transplant perspective in liver bioengineering. J Tissue Eng 2022; 13:20417314221105305. [PMID: 35756167 PMCID: PMC9218891 DOI: 10.1177/20417314221105305] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/19/2022] [Indexed: 11/15/2022] Open
Abstract
Liver transplantation is the only definitive treatment for many diseases that affect this organ, however, its quantity and viability are reduced. The study of liver scaffolds based on an extracellular matrix is a tissue bioengineering strategy with great application in regenerative medicine. Collectively, recent studies suggest that liver scaffold transplantation may assist in reestablishing hepatic function in preclinical diseased animals, which represents a great potential for application as a treatment for patients with liver disease in the future. This review focuses on useful strategies to promote liver scaffold transplantation and the main open questions about this context. We outline the current knowledge about ex vivo bioengineered liver transplantation, including the surgical techniques, recipient survival time, scaffold preparation before transplantation, and liver disease models. We also highlight the current limitations and future directions regarding in vivo bioengineering techniques.
Collapse
Affiliation(s)
- Marlon Lemos Dias
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa - INCT - REGENERA, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Bruno Andrade Paranhos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa - INCT - REGENERA, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Regina Coeli Dos Santos Goldenberg
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa - INCT - REGENERA, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| |
Collapse
|
33
|
Lock R, Al Asafen H, Fleischer S, Tamargo M, Zhao Y, Radisic M, Vunjak-Novakovic G. A framework for developing sex-specific engineered heart models. NATURE REVIEWS. MATERIALS 2021; 7:295-313. [PMID: 34691764 PMCID: PMC8527305 DOI: 10.1038/s41578-021-00381-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/20/2021] [Indexed: 05/02/2023]
Abstract
The convergence of tissue engineering and patient-specific stem cell biology has enabled the engineering of in vitro tissue models that allow the study of patient-tailored treatment modalities. However, sex-related disparities in health and disease, from systemic hormonal influences to cellular-level differences, are often overlooked in stem cell biology, tissue engineering and preclinical screening. The cardiovascular system, in particular, shows considerable sex-related differences, which need to be considered in cardiac tissue engineering. In this Review, we analyse sex-related properties of the heart muscle in the context of health and disease, and discuss a framework for including sex-based differences in human cardiac tissue engineering. We highlight how sex-based features can be implemented at the cellular and tissue levels, and how sex-specific cardiac models could advance the study of cardiovascular diseases. Finally, we define design criteria for sex-specific cardiac tissue engineering and provide an outlook to future research possibilities beyond the cardiovascular system.
Collapse
Affiliation(s)
- Roberta Lock
- Department of Biomedical Engineering, Columbia University, New York, NY USA
| | - Hadel Al Asafen
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario Canada
| | - Sharon Fleischer
- Department of Biomedical Engineering, Columbia University, New York, NY USA
| | - Manuel Tamargo
- Department of Biomedical Engineering, Columbia University, New York, NY USA
| | - Yimu Zhao
- Department of Biomedical Engineering, Columbia University, New York, NY USA
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario Canada
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY USA
- Department of Medicine, Columbia University, New York, NY USA
| |
Collapse
|
34
|
Pluta KD, Ciezkowska M, Wisniewska M, Wencel A, Pijanowska DG. Cell-based clinical and experimental methods for assisting the function of impaired livers – Present and future of liver support systems. Biocybern Biomed Eng 2021. [DOI: 10.1016/j.bbe.2021.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
35
|
Khajavi M, Hashemi M, Kalalinia F. Recent advances in optimization of liver decellularization procedures used for liver regeneration. Life Sci 2021; 281:119801. [PMID: 34229008 DOI: 10.1016/j.lfs.2021.119801] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 06/19/2021] [Accepted: 06/29/2021] [Indexed: 10/20/2022]
Abstract
Severe liver diseases have been considered the most common causes of adult deaths worldwide. Until now, liver transplantation is known as the only effective treatment for end stage liver disease. However, it is associated with several problems, most importantly, the side effects of immunosuppressive drugs that should be used after transplantation, and the shortage of tissue donors compared to the increasing number of patients requiring liver transplantation. Currently, tissue/organ decellularization as a new approach in tissue engineering is becoming a valid substitute for managing these kinds of problems. Decellularization of a whole liver is an attractive procedure to create three-dimensional (3D) scaffolds that micro-architecturally and structurally are similar to the native one and could support the repair or replacement of damaged or injured tissue. In this review, the different methods used for decellularization of liver tissue have been reviewed. In addition, the current approaches to overcome the challenges in these techniques are discussed.
Collapse
Affiliation(s)
- Mohaddeseh Khajavi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Hashemi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Kalalinia
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
36
|
Cell Therapy and Bioengineering in Experimental Liver Regenerative Medicine: In Vivo Injury Models and Grafting Strategies. CURRENT TRANSPLANTATION REPORTS 2021. [DOI: 10.1007/s40472-021-00325-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Abstract
Purpose of Review
To describe experimental liver injury models used in regenerative medicine, cell therapy strategies to repopulate damaged livers and the efficacy of liver bioengineering.
Recent Findings
Several animal models have been developed to study different liver conditions. Multiple strategies and modified protocols of cell delivery have been also reported. Furthermore, using bioengineered liver scaffolds has shown promising results that could help in generating a highly functional cell delivery system and/or a whole transplantable liver.
Summary
To optimize the most effective strategies for liver cell therapy, further studies are required to compare among the performed strategies in the literature and/or innovate a novel modifying technique to overcome the potential limitations. Coating of cells with polymers, decellularized scaffolds, or microbeads could be the most appropriate solution to improve cellular efficacy. Besides, overcoming the problems of liver bioengineering may offer a radical treatment for end-stage liver diseases.
Collapse
|
37
|
Padma AM, Carrière L, Krokström Karlsson F, Sehic E, Bandstein S, Tiemann TT, Oltean M, Song MJ, Brännström M, Hellström M. Towards a bioengineered uterus: bioactive sheep uterus scaffolds are effectively recellularized by enzymatic preconditioning. NPJ Regen Med 2021; 6:26. [PMID: 34021161 PMCID: PMC8140118 DOI: 10.1038/s41536-021-00136-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 04/20/2021] [Indexed: 12/23/2022] Open
Abstract
Uterine factor infertility was considered incurable until recently when we reported the first successful live birth after uterus transplantation. However, risky donor surgery and immunosuppressive therapy are factors that may be avoided with bioengineering. For example, transplanted recellularized constructs derived from decellularized tissue restored fertility in rodent models and mandate translational studies. In this study, we decellularized whole sheep uterus with three different protocols using 0.5% sodium dodecyl sulfate, 2% sodium deoxycholate (SDC) or 2% SDC, and 1% Triton X-100. Scaffolds were then assessed for bioactivity using the dorsal root ganglion and chorioallantoic membrane assays, and we found that all the uterus scaffolds exhibited growth factor activity that promoted neurogenesis and angiogenesis. Extensive recellularization optimization was conducted using multipotent sheep fetal stem cells and we report results from the following three in vitro conditions; (a) standard cell culturing conditions, (b) constructs cultured in transwells, and (c) scaffolds preconditioned with matrix metalloproteinase 2 and 9. The recellularization efficiency was improved short-term when transwells were used compared with standard culturing conditions. However, the recellularization efficiency in scaffolds preconditioned with matrix metalloproteinases was 200–300% better than the other strategies evaluated herein, independent of decellularization protocol. Hence, a major recellularization hurdle has been overcome with the improved recellularization strategies and in vitro platforms described herein. These results are an important milestone and should facilitate the production of large bioengineered grafts suitable for future in vivo applications in the sheep, which is an essential step before considering these principles in a clinical setting.
Collapse
Affiliation(s)
- Arvind Manikantan Padma
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Obstetrics and Gynecology, Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Laura Carrière
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Obstetrics and Gynecology, Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Frida Krokström Karlsson
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Obstetrics and Gynecology, Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Edina Sehic
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Obstetrics and Gynecology, Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sara Bandstein
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Obstetrics and Gynecology, Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tom Tristan Tiemann
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Obstetrics and Gynecology, Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Gynecology and Obstetrics, University Hospital of Heidelberg, Heidelberg, Germany
| | - Mihai Oltean
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Surgery, Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Min Jong Song
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Obstetrics and Gynecology, Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Obstetrics and Gynecology, Yeouido St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mats Brännström
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Obstetrics and Gynecology, Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Stockholm IVF-EUGIN, Hammarby allé 93, Stockholm, Sweden
| | - Mats Hellström
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden. .,Department of Obstetrics and Gynecology, Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
38
|
Park Y, Huh KM, Kang SW. Applications of Biomaterials in 3D Cell Culture and Contributions of 3D Cell Culture to Drug Development and Basic Biomedical Research. Int J Mol Sci 2021; 22:2491. [PMID: 33801273 PMCID: PMC7958286 DOI: 10.3390/ijms22052491] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 01/10/2023] Open
Abstract
The process of evaluating the efficacy and toxicity of drugs is important in the production of new drugs to treat diseases. Testing in humans is the most accurate method, but there are technical and ethical limitations. To overcome these limitations, various models have been developed in which responses to various external stimuli can be observed to help guide future trials. In particular, three-dimensional (3D) cell culture has a great advantage in simulating the physical and biological functions of tissues in the human body. This article reviews the biomaterials currently used to improve cellular functions in 3D culture and the contributions of 3D culture to cancer research, stem cell culture and drug and toxicity screening.
Collapse
Affiliation(s)
- Yujin Park
- Department of Polymer Science and Engineering & Chemical Engineering and Applied Chemistry, Chungnam National University, Daejeon 34134, Korea;
- Predictive Model Research Center, Korea Institute of Toxicology, Daejeon 34114, Korea
| | - Kang Moo Huh
- Department of Polymer Science and Engineering & Chemical Engineering and Applied Chemistry, Chungnam National University, Daejeon 34134, Korea;
| | - Sun-Woong Kang
- Predictive Model Research Center, Korea Institute of Toxicology, Daejeon 34114, Korea
- Human and Environmental Toxicology Program, University of Science and Technology, Daejeon 34114, Korea
| |
Collapse
|
39
|
Ali M, Payne SL. Biomaterial-based cell delivery strategies to promote liver regeneration. Biomater Res 2021; 25:5. [PMID: 33632335 PMCID: PMC7905561 DOI: 10.1186/s40824-021-00206-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/05/2021] [Indexed: 02/08/2023] Open
Abstract
Chronic liver disease and cirrhosis is a widespread and untreatable condition that leads to lifelong impairment and eventual death. The scarcity of liver transplantation options requires the development of new strategies to attenuate disease progression and reestablish liver function by promoting regeneration. Biomaterials are becoming an increasingly promising option to both culture and deliver cells to support in vivo viability and long-term function. There is a wide variety of both natural and synthetic biomaterials that are becoming established as delivery vehicles with their own unique advantages and disadvantages for liver regeneration. We review the latest developments in cell transplantation strategies to promote liver regeneration, with a focus on the use of both natural and synthetic biomaterials for cell culture and delivery. We conclude that future work will need to refine the use of these biomaterials and combine them with novel strategies that recapitulate liver organization and function in order to translate this strategy to clinical use.
Collapse
Affiliation(s)
- Maqsood Ali
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, South Korea
| | - Samantha L Payne
- Department of Biomedical Engineering, School of Engineering, Tufts University, Medford, MA, 02155, USA.
| |
Collapse
|
40
|
Bate TSR, Gadd VL, Forbes SJ, Callanan A. Response differences of HepG2 and Primary Mouse Hepatocytes to morphological changes in electrospun PCL scaffolds. Sci Rep 2021; 11:3059. [PMID: 33542251 PMCID: PMC7862353 DOI: 10.1038/s41598-021-81761-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 12/02/2020] [Indexed: 01/30/2023] Open
Abstract
Liver disease cases are rapidly expanding across the globe and the only effective cure for end-stage disease is a transplant. Transplant procedures are costly and current supply of donor livers does not satisfy demand. Potential drug treatments and regenerative therapies that are being developed to tackle these pressing issues require effective in-vitro culture platforms. Electrospun scaffolds provide bio-mimetic structures upon which cells are cultured to regulate function in-vitro. This study aims to shed light on the effects of electrospun PCL morphology on the culture of an immortalised hepatic cell line and mouse primary hepatocytes. Each cell type was cultured on large 4-5 µm fibres and small 1-2 µm fibres with random, aligned and highly porous cryogenically spun configurations. Cell attachment, proliferation, morphology and functional protein and gene expression was analysed. Results show that fibre morphology has a measurable influence on cellular morphology and function, with the alteration of key functional markers such as CYP1A2 expression.
Collapse
Affiliation(s)
- Thomas S R Bate
- Institute for Bioengineering, School of Engineering, University of Edinburgh, Edinburgh, UK
| | - Victoria L Gadd
- Scottish Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Stuart J Forbes
- Scottish Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Anthony Callanan
- Institute for Bioengineering, School of Engineering, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
41
|
Pennarossa G, Arcuri S, De Iorio T, Gandolfi F, Brevini TAL. Current Advances in 3D Tissue and Organ Reconstruction. Int J Mol Sci 2021; 22:E830. [PMID: 33467648 PMCID: PMC7830719 DOI: 10.3390/ijms22020830] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/31/2020] [Accepted: 01/13/2021] [Indexed: 12/11/2022] Open
Abstract
Bi-dimensional culture systems have represented the most used method to study cell biology outside the body for over a century. Although they convey useful information, such systems may lose tissue-specific architecture, biomechanical effectors, and biochemical cues deriving from the native extracellular matrix, with significant alterations in several cellular functions and processes. Notably, the introduction of three-dimensional (3D) platforms that are able to re-create in vitro the structures of the native tissue, have overcome some of these issues, since they better mimic the in vivo milieu and reduce the gap between the cell culture ambient and the tissue environment. 3D culture systems are currently used in a broad range of studies, from cancer and stem cell biology, to drug testing and discovery. Here, we describe the mechanisms used by cells to perceive and respond to biomechanical cues and the main signaling pathways involved. We provide an overall perspective of the most recent 3D technologies. Given the breadth of the subject, we concentrate on the use of hydrogels, bioreactors, 3D printing and bioprinting, nanofiber-based scaffolds, and preparation of a decellularized bio-matrix. In addition, we report the possibility to combine the use of 3D cultures with functionalized nanoparticles to obtain highly predictive in vitro models for use in the nanomedicine field.
Collapse
Affiliation(s)
- Georgia Pennarossa
- Laboratory of Biomedical Embryology, Department of Health, Animal Science and Food Safety and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; (G.P.); (S.A.); (T.D.I.)
| | - Sharon Arcuri
- Laboratory of Biomedical Embryology, Department of Health, Animal Science and Food Safety and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; (G.P.); (S.A.); (T.D.I.)
| | - Teresina De Iorio
- Laboratory of Biomedical Embryology, Department of Health, Animal Science and Food Safety and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; (G.P.); (S.A.); (T.D.I.)
| | - Fulvio Gandolfi
- Department of Agricultural and Environmental Sciences—Production, Landscape, Agroenergy and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 2, 20133 Milan, Italy;
| | - Tiziana A. L. Brevini
- Laboratory of Biomedical Embryology, Department of Health, Animal Science and Food Safety and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; (G.P.); (S.A.); (T.D.I.)
| |
Collapse
|
42
|
Huang Y, Miyamoto D, Hidaka M, Adachi T, Gu WL, Eguchi S. Regenerative medicine for the hepatobiliary system: A review. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2020; 28:913-930. [PMID: 33314713 DOI: 10.1002/jhbp.882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/05/2020] [Accepted: 11/20/2020] [Indexed: 12/16/2022]
Abstract
Liver transplantation, the only proven treatment for end-stage liver disease and acute liver failure, is hampered by the scarcity of donors. Regenerative medicine provides an alternative therapeutic approach. Tremendous efforts dedicated to liver regenerative medicine include the delivery of transplantable cells, microtissues, and bioengineered whole livers via tissue engineering and the maintenance of partial liver function via extracorporeal support. This brief review summarizes the current status of regenerative medicine for the hepatobiliary system. For liver regenerative medicine, the focus is on strategies for expansion of transplantable hepatocytes, generation of hepatocyte-like cells, and therapeutic potential of engineered tissues in liver disease models. For biliary regenerative medicine, the discussion concentrates on the methods for generation of cholangiocyte-like cells and strategies in the treatment of biliary disease. Significant advances have been made in large-scale and long-term expansion of liver cells. The development of tissue engineering and stem cell induction technology holds great promise for the future treatment of hepatobiliary diseases. The application of regenerative medicine in liver still lacks extensive animal experiments. Therefore, a large number of preclinical studies are necessary to provide sufficient evidence for their therapeutic effectiveness. Much remains to be done for the treatment of hepatobiliary diseases with regenerative medicine.
Collapse
Affiliation(s)
- Yu Huang
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Department of Surgery, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangdong, China
| | - Daisuke Miyamoto
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Masaaki Hidaka
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tomohiko Adachi
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Wei-Li Gu
- Department of Surgery, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangdong, China
| | - Susumu Eguchi
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
43
|
Dias ML, Batista CMP, Secomandi VJK, Silva AC, Monteiro VRS, Faccioli LA, Goldenberg RCS. Surgical Models to Explore Acellular Liver Scaffold Transplantation: Step-by-Step. Organogenesis 2020; 16:95-112. [PMID: 32799604 DOI: 10.1080/15476278.2020.1801273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Acellular liver scaffolds (ALS) have arisen as potential candidates for transplantation. Until now, all reports involving ALS transplantation failed in surgical method descriptions and do not offer support to scientists to reproduce the procedures used in experimental microsurgery to make the results comparable to literature. To overcome the lack of detail information, we described surgical steps details to perform heterotopic and partial orthotopic surgical models to promote ALS transplantation. After preservation and vessel cannulation steps, the liver grafts were decellularized. In addition, ex vivo blood perfusion tests were performed to obtain a successful anticoagulation treatment prior in vivo transplantation. Then, methods of partial liver resection, combination of hand-suture and cuff techniques to complete end-to-end anastomosis between the scaffold and the recipient animal were performed. These procedures which take 30-60 min and were efficient to allow acellular liver scaffold viability and recellularization of different types of cell post-surgery. In conclusion, our methods are practical and simple promising approach that provides the opportunity to investigate ways to achieve sufficient liver function post-transplantation in vivo.
Collapse
Affiliation(s)
- Marlon L Dias
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio De Janeiro , Rio De Janeiro, Brazil
| | - Cíntia M P Batista
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio De Janeiro , Rio De Janeiro, Brazil
| | - Victor J K Secomandi
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio De Janeiro , Rio De Janeiro, Brazil
| | - Alexandre C Silva
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio De Janeiro , Rio De Janeiro, Brazil.,Department of Surgery, Clementino Fraga Filho Universitary Hospital, Federal University of Rio De Janeiro , Rio De Janeiro, Brazil
| | - Victoria R S Monteiro
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio De Janeiro , Rio De Janeiro, Brazil
| | - Lanuza A Faccioli
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio De Janeiro , Rio De Janeiro, Brazil
| | - Regina C S Goldenberg
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio De Janeiro , Rio De Janeiro, Brazil.,National Institute of Science and Technology in Regenerative Medicine- REGENERA, Federal University of Rio De Janeiro , Rio De Janeiro, Brazil
| |
Collapse
|
44
|
Nevzorova YA, Boyer-Diaz Z, Cubero FJ, Gracia-Sancho J. Animal models for liver disease - A practical approach for translational research. J Hepatol 2020; 73:423-440. [PMID: 32330604 DOI: 10.1016/j.jhep.2020.04.011] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 12/11/2022]
Abstract
Animal models are crucial for improving our understanding of human pathogenesis, enabling researchers to identify therapeutic targets and test novel drugs. In the current review, we provide a comprehensive summary of the most widely used experimental models of chronic liver disease, starting from early stages of fatty liver disease (non-alcoholic and alcoholic) to steatohepatitis, advanced cirrhosis and end-stage primary liver cancer. We focus on aspects such as reproducibility and practicality, discussing the advantages and weaknesses of available models for researchers who are planning to perform animal studies in the near future. Additionally, we summarise current and prospective models based on human tissue bioengineering.
Collapse
Affiliation(s)
- Yulia A Nevzorova
- Department of Genetics, Physiology and Microbiology, Faculty of Biology, Complutense University, Madrid, Spain; 12 de Octubre Health Research Institute (imas12), Madrid, Spain; Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Zoe Boyer-Diaz
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Unit, IDIBAPS Biomedical Research Institute, Barcelona, Spain; Barcelona Liver Bioservices, Barcelona, Spain
| | - Francisco Javier Cubero
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain; Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain.
| | - Jordi Gracia-Sancho
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Unit, IDIBAPS Biomedical Research Institute, Barcelona, Spain; Barcelona Liver Bioservices, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain; Hepatology, Department of Biomedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
45
|
Saleh T, Ahmed E, Yu L, Song SH, Park KM, Kwak HH, Woo HM. Conjugating homogenized liver-extracellular matrix into decellularized hepatic scaffold for liver tissue engineering. J Biomed Mater Res A 2020; 108:1991-2004. [PMID: 32180336 DOI: 10.1002/jbm.a.36920] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/29/2020] [Accepted: 03/09/2020] [Indexed: 12/13/2022]
Abstract
The generation of a transplantable liver scaffold is crucial for the treatment of end-stage liver failure. Unfortunately, decellularized liver scaffolds suffer from lack of bioactive molecules and functionality. In this study, we conjugated homogenized liver-extracellular matrix (ECM) into a decellularized liver in a rat model to improve its structural and functional properties. The homogenized ECM was prepared, characterized, and subsequently perfused into ethyl carbodiimide hydrochloride (EDC)/N-hydroxysuccinimide (NHS) activated liver scaffolds. Various techniques were performed to confirm the improvements that were accomplished through the conjugation process; these included micro/ultra-structural analyses, biochemical analysis of ECM components, DNA quantification, swelling ratio, structural stability, calcification properties, platelet activation study, static and dynamic seeding with EAhy926 endothelial cells and HepG2 hepatocarcinoma cells, subcutaneous implantation and intrahepatic transplantation. The results showed that the conjugated scaffolds have superior micro- and ultrastructural and biochemical characteristics. In addition, DNA contents, swelling ratios, calcification properties, platelet reactions, and host inflammatory reactions were not altered with the conjugation process. The conjugated scaffolds revealed better cellular spreading and popularity compared to the non-conjugated scaffolds. Intrahepatic transplantation showed that the conjugated scaffold had higher popularity of hepatic regenerative cells with better angiogenesis. The conjugation of the decellularized liver scaffold with homogenized liver-ECM is a promising tool to improve the quality of the generated scaffold for further transplantation.
Collapse
Affiliation(s)
- Tarek Saleh
- Department of Veterinary Science, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Ebtehal Ahmed
- Department of Veterinary Science, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Lina Yu
- Department of Veterinary Science, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Su-Hyeon Song
- Department of Veterinary Science, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Kyung-Mee Park
- College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Ho-Hyun Kwak
- Department of Veterinary Science, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Heung-Myong Woo
- Department of Veterinary Science, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Republic of Korea
| |
Collapse
|
46
|
Gao Y, Li Z, Hong Y, Li T, Hu X, Sun L, Chen Z, Chen Z, Luo Z, Wang X, Kong J, Li G, Wang HL, Leo HL, Yu H, Xi L, Guo Q. Decellularized liver as a translucent ex vivo model for vascular embolization evaluation. Biomaterials 2020; 240:119855. [DOI: 10.1016/j.biomaterials.2020.119855] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 02/06/2020] [Accepted: 02/08/2020] [Indexed: 12/25/2022]
|
47
|
Kuipers ME, Nolte-'t Hoen ENM, van der Ham AJ, Ozir-Fazalalikhan A, Nguyen DL, de Korne CM, Koning RI, Tomes JJ, Hoffmann KF, Smits HH, Hokke CH. DC-SIGN mediated internalisation of glycosylated extracellular vesicles from Schistosoma mansoni increases activation of monocyte-derived dendritic cells. J Extracell Vesicles 2020; 9:1753420. [PMID: 32489529 PMCID: PMC7241508 DOI: 10.1080/20013078.2020.1753420] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/20/2020] [Accepted: 03/24/2020] [Indexed: 12/13/2022] Open
Abstract
Helminths like Schistosoma mansoni release excretory/secretory (E/S) products that modulate host immunity to enable infection. Extracellular vesicles (EVs) are among these E/S products, yet molecular mechanisms and functionality of S. mansoni EV interaction with host immune cells is unknown. Here we demonstrate that EVs released by S. mansoni schistosomula are internalised by human monocyte-derived dendritic cells (moDCs). Importantly, we show that this uptake was mainly mediated via DC-SIGN (CD209). Blocking DC-SIGN almost completely abrogated EV uptake, while blocking mannose receptor (MR, CD206) or dendritic cell immunoreceptor (DCIR, CLEC4A) had no effect on EV uptake. Mass spectrometric analysis of EV glycans revealed the presence of surface N-glycans with terminal Galβ1-4(Fucα1-3)GlcNAc (LewisX) motifs, and a wide array of fucosylated lipid-linked glycans, including LewisX, a known ligand for DC-SIGN. Stimulation of moDCs with schistosomula EVs led to increased expression of costimulatory molecules CD86 and CD80 and regulatory surface marker PD-L1. Furthermore, schistosomula EVs increased expression of IL-12 and IL-10 by moDCs, which was partly dependent on the interaction with DC-SIGN. These results provide the first evidence that glycosylation of S. mansoni EVs facilitates the interaction with host immune cells and reveals a role for DC-SIGN and EV-associated glycoconjugates in parasite-induced immune modulation.
Collapse
Affiliation(s)
- Marije E Kuipers
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands.,Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Esther N M Nolte-'t Hoen
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Alwin J van der Ham
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | | | - D Linh Nguyen
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Clarize M de Korne
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Roman I Koning
- Department of Cell & Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - John J Tomes
- Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth University, Aberystwyth, UK
| | - Karl F Hoffmann
- Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth University, Aberystwyth, UK
| | - Hermelijn H Smits
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Cornelis H Hokke
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
48
|
Cernigliaro V, Peluso R, Zedda B, Silengo L, Tolosano E, Pellicano R, Altruda F, Fagoonee S. Evolving Cell-Based and Cell-Free Clinical Strategies for Treating Severe Human Liver Diseases. Cells 2020; 9:386. [PMID: 32046114 PMCID: PMC7072646 DOI: 10.3390/cells9020386] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/21/2020] [Accepted: 02/06/2020] [Indexed: 02/07/2023] Open
Abstract
Liver diseases represent a major global health issue, and currently, liver transplantation is the only viable alternative to reduce mortality rates in patients with end-stage liver diseases. However, scarcity of donor organs and risk of recidivism requiring a re-transplantation remain major obstacles. Hence, much hope has turned towards cell-based therapy. Hepatocyte-like cells obtained from embryonic stem cells or adult stem cells bearing multipotent or pluripotent characteristics, as well as cell-based systems, such as organoids, bio-artificial liver devices, bioscaffolds and organ printing are indeed promising. New approaches based on extracellular vesicles are also being investigated as cell substitutes. Extracellular vesicles, through the transfer of bioactive molecules, can modulate liver regeneration and restore hepatic function. This review provides an update on the current state-of-art cell-based and cell-free strategies as alternatives to liver transplantation for patients with end-stage liver diseases.
Collapse
Affiliation(s)
- Viviana Cernigliaro
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy; (V.C.); (R.P.); (B.Z.)
- Maria Pia Hospital, 10126 Turin, Italy
| | - Rossella Peluso
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy; (V.C.); (R.P.); (B.Z.)
- Maria Pia Hospital, 10126 Turin, Italy
| | - Beatrice Zedda
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy; (V.C.); (R.P.); (B.Z.)
- Maria Pia Hospital, 10126 Turin, Italy
| | - Lorenzo Silengo
- Molecular Biotechnology Center, Departmet of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy; (L.S.); (E.T.)
| | - Emanuela Tolosano
- Molecular Biotechnology Center, Departmet of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy; (L.S.); (E.T.)
| | | | - Fiorella Altruda
- Molecular Biotechnology Center, Departmet of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy; (L.S.); (E.T.)
| | - Sharmila Fagoonee
- Institute of Biostructure and Bioimaging, National Research Council, Molecular Biotechnology Center, Via Nizza 52, 10126 Turin, Italy
| |
Collapse
|