1
|
Gupta S, Sharma A, Rajakannu M, Bisevac J, Rela M, Verma RS. Small Molecule-Mediated Stage-Specific Reprogramming of MSCs to Hepatocyte-Like Cells and Hepatic Tissue for Liver Injury Treatment. Stem Cell Rev Rep 2024; 20:2215-2235. [PMID: 39259445 PMCID: PMC11554881 DOI: 10.1007/s12015-024-10771-x] [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] [Accepted: 08/04/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Derivation of hepatocytes from stem cells has been established through various protocols involving growth factor (GF) and small molecule (SM) agents, among others. However, mesenchymal stem cell-based derivation of hepatocytes still remains expensive due to the use of a cocktail of growth factors, and a long duration of differentiation is needed, thus limiting its potential clinical application. METHODS In this study, we developed a chemically defined differentiation strategy that is exclusively based on SM and takes 14 days, while the GF-based protocol requires 23-28 days. RESULTS We optimized a stage-specific differentiation protocol for the differentiation of rat bone marrow-derived mesenchymal stem cells (MSCs) into functional hepatocyte-like cells (dHeps) that involved four stages, i.e., definitive endoderm (DE), hepatic competence (HC), hepatic specification (HS) and hepatic differentiation and growth. We further generated hepatic tissue using human decellularized liver extracellular matrix and compared it with hepatic tissue derived from the growth factor-based protocol at the transcriptional level. dHep, upon transplantation in a rat model of acute liver injury (ALI), was capable of ameliorating liver injury in rats and improving liver function and tissue damage compared to those in the ALI model. CONCLUSIONS In summary, this is the first study in which hepatocytes and hepatic tissue were derived from MSCs utilizing a stage-specific strategy by exclusively using SM as a differentiation factor.
Collapse
Affiliation(s)
- Santosh Gupta
- Stem Cell and Molecular Biology, Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu, 600036, India.
- Centre for Eye Research and Innovative Diagnostics, Department of Ophthalmology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Akriti Sharma
- Stem Cell and Molecular Biology, Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu, 600036, India
| | - Muthukumarassamy Rajakannu
- The Institute of Liver Disease & Transplantation, Dr. Rela Institute & Medical Centre, Bharath Institute of Higher Education & Research, Chromepet, Tamil Nadu, India
| | - Jovana Bisevac
- Centre for Eye Research and Innovative Diagnostics, Department of Ophthalmology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Mohamed Rela
- The Institute of Liver Disease & Transplantation, Dr. Rela Institute & Medical Centre, Bharath Institute of Higher Education & Research, Chromepet, Tamil Nadu, India
| | - Rama Shanker Verma
- Stem Cell and Molecular Biology, Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu, 600036, India.
| |
Collapse
|
2
|
Gao Y, Gadd VL, Heim M, Grant R, Bate TSR, Esser H, Gonzalez SF, Man TY, Forbes SJ, Callanan A. Combining human liver ECM with topographically featured electrospun scaffolds for engineering hepatic microenvironment. Sci Rep 2024; 14:23192. [PMID: 39369012 PMCID: PMC11455933 DOI: 10.1038/s41598-024-73827-5] [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: 05/29/2024] [Accepted: 09/20/2024] [Indexed: 10/07/2024] Open
Abstract
Liver disease cases are rapidly expanding worldwide, and transplantation remains the only effective cure for end-stage disease. There is an increasing demand for developing potential drug treatments, and regenerative therapies using in-vitro culture platforms. Human decellularized extracellular matrix (dECM) is an appealing alternative to conventional animal tissues as it contains human-specific proteins and can serve as scaffolding materials. Herein we exploit this with human donor tissue from discarded liver which was not suitable for transplant using a synergistic approach to combining biological and topographical cues in electrospun materials as an in-vitro culture platform. To realise this, we developed a methodology for incorporating human liver dECM into electrospun polycaprolactone (PCL) fibres with surface nanotopographies (230-580 nm). The hybrid scaffolds were fabricated using varying concentrations of dECM; their morphology, mechanical properties, hydrophilicity and stability were analysed. The scaffolds were validated using HepG2 and primary mouse hepatocytes, with subsequent results indicating that the modified scaffolds-maintained cell growth and influenced cell attachment, proliferation and hepatic-related gene expression. This work demonstrates a novel approach to harvesting the potential from decellularized human tissues in the form of innovative in-vitro culture platforms for liver.
Collapse
Affiliation(s)
- Yunxi Gao
- Institute for Bioengineering, School of Engineering, University of Edinburgh, Edinburgh, UK
- Foundation of Liver Research, The Roger Williams Institute of Liver Study, London, UK
| | - Victoria L Gadd
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Maria Heim
- Institute for Bioengineering, School of Engineering, University of Edinburgh, Edinburgh, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Rhiannon Grant
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Thomas S R Bate
- Institute for Bioengineering, School of Engineering, University of Edinburgh, Edinburgh, UK
- Vanderbilt University Medical Center, Nashville, USA
| | - Hannah Esser
- Institute for Bioengineering, School of Engineering, University of Edinburgh, Edinburgh, UK
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Sofia Ferreira Gonzalez
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Tak Yung Man
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Stuart J Forbes
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Anthony Callanan
- Institute for Bioengineering, School of Engineering, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
3
|
Arteel GE. Hepatic Extracellular Matrix and Its Role in the Regulation of Liver Phenotype. Semin Liver Dis 2024; 44:343-355. [PMID: 39191427 DOI: 10.1055/a-2404-7973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
The hepatic extracellular matrix (ECM) is most accurately depicted as a dynamic compartment that comprises a diverse range of players that work bidirectionally with hepatic cells to regulate overall homeostasis. Although the classic meaning of the ECM referred to only proteins directly involved in generating the ECM structure, such as collagens, proteoglycans, and glycoproteins, the definition of the ECM is now broader and includes all components associated with this compartment. The ECM is critical in mediating phenotype at the cellular, organ, and even organismal levels. The purpose of this review is to summarize the prevailing mechanisms by which ECM mediates hepatic phenotype and discuss the potential or established role of this compartment in the response to hepatic injury in the context of steatotic liver disease.
Collapse
Affiliation(s)
- Gavin E Arteel
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
4
|
Guo WY, Wang WH, Xu PY, Kankala RK, Chen AZ. Decellularised extracellular matrix-based injectable hydrogels for tissue engineering applications. BIOMATERIALS TRANSLATIONAL 2024; 5:114-128. [PMID: 39351160 PMCID: PMC11438603 DOI: 10.12336/biomatertransl.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/17/2024] [Accepted: 05/30/2024] [Indexed: 10/04/2024]
Abstract
Decellularised extracellular matrix (dECM) is a biomaterial derived from natural tissues that has attracted considerable attention from tissue engineering researchers due to its exceptional biocompatibility and malleability attributes. These advantageous properties often facilitate natural cell infiltration and tissue reconstruction for regenerative medicine. Due to their excellent fluidity, the injectable hydrogels can be administered in a liquid state and subsequently formed into a gel state in vivo, stabilising the target area and serving in a variety of ways, such as support, repair, and drug release functions. Thus, dECM-based injectable hydrogels have broad prospects for application in complex organ structures and various tissue injury models. This review focuses on exploring research advances in dECM-based injectable hydrogels, primarily focusing on the applications and prospects of dECM hydrogels in tissue engineering. Initially, the recent developments of the dECM-based injectable hydrogels are explained, summarising the different preparation methods with the evaluation of injectable hydrogel properties. Furthermore, some specific examples of the applicability of dECM-based injectable hydrogels are presented. Finally, we summarise the article with interesting prospects and challenges of dECM-based injectable hydrogels, providing insights into the development of these composites in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Wan-Ying Guo
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian Province, China
| | - Wei-Huang Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian Province, China
| | - Pei-Yao Xu
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian Province, China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian Province, China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian Province, China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian Province, China
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian Province, China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian Province, China
| |
Collapse
|
5
|
Li Y, Zhang Y, Zhong K, Liao S, Zhang G. The Development of a 3D PET Fibrous Scaffold Modified with an Umbilical Cord dECM for Liver Tissue Engineering. Polymers (Basel) 2024; 16:1794. [PMID: 39000651 PMCID: PMC11243929 DOI: 10.3390/polym16131794] [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: 05/10/2024] [Revised: 06/07/2024] [Accepted: 06/17/2024] [Indexed: 07/17/2024] Open
Abstract
Organ and tissue dysfunction represents a clinically significant condition. By integrating cell biology with materials science, tissue engineering enables the reconstruction and restoration of damaged tissues or organs, offering a noninvasive repair approach. In our study, we replicated the cellular growth environment by utilizing a human umbilical cord-derived decellularized extracellular matrix (dECM) as a modifying agent for the polyethylene terephthalate (PET) polymeric fiber scaffold. This allowed us to create a dECM-coated polyester fiber-based scaffold, PET-dECM, tailored for liver tissue engineering purposes. We effectively produced a decellularized human umbilical cord-derived ECM through a combined decellularization process involving trypsin/EDTA, TritonX-100, and sodium deoxycholate. The application of the dECM coating onto the PET material was accomplished through several steps, such as ester hydrolysis, EDC/NHS-activated crosslinking, and dECM conjugation. The biological performance of the PET-dECM was validated using RG cell culture assays. Notably, the dECM coating significantly improved PET's hydrophilicity and biocompatibility, thereby aiding cell adhesion, proliferation, and functional differentiation (p < 0.05). It was further found that the hepatocyte function of HepaRG was significantly enhanced on the PET-dECM, which may be attributed to the dECM's ability to facilitate the restoration of cell polarity. The PET-dECM holds promise as an effective hepatocyte culture carrier and could potentially find application in liver tissue engineering.
Collapse
Affiliation(s)
- Yang Li
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical and Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
- Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yang Zhang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Kebo Zhong
- Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Shuguang Liao
- Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Guifeng Zhang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical and Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
6
|
Wu TY, Hsieh YC, Yin WR, Cheng KY, Hou YT. Fabrication of a decellularized liver matrix-based hepatic patch for the repair of CCl4-induced liver injury. Biotechnol J 2024; 19:e2300570. [PMID: 38864387 DOI: 10.1002/biot.202300570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 04/23/2024] [Accepted: 05/16/2024] [Indexed: 06/13/2024]
Abstract
This article primarily introduces a new treatment for liver fibrosis/cirrhosis. We developed a hepatic patch by combining decellularized liver matrix (DLM) with the hepatocyte growth factor (HGF)/heparin-complex and evaluated its restorative efficacy. In vitro prophylactic results, the HGF/heparin-DLM patches effectively mitigated CCl4-induced hepatocyte toxicity and restored the cytotoxicity levels to the baseline levels by day 5. Furthermore, these patches restored albumin synthesis of injured hepatocytes to more than 70% of the normal levels within 5 days. In vitro therapeutic results, the urea synthesis of the injured hepatocytes reached 91% of the normal levels after 10 days of culture, indicating successful restoration of hepatic function by the HGF/heparin-DLM patches in both prophylactic and therapeutic models. In vivo results, HGF/heparin-DLM patches attached to the liver and gut exhibited a significant decrease in collagen content (4.44 times and 2.77 times, respectively) and an increase in glycogen content (1.19 times and 1.12 times, respectively) compared to the fibrosis group after 1 week, separately. In summary, liver function was restored and inflammation was inhibited through the combined effects of DLM and the HGF/heparin-complex in fibrotic liver. The newly designed hepatic patch holds promise for both in vitro and in vivo regeneration therapy and preventive health care for liver tissue engineering.
Collapse
Affiliation(s)
- Ting-Yi Wu
- Department of Biomechatronics Engineering, National Taiwan University, Taipei, Taiwan
| | - Yi-Cheng Hsieh
- Department of Biomechatronics Engineering, National Taiwan University, Taipei, Taiwan
| | - Wei-Rong Yin
- Department of Biomechatronics Engineering, National Taiwan University, Taipei, Taiwan
| | - Kai-Yi Cheng
- Department of Biomechatronics Engineering, National Taiwan University, Taipei, Taiwan
| | - Yung-Te Hou
- Department of Biomechatronics Engineering, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
7
|
Tamo AK, Djouonkep LDW, Selabi NBS. 3D Printing of Polysaccharide-Based Hydrogel Scaffolds for Tissue Engineering Applications: A Review. Int J Biol Macromol 2024; 270:132123. [PMID: 38761909 DOI: 10.1016/j.ijbiomac.2024.132123] [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: 12/05/2023] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/20/2024]
Abstract
In tissue engineering, 3D printing represents a versatile technology employing inks to construct three-dimensional living structures, mimicking natural biological systems. This technology efficiently translates digital blueprints into highly reproducible 3D objects. Recent advances have expanded 3D printing applications, allowing for the fabrication of diverse anatomical components, including engineered functional tissues and organs. The development of printable inks, which incorporate macromolecules, enzymes, cells, and growth factors, is advancing with the aim of restoring damaged tissues and organs. Polysaccharides, recognized for their intrinsic resemblance to components of the extracellular matrix have garnered significant attention in the field of tissue engineering. This review explores diverse 3D printing techniques, outlining distinctive features that should characterize scaffolds used as ideal matrices in tissue engineering. A detailed investigation into the properties and roles of polysaccharides in tissue engineering is highlighted. The review also culminates in a profound exploration of 3D polysaccharide-based hydrogel applications, focusing on recent breakthroughs in regenerating different tissues such as skin, bone, cartilage, heart, nerve, vasculature, and skeletal muscle. It further addresses challenges and prospective directions in 3D printing hydrogels based on polysaccharides, paving the way for innovative research to fabricate functional tissues, enhancing patient care, and improving quality of life.
Collapse
Affiliation(s)
- Arnaud Kamdem Tamo
- Institute of Microsystems Engineering IMTEK, University of Freiburg, 79110 Freiburg, Germany; Freiburg Center for Interactive Materials and Bioinspired Technologies FIT, University of Freiburg, 79110 Freiburg, Germany; Freiburg Materials Research Center FMF, University of Freiburg, 79104 Freiburg, Germany; Ingénierie des Matériaux Polymères (IMP), Université Claude Bernard Lyon 1, INSA de Lyon, Université Jean Monnet, CNRS, UMR 5223, 69622 Villeurbanne CEDEX, France.
| | - Lesly Dasilva Wandji Djouonkep
- College of Petroleum Engineering, Yangtze University, Wuhan 430100, China; Key Laboratory of Drilling and Production Engineering for Oil and Gas, Wuhan 430100, China
| | - Naomie Beolle Songwe Selabi
- Institute of Advanced Materials and Nanotechnology, Wuhan University of Science and Technology, Wuhan 430081, China
| |
Collapse
|
8
|
Ojha AK, Rajasekaran R, Hansda AK, Choudhury P, Biswas A, Sharma S, Chaudhuri PP, Dogra N, Goswami R, Chaudhury K, Dhara S. Biochemical and immunomodulatory insights of extracellular matrix from decellularized human whole cervix: recellularization and in vivoECM remodeling interplay. Biofabrication 2024; 16:035014. [PMID: 38663394 DOI: 10.1088/1758-5090/ad4393] [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: 07/15/2023] [Accepted: 04/25/2024] [Indexed: 05/08/2024]
Abstract
Extracellular matrix (ECM) rich whole organ bio-scaffolds, preserving structural integrity and essential growth factors, has potential towards regeneration and reconstruction. Women with cervical anomalies or trauma can benefit from clinical cervicovaginal repair using constructs rich in site specific ECM. In this study, complete human cervix decellularization was achieved using a modified perfusion-based stir bench top decellularization method. This was followed by physico-chemical processes including perfusion of ionic agents, enzymatic treatment and washing using detergent solutions for a duration of 10-12 d. Histopathological analysis, as well as DNA quantification confirmed the efficacy of the decellularization process. Tissue ultrastructure integrity was preserved and the same was validated via scanning electron microscopy and transmission electron microscopy studies. Biochemical analysis and structural characterizations like Fourier transform infrared, Raman spectroscopy of decellularized tissues demonstrated preservation of important proteins, crucial growth factors, collagen, and glycosaminoglycans.In vitrostudies, using THP-1 and human umbilical vein endothelial cell (HUVEC) cells, demonstrated macrophage polarization from M1 to M2 and vascular functional genes enhancement, respectively, when treated with decellularized human cervical matrix (DHCp). Crosslinked DHC scaffolds were recellularized with site specific human cervical epithelial cells and HUVEC, showing non-cytotoxic cell viability and enhanced proliferation. Furthermore, DHC scaffolds showed immunomodulatory effectsin vivoon small rodent model via upregulation of M2 macrophage genes as compared to decellularized rat cervix matrix scaffolds (DRC). DHC scaffolds underwent neo-vascularization followed by ECM remodeling with enhanced tissue integration.
Collapse
Affiliation(s)
- Atul Kumar Ojha
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Ragavi Rajasekaran
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Arman Kunwar Hansda
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Priyanka Choudhury
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Asmita Biswas
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Sunita Sharma
- Institute of Reproductive Medicine, Salt Lake, Kolkata, India
| | - Prithwis Pal Chaudhuri
- Department of Obstetrics and Gynecology, Zenith Super specialty hospital, Kolkata, India
| | - Nantu Dogra
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Ritobrata Goswami
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Koel Chaudhury
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Santanu Dhara
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
9
|
Wasyłeczko M, Wojciechowski C, Chwojnowski A. Polyethersulfone Polymer for Biomedical Applications and Biotechnology. Int J Mol Sci 2024; 25:4233. [PMID: 38673817 PMCID: PMC11049998 DOI: 10.3390/ijms25084233] [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: 03/07/2024] [Revised: 04/03/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Polymers stand out as promising materials extensively employed in biomedicine and biotechnology. Their versatile applications owe much to the field of tissue engineering, which seamlessly integrates materials engineering with medical science. In medicine, biomaterials serve as prototypes for organ development and as implants or scaffolds to facilitate body regeneration. With the growing demand for innovative solutions, synthetic and hybrid polymer materials, such as polyethersulfone, are gaining traction. This article offers a concise characterization of polyethersulfone followed by an exploration of its diverse applications in medical and biotechnological realms. It concludes by summarizing the significant roles of polyethersulfone in advancing both medicine and biotechnology, as outlined in the accompanying table.
Collapse
Affiliation(s)
- Monika Wasyłeczko
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Ksiecia Trojdena 4, 02-109 Warsaw, Poland; (C.W.); (A.C.)
| | | | | |
Collapse
|
10
|
Zeshan M, Amjed N, Ashraf H, Farooq A, Akram N, Zia KM. A review on the application of chitosan-based polymers in liver tissue engineering. Int J Biol Macromol 2024; 262:129350. [PMID: 38242400 DOI: 10.1016/j.ijbiomac.2024.129350] [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: 08/28/2023] [Revised: 12/29/2023] [Accepted: 01/07/2024] [Indexed: 01/21/2024]
Abstract
Chitosan-based polymers have enormous structural tendencies to build bioactive materials with novel characteristics, functions, and various applications, mainly in liver tissue engineering (LTE). The specific physicochemical, biological, mechanical, and biodegradation properties give the effective ways to blend these biopolymers with synthetic and natural polymers to fabricate scaffolds matrixes, sponges, and complexes. A variety of natural and synthetic biomaterials, including chitosan (CS), alginate (Alg), collagen (CN), gelatin (GL), hyaluronic acid (HA), hydroxyapatite (HAp), polyethylene glycol (PEG), polycaprolactone (PCL), poly(lactic-co-glycolic) acid (PGLA), polylactic acid (PLA), and silk fibroin gained considerable attention due to their structure-properties relationship. The incorporation of CS within the polymer matrix results in increased mechanical strength and also imparts biological behavior to the designed PU formulations. The significant and growing interest in the LTE sector, this review aims to be a detailed exploration of CS-based polymers biomaterials for LTE. A brief explanation of the sources and extraction, properties, structure, and scope of CS is described in the introduction. After that, a full overview of the liver, its anatomy, issues, hepatocyte transplantation, LTE, and CS LTE applications are discussed.
Collapse
Affiliation(s)
- Muhammad Zeshan
- Department of Chemistry, University of Agriculture, Faisalabad, Pakistan
| | - Nyla Amjed
- Department of Chemistry, The University of Lahore, Lahore, Pakistan
| | - Humna Ashraf
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Ariba Farooq
- Department of Chemistry, The University of Lahore, Lahore, Pakistan
| | - Nadia Akram
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Khalid Mahmood Zia
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan.
| |
Collapse
|
11
|
Golebiowska AA, Intravaia JT, Sathe VM, Kumbar SG, Nukavarapu SP. Decellularized extracellular matrix biomaterials for regenerative therapies: Advances, challenges and clinical prospects. Bioact Mater 2024; 32:98-123. [PMID: 37927899 PMCID: PMC10622743 DOI: 10.1016/j.bioactmat.2023.09.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 11/07/2023] Open
Abstract
Tissue engineering and regenerative medicine have shown potential in the repair and regeneration of tissues and organs via the use of engineered biomaterials and scaffolds. However, current constructs face limitations in replicating the intricate native microenvironment and achieving optimal regenerative capacity and functional recovery. To address these challenges, the utilization of decellularized tissues and cell-derived extracellular matrix (ECM) has emerged as a promising approach. These biocompatible and bioactive biomaterials can be engineered into porous scaffolds and grafts that mimic the structural and compositional aspects of the native tissue or organ microenvironment, both in vitro and in vivo. Bioactive dECM materials provide a unique tissue-specific microenvironment that can regulate and guide cellular processes, thereby enhancing regenerative therapies. In this review, we explore the emerging frontiers of decellularized tissue-derived and cell-derived biomaterials and bio-inks in the field of tissue engineering and regenerative medicine. We discuss the need for further improvements in decellularization methods and techniques to retain structural, biological, and physicochemical characteristics of the dECM products in a way to mimic native tissues and organs. This article underscores the potential of dECM biomaterials to stimulate in situ tissue repair through chemotactic effects for the development of growth factor and cell-free tissue engineering strategies. The article also identifies the challenges and opportunities in developing sterilization and preservation methods applicable for decellularized biomaterials and grafts and their translation into clinical products.
Collapse
Affiliation(s)
| | - Jonathon T. Intravaia
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Vinayak M. Sathe
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA
| | - Sangamesh G. Kumbar
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA
| | - Syam P. Nukavarapu
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA
| |
Collapse
|
12
|
Carvalho AM, Bansal R, Barrias CC, Sarmento B. The Material World of 3D-Bioprinted and Microfluidic-Chip Models of Human Liver Fibrosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307673. [PMID: 37961933 DOI: 10.1002/adma.202307673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/06/2023] [Indexed: 11/15/2023]
Abstract
Biomaterials are extensively used to mimic cell-matrix interactions, which are essential for cell growth, function, and differentiation. This is particularly relevant when developing in vitro disease models of organs rich in extracellular matrix, like the liver. Liver disease involves a chronic wound-healing response with formation of scar tissue known as fibrosis. At early stages, liver disease can be reverted, but as disease progresses, reversion is no longer possible, and there is no cure. Research for new therapies is hampered by the lack of adequate models that replicate the mechanical properties and biochemical stimuli present in the fibrotic liver. Fibrosis is associated with changes in the composition of the extracellular matrix that directly influence cell behavior. Biomaterials could play an essential role in better emulating the disease microenvironment. In this paper, the recent and cutting-edge biomaterials used for creating in vitro models of human liver fibrosis are revised, in combination with cells, bioprinting, and/or microfluidics. These technologies have been instrumental to replicate the intricate structure of the unhealthy tissue and promote medium perfusion that improves cell growth and function, respectively. A comprehensive analysis of the impact of material hints and cell-material interactions in a tridimensional context is provided.
Collapse
Affiliation(s)
- Ana Margarida Carvalho
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Ruchi Bansal
- Translational Liver Research, Department of Medical Cell Biophysics, Technical Medical Center, Faculty of Science and Technology, University of Twente, Enschede, 7522 NB, The Netherlands
| | - Cristina C Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- IUCS - Instituto Universitário de Ciências da Saúde, CESPU, Rua Central de Gandra 1317, Gandra, 4585-116, Portugal
| |
Collapse
|
13
|
Faccioli LA, Dias ML, Martins-Santos R, Paredes BD, Takiya CM, dos Santos Goldenberg RC. Resident Liver Stem Cells. RESIDENT STEM CELLS AND REGENERATIVE THERAPY 2024:23-51. [DOI: 10.1016/b978-0-443-15289-4.00015-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
14
|
Kasturi M, Mathur V, Gadre M, Srinivasan V, Vasanthan KS. Three Dimensional Bioprinting for Hepatic Tissue Engineering: From In Vitro Models to Clinical Applications. Tissue Eng Regen Med 2024; 21:21-52. [PMID: 37882981 PMCID: PMC10764711 DOI: 10.1007/s13770-023-00576-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 10/27/2023] Open
Abstract
Fabrication of functional organs is the holy grail of tissue engineering and the possibilities of repairing a partial or complete liver to treat chronic liver disorders are discussed in this review. Liver is the largest gland in the human body and plays a responsible role in majority of metabolic function and processes. Chronic liver disease is one of the leading causes of death globally and the current treatment strategy of organ transplantation holds its own demerits. Hence there is a need to develop an in vitro liver model that mimics the native microenvironment. The developed model should be a reliable to understand the pathogenesis, screen drugs and assist to repair and replace the damaged liver. The three-dimensional bioprinting is a promising technology that recreates in vivo alike in vitro model for transplantation, which is the goal of tissue engineers. The technology has great potential due to its precise control and its ability to homogeneously distribute cells on all layers in a complex structure. This review gives an overview of liver tissue engineering with a special focus on 3D bioprinting and bioinks for liver disease modelling and drug screening.
Collapse
Affiliation(s)
- Meghana Kasturi
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Vidhi Mathur
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Mrunmayi Gadre
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Varadharajan Srinivasan
- Department of Civil Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Kirthanashri S Vasanthan
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
15
|
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
|
16
|
Yadav P, Singh SK, Rajput S, Allawadhi P, Khurana A, Weiskirchen R, Navik U. Therapeutic potential of stem cells in regeneration of liver in chronic liver diseases: Current perspectives and future challenges. Pharmacol Ther 2024; 253:108563. [PMID: 38013053 DOI: 10.1016/j.pharmthera.2023.108563] [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: 09/15/2023] [Revised: 11/04/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023]
Abstract
The deposition of extracellular matrix and hyperplasia of connective tissue characterizes chronic liver disease called hepatic fibrosis. Progression of hepatic fibrosis may lead to hepatocellular carcinoma. At this stage, only liver transplantation is a viable option. However, the number of possible liver donors is less than the number of patients needing transplantation. Consequently, alternative cell therapies based on non-stem cells (e.g., fibroblasts, chondrocytes, keratinocytes, and hepatocytes) therapy may be able to postpone hepatic disease, but they are often ineffective. Thus, novel stem cell-based therapeutics might be potentially important cutting-edge approaches for treating liver diseases and reducing patient' suffering. Several signaling pathways provide targets for stem cell interventions. These include pathways such as TGF-β, STAT3/BCL-2, NADPH oxidase, Raf/MEK/ERK, Notch, and Wnt/β-catenin. Moreover, mesenchymal stem cells (MSCs) stimulate interleukin (IL)-10, which inhibits T-cells and converts M1 macrophages into M2 macrophages, producing an anti-inflammatory environment. Furthermore, it inhibits the action of CD4+ and CD8+ T cells and reduces the activity of TNF-α and interferon cytokines by enhancing IL-4 synthesis. Consequently, the immunomodulatory and anti-inflammatory capabilities of MSCs make them an attractive therapeutic approach. Importantly, MSCs can inhibit the activation of hepatic stellate cells, causing their apoptosis and subsequent promotion of hepatocyte proliferation, thereby replacing dead hepatocytes and reducing liver fibrosis. This review discusses the multidimensional therapeutic role of stem cells as cell-based therapeutics in liver fibrosis.
Collapse
Affiliation(s)
- Poonam Yadav
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India
| | - Sumeet Kumar Singh
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India
| | - Sonu Rajput
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India
| | - Prince Allawadhi
- Department of Pharmacy, Vaish Institute of Pharmaceutical Education and Research (VIPER), Pandit Bhagwat Dayal Sharma University of Health Sciences (Pt. B. D. S. UHS), Rohtak, Haryana 124001, India
| | - Amit Khurana
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India; Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, Pauwelsstr. 30, D-52074 Aachen, Germany.
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, Pauwelsstr. 30, D-52074 Aachen, Germany.
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India; Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, Pauwelsstr. 30, D-52074 Aachen, Germany.
| |
Collapse
|
17
|
Ghosh S, Pati F. Decellularized extracellular matrix and silk fibroin-based hybrid biomaterials: A comprehensive review on fabrication techniques and tissue-specific applications. Int J Biol Macromol 2023; 253:127410. [PMID: 37844823 DOI: 10.1016/j.ijbiomac.2023.127410] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/01/2023] [Accepted: 10/10/2023] [Indexed: 10/18/2023]
Abstract
Biomaterials play a fundamental role in tissue engineering by providing biochemical and physical cues that influence cellular fate and matrix development. Decellularized extracellular matrix (dECM) as a biomaterial is distinguished by its abundant composition of matrix proteins, such as collagen, elastin, fibronectin, and laminin, as well as glycosaminoglycans and proteoglycans. However, the mechanical properties of only dECM-based constructs may not always meet tissue-specific requirements. Recent advancements address this challenge by utilizing hybrid biomaterials that harness the strengths of silk fibroin (SF), which contributes the necessary mechanical properties, while dECM provides essential cellular cues for in vitro studies and tissue regeneration. This review discusses emerging trends in developing such biopolymer blends, aiming to synergistically combine the advantages of SF and dECM through optimal concentrations and desired cross-linking density. We focus on different fabrication techniques and cross-linking methods that have been utilized to fabricate various tissue-engineered hybrid constructs. Furthermore, we survey recent applications of such biomaterials for the regeneration of various tissues, including bone, cartilage, trachea, bladder, vascular graft, heart, skin, liver, and other soft tissues. Finally, the trajectory and prospects of the constructs derived from this blend in the tissue engineering field have been summarized, highlighting their potential for clinical translation.
Collapse
Affiliation(s)
- Soham Ghosh
- BioFab Lab, Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502285, Telangana, India
| | - Falguni Pati
- BioFab Lab, Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502285, Telangana, India.
| |
Collapse
|
18
|
Homayoon Vala M, Bagheri H, Sargazi Z, Bakhtiary N, Pourbeiranvand S, Salehnia M. Evaluation of Vascular Endothelial Growth Factor Gene Expression in Recellularized Liver Tissue by Mouse Embryo Fibroblast. IRANIAN BIOMEDICAL JOURNAL 2023; 27:340-8. [PMID: 37950395 PMCID: PMC10826915 DOI: 10.61186/ibj.3862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 06/10/2023] [Indexed: 12/25/2023]
Abstract
Background The aim of the present study was to evaluate alterations in the vegf gene expression as an angiogenic factor in mouse embryo fibroblasts seeded on the decellularized liver fragments. Methods Liver tissue samples (n = 10) collected from adult male mice were randomly divided into decellularized and native control groups. Tissues were decellularized by treating with 1% Triton X-100 and 0.1% SDS for 24 hours and assessed by H&E staining and SEM. Then DNA content analysis and toxicity tests were performed. By centrifugation, DiI-labeled mouse embryo fibroblasts were seeded on each scaffold and cultured for one week. The recellularized scaffolds were studied by H&E staining, SEM, and LSCM. After RNA extraction and cDNA synthesis, the expression of the vegf gene in these samples was investigated using real-time RT-PCR. Results Our observations showed that the decellularized tissues had morphology and porous structure similar to the control group, and their DNA content significantly reduced (p < 0.05) and reached to 4.12% of the control group. The MTT test indicated no significant cellular toxicity for the decellularized scaffolds. Light microscopy, SEM, and LSCM observations confirmed the attachment and penetration of embryonic fibroblast cells on the surface and into different depths of the scaffolds. There was no statistically significant difference in terms of vegf gene expression in the cultured cells in the presence and absence of a scaffold. Conclusion The reconstructed scaffold had no effect on vegf gene expression. Decellularized mouse liver tissue recellularized by embryonic fibroblasts could have an application in regenerative medicine.
Collapse
Affiliation(s)
- Motahare Homayoon Vala
- Department of Biomaterials, Faculty of Interdisciplinary Science and Technology, Tarbiat Modares University, Tehran, Iran
| | - Hamed Bagheri
- Department of Biomaterials, Faculty of Interdisciplinary Science and Technology, Tarbiat Modares University, Tehran, Iran
| | - Zinat Sargazi
- Department of Anatomical Sciences, School of Medical Sciences, Zahedan University, Zahedan, Iran
| | - Negar Bakhtiary
- Department of Biomaterials, Faculty of Interdisciplinary Science and Technology, Tarbiat Modares University, Tehran, Iran
- Burn Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Shahram Pourbeiranvand
- Department of Anatomical Sciences, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mojdeh Salehnia
- Department of Biomaterials, Faculty of Interdisciplinary Science and Technology, Tarbiat Modares University, Tehran, Iran
- Department of Anatomical Sciences, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
19
|
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
|
20
|
Zhang H, Wang M, Wu R, Guo J, Sun A, Li Z, Ye R, Xu G, Cheng Y. From materials to clinical use: advances in 3D-printed scaffolds for cartilage tissue engineering. Phys Chem Chem Phys 2023; 25:24244-24263. [PMID: 37698006 DOI: 10.1039/d3cp00921a] [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: 09/13/2023]
Abstract
Osteoarthritis caused by articular cartilage defects is a particularly common orthopedic disease that can involve the entire joint, causing great pain to its sufferers. A global patient population of approximately 250 million people has an increasing demand for new therapies with excellent results, and tissue engineering scaffolds have been proposed as a potential strategy for the repair and reconstruction of cartilage defects. The precise control and high flexibility of 3D printing provide a platform for subversive innovation. In this perspective, cartilage tissue engineering (CTE) scaffolds manufactured using different biomaterials are summarized from the perspective of 3D printing strategies, the bionic structure strategies and special functional designs are classified and discussed, and the advantages and limitations of these CTE scaffold preparation strategies are analyzed in detail. Finally, the application prospect and challenges of 3D printed CTE scaffolds are discussed, providing enlightening insights for their current research.
Collapse
Affiliation(s)
- Hewen Zhang
- School of the Faculty of Mechanical Engineering and Mechanic, Ningbo University, Ningbo, Zhejiang Province, 315211, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- Zhejiang Key Laboratory of Additive Manufacturing Materials, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China.
| | - Meng Wang
- Department of Joint Surgery, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, 315020, China.
| | - Rui Wu
- Department of Orthopedics, Ningbo First Hospital Longshan Hospital Medical and Health Group, Ningbo 315201, P. R. China
| | - Jianjun Guo
- Zhejiang Key Laboratory of Additive Manufacturing Materials, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China.
| | - Aihua Sun
- Zhejiang Key Laboratory of Additive Manufacturing Materials, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China.
| | - Zhixiang Li
- Zhejiang Key Laboratory of Additive Manufacturing Materials, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China.
| | - Ruqing Ye
- Department of Joint Surgery, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, 315020, China.
| | - Gaojie Xu
- Zhejiang Key Laboratory of Additive Manufacturing Materials, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China.
| | - Yuchuan Cheng
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- Zhejiang Key Laboratory of Additive Manufacturing Materials, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China.
| |
Collapse
|
21
|
Kim MK, Jeong W, Kang HW. Liver dECM-Gelatin Composite Bioink for Precise 3D Printing of Highly Functional Liver Tissues. J Funct Biomater 2023; 14:417. [PMID: 37623662 PMCID: PMC10455418 DOI: 10.3390/jfb14080417] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 07/31/2023] [Accepted: 08/04/2023] [Indexed: 08/26/2023] Open
Abstract
In recent studies, liver decellularized extracellular matrix (dECM)-based bioinks have gained significant attention for their excellent compatibility with hepatocytes. However, their low printability limits the fabrication of highly functional liver tissue. In this study, a new liver dECM-gelatin composite bioink (dECM gBioink) was developed to overcome this limitation. The dECM gBioink was prepared by incorporating a viscous gelatin mixture into the liver dECM material. The novel dECM gBioink showed 2.44 and 10.71 times higher bioprinting resolution and compressive modulus, respectively, than a traditional dECM bioink. In addition, the new bioink enabled stable stacking with 20 or more layers, whereas a structure printed with the traditional dECM bioink collapsed. Moreover, the proposed dECM gBioink exhibited excellent hepatocyte and endothelial cell compatibility. At last, the liver lobule mimetic structure was successfully fabricated with a precisely patterned endothelial cell cord-like pattern and primary hepatocytes using the dECM gBioink. The fabricated lobule structure exhibited excellent hepatic functionalities and dose-dependent responses to hepatotoxic drugs. These results demonstrated that the gelatin mixture can significantly improve the printability and mechanical properties of the liver dECM materials while maintaining good cytocompatibility. This novel liver dECM gBioink with enhanced 3D printability and resolution can be used as an advanced tool for engineering highly functional liver tissues.
Collapse
Affiliation(s)
| | | | - Hyun-Wook Kang
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), UNIST 50, UNIST-gil, Ulsan 44919, Republic of Korea; (M.K.K.); (W.J.)
| |
Collapse
|
22
|
Shagidulin M, Onishchenko N, Sevastianov V, Krasheninnikov M, Lyundup A, Nikolskaya A, Kryzhanovskaya A, Voznesenskaia S, Gorelova M, Perova N, Kozlov I, Venediktov A, Piavchenko G, Gautier S. Experimental Correction and Treatment of Chronic Liver Failure Using Implantable Cell-Engineering Constructs of the Auxiliary Liver Based on a Bioactive Heterogeneous Biopolymer Hydrogel. Gels 2023; 9:456. [PMID: 37367127 DOI: 10.3390/gels9060456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/18/2023] [Accepted: 05/25/2023] [Indexed: 06/28/2023] Open
Abstract
Our study sought approaches for chronic liver failure (CLF) treatment and correction via cell-engineered constructs (CECs). They are built from biopolymer-based, microstructured, and collagen-containing hydrogel (BMCG). We also strove to evaluate the functional activity of BMCG in liver regeneration. MATERIALS AND METHODS Allogeneic liver cells (namely, hepatocytes; LC) together with mesenchymal multipotent stem cells of bone marrow origin (MMSC BM; BMSCs) were adhered to our BMCG to compose implanted liver CECs. Thereafter, we investigated a model of CLF in rats receiving the implanted CECs. The CLF had been provoked by long-term exposure to carbon tetrachloride. The study comprised male Wistar rats (n = 120) randomized into 3 groups: Group 1 was a control group with the saline treatment of the hepatic parenchyma (n = 40); Group 2 received BMCG only (n = 40); and Group 3 was loaded with CECs implanted into the parenchyma of their livers (n = 40). August rats (n = 30) made up a donor population for LCs and MMSC BM to develop grafts for animals from Group 3. The study length was 90 days. RESULTS CECs were shown to affect both biochemical test values and morphological parameters in rats with CLF. CONCLUSION We found BMCG-derived CECs to be operational and active, with regenerative potential. Group 3 showed significant evidence of forced liver regeneration that tended to persist until the end of the study (day 90). The phenomenon is reflected by biochemical signs of hepatic functional recovery by day 30 after grafting (compared to Groups 1 and 2), whereas structural features of liver repair (necrosis prevention, missing formation of vacuoles, degenerating LC number decrease, and delay of hepatic fibrotic transformation). Such implantation of BMCG-derived CECs with allogeneic LCs and MMSC BM might represent a proper option to correct and treat CLF, as well as to maintain affected liver function in patients with liver grafting needed.
Collapse
Affiliation(s)
- Murat Shagidulin
- Federal State Budgetary Institution "Shumakov National Medical Research Centre of Transplantology and Artificial Organs" of the Ministry of Health of the Russian Federation, 123182 Moscow, Russia
- Federal State Autonomous Educational Institution of Higher Education, "I.M. Sechenov First Moscow State Medical University" of the Ministry of Health of the Russian Federation (Sechenov University), 119435 Moscow, Russia
| | - Nina Onishchenko
- Federal State Budgetary Institution "Shumakov National Medical Research Centre of Transplantology and Artificial Organs" of the Ministry of Health of the Russian Federation, 123182 Moscow, Russia
| | - Victor Sevastianov
- Federal State Budgetary Institution "Shumakov National Medical Research Centre of Transplantology and Artificial Organs" of the Ministry of Health of the Russian Federation, 123182 Moscow, Russia
| | - Mikhail Krasheninnikov
- Research and Education Resource Centre for Cellular Technologies, Peoples' Friendship University of Russia (RUDN University), 117198 Moscow, Russia
- M.V. Lomonosov Moscow State Academy of Fine Chemical Technology (MITKhT), 119571 Moscow, Russia
| | - Aleksey Lyundup
- Research and Education Resource Centre for Cellular Technologies, Peoples' Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Alla Nikolskaya
- Federal State Budgetary Institution "Shumakov National Medical Research Centre of Transplantology and Artificial Organs" of the Ministry of Health of the Russian Federation, 123182 Moscow, Russia
| | - Alena Kryzhanovskaya
- Federal State Autonomous Educational Institution of Higher Education, "I.M. Sechenov First Moscow State Medical University" of the Ministry of Health of the Russian Federation (Sechenov University), 119435 Moscow, Russia
| | - Sofia Voznesenskaia
- Federal State Autonomous Educational Institution of Higher Education, "I.M. Sechenov First Moscow State Medical University" of the Ministry of Health of the Russian Federation (Sechenov University), 119435 Moscow, Russia
| | - Mariia Gorelova
- Federal State Autonomous Educational Institution of Higher Education, "I.M. Sechenov First Moscow State Medical University" of the Ministry of Health of the Russian Federation (Sechenov University), 119435 Moscow, Russia
| | - Nadezhda Perova
- ANO "Institute Biomedical Research and Technology", 123557 Moscow, Russia
| | - Igor Kozlov
- Federal State Autonomous Educational Institution of Higher Education, "I.M. Sechenov First Moscow State Medical University" of the Ministry of Health of the Russian Federation (Sechenov University), 119435 Moscow, Russia
| | - Artem Venediktov
- Federal State Autonomous Educational Institution of Higher Education, "I.M. Sechenov First Moscow State Medical University" of the Ministry of Health of the Russian Federation (Sechenov University), 119435 Moscow, Russia
| | - Gennadii Piavchenko
- Federal State Autonomous Educational Institution of Higher Education, "I.M. Sechenov First Moscow State Medical University" of the Ministry of Health of the Russian Federation (Sechenov University), 119435 Moscow, Russia
| | - Sergey Gautier
- Federal State Budgetary Institution "Shumakov National Medical Research Centre of Transplantology and Artificial Organs" of the Ministry of Health of the Russian Federation, 123182 Moscow, Russia
- Federal State Autonomous Educational Institution of Higher Education, "I.M. Sechenov First Moscow State Medical University" of the Ministry of Health of the Russian Federation (Sechenov University), 119435 Moscow, Russia
| |
Collapse
|
23
|
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
|
24
|
Kokorev OV, Marchenko ES, Khlusov IA, Volinsky AA, Yasenchuk YF, Monogenov AN. Engineered Fibrous NiTi Scaffolds with Cultured Hepatocytes for Liver Regeneration in Rats. ACS Biomater Sci Eng 2023; 9:1558-1569. [PMID: 36802492 DOI: 10.1021/acsbiomaterials.2c01268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
At present, the use of alternative systems to replenish the lost functions of hepatic metabolism and partial replacement of liver organ failure is relevant, due to an increase in the incidence of various liver disorders, insufficiency, and cost of organs for transplantation, as well as the high cost of using the artificial liver systems. The development of low-cost intracorporeal systems for maintaining hepatic metabolism using tissue engineering, as a bridge before liver transplantation or completely replacing liver function, deserves special attention. In vivo applications of intracorporeal fibrous nickel-titanium scaffolds (FNTSs) with cultured hepatocytes are described. Hepatocytes cultured in FNTSs are superior to their injections in terms of liver function, survival time, and recovery in a CCl4-induced cirrhosis rats' model. 232 animals were divided into 5 groups: control, CCl4-induced cirrhosis, CCl4-induced cirrhosis followed by implantation of cell-free FNTSs (sham surgery), CCl4-induced cirrhosis followed by infusion of hepatocytes (2 mL, 107 cells/mL), and CCl4-induced cirrhosis followed by FNTS implantation with hepatocytes. Restoration of hepatocyte function in the FNTS implantation with the hepatocytes group was accompanied by a significant decrease in the level of aspartate aminotransferase (AsAT) in blood serum compared to the cirrhosis group. A significant decrease in the level of AsAT was noted after 15 days in the infused hepatocytes group. However, on the 30th day, the AsAT level increased and was close to the cirrhosis group due to the short-term effect after the introduction of hepatocytes without a scaffold. The changes in alanine aminotransferase (AlAT), alkaline phosphatase (AlP), total and direct bilirubin, serum protein, triacylglycerol, lactate, albumin, and lipoproteins were similar to those in AsAT. The survival time of animals was significantly longer in the FNTS implantation with hepatocytes group. The obtained results showed the scaffolds' ability to support hepatocellular metabolism. The development of hepatocytes in FNTS was studied in vivo using 12 animals using scanning electron microscopy. Hepatocytes demonstrated good adhesion to the scaffold wireframe and survival in allogeneic conditions. Mature tissue, including cellular and fibrous, filled the scaffold space by 98% in 28 days. The study shows the extent to which an implantable "auxiliary liver" compensates for the lack of liver function without replacement in rats.
Collapse
Affiliation(s)
- Oleg V Kokorev
- National Research Tomsk State University, 36 Lenin Ave., Tomsk 634050, Russia
- Siberian State Medical University, 2 Moskovsky Trakt, Tomsk 634050, Russia
| | | | - Igor A Khlusov
- Siberian State Medical University, 2 Moskovsky Trakt, Tomsk 634050, Russia
| | - Alex A Volinsky
- National Research Tomsk State University, 36 Lenin Ave., Tomsk 634050, Russia
- Department of Mechanical Engineering, University of South Florida, 4202 E. Fowler Ave. ENG030, Tampa, Florida 33620, United States
| | - Yuri F Yasenchuk
- National Research Tomsk State University, 36 Lenin Ave., Tomsk 634050, Russia
| | | |
Collapse
|
25
|
Mirdamadi ES, Khosrowpour Z, Jafari D, Gholipourmalekabadi M, Solati-Hashjin M. 3D-printed PLA/Gel hybrid in liver tissue engineering: Effects of architecture on biological functions. Biotechnol Bioeng 2023; 120:836-851. [PMID: 36479982 DOI: 10.1002/bit.28301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/28/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022]
Abstract
The liver is one of the vital organs in the body, and the gold standard of treatment for liver function impairment is liver transplantation, which poses many challenges. The specific three-dimensional (3D) structure of liver, which significantly impacts the growth and function of its cells, has made biofabrication with the 3D printing of scaffolds suitable for this approach. In this study, to investigate the effect of scaffold geometry on the performance of HepG2 cells, poly-lactic acid (PLA) polymer was used as the input of the fused deposition modeling (FDM) 3D-printing machine. Samples with simple square and bioinspired hexagonal cross-sectional designs were printed. One percent and 2% of gelatin coating were applied to the 3D printed PLA to improve the wettability and surface properties of the scaffold. Scanning electron microscopy pictures were used to analyze the structural properties of PLA-Gel hybrid scaffolds, energy dispersive spectroscopy to investigate the presence of gelatin, water contact angle measurement for wettability, and weight loss for degradation. In vitro tests were performed by culturing HepG2 cells on the scaffold to evaluate the cell adhesion, viability, cytotoxicity, and specific liver functions. Then, high-precision scaffolds were printed and the presence of gelatin was detected. Also, the effect of geometry on cell function was confirmed in viability, adhesion, and functional tests. The albumin and urea production of the Hexagonal PLA scaffold was about 1.22 ± 0.02-fold higher than the square design in 3 days. This study will hopefully advance our understanding of liver tissue engineering toward a promising perspective for liver regeneration.
Collapse
Affiliation(s)
- Elnaz Sadat Mirdamadi
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.,Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA
| | - Zahra Khosrowpour
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Davod Jafari
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mazaher Gholipourmalekabadi
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.,Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehran Solati-Hashjin
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| |
Collapse
|
26
|
Mirhaidari GJ, Barker JC, Breuer CK, Reinhardt JW. Implanted Tissue-Engineered Vascular Graft Cell Isolation with Single-Cell RNA Sequencing Analysis. Tissue Eng Part C Methods 2023; 29:72-84. [PMID: 36719780 PMCID: PMC9968626 DOI: 10.1089/ten.tec.2022.0189] [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: 11/12/2022] [Accepted: 01/17/2023] [Indexed: 02/01/2023] Open
Abstract
The advent of single-cell RNA sequencing (scRNA-Seq) has brought with it the ability to gain greater insights into the cellular composition of tissues and heterogeneity in gene expression within specific cell types. For tissue-engineered blood vessels, this is particularly impactful to better understand how neotissue forms and remodels into tissue resembling a native vessel. A notable challenge, however, is the ability to separate cells from synthetic biomaterials to generate high-quality single-cell suspensions to interrogate the cellular composition of our tissue-engineered vascular grafts (TEVGs) during active remodeling in situ. We present here a simple, commercially available approach to separate cells within our TEVG from the residual scaffold for downstream use in a scRNA-Seq workflow. Utilizing this method, we identified the cell populations comprising explanted TEVGs and compared these with results from immunohistochemical analysis. The process began with explanted TEVGs undergoing traditional mechanical and enzymatic dissociation to separate cells from scaffold and extracellular matrix proteins. Magnetically labeled antibodies targeting murine origin cells were incubated with enzymatic digests of TEVGs containing cells and scaffold debris in suspension allowing for separation by utilizing a magnetic separator column. Single-cell suspensions were processed through 10 × Genomics and data were analyzed utilizing R to generate cell clusters. Expression data provided new insights into a diverse composition of phenotypically unique subclusters within the fibroblast, macrophage, smooth muscle cell, and endothelial cell populations contributing to the early neotissue remodeling stages of TEVGs. These populations were correlated qualitatively and quantitatively with immunohistochemistry highlighting for the first time the potential of scRNA-Seq to provide exquisite detail into the host cellular response to an implanted TEVG. These results additionally demonstrate magnetic cell isolation is an effective method for generating high-quality cell suspensions for scRNA-Seq. While this method was utilized for our group's TEVGs, it has broader applications to other implantable materials that use biodegradable synthetic materials as part of scaffold composition. Impact statement Single-cell RNA sequencing is an evolving technology with the ability to provide detailed information on the cellular composition of remodeling biomaterials in vivo. This present work details an effective approach for separating nondegraded biomaterials from cells for downstream RNA-sequencing analysis. We applied this method to implanted tissue-engineered vascular grafts and for the first time describe the cellular composition of the remodeling graft at a single-cell gene expression level. While this method was effective in our scaffold, it has broad applicability to other implanted biomaterials that necessitate separation of cell from residual scaffold materials for single-cell RNA sequencing.
Collapse
Affiliation(s)
- Gabriel J.M. Mirhaidari
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Jenny C. Barker
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Plastic and Reconstructive Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Christopher K. Breuer
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - James W. Reinhardt
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| |
Collapse
|
27
|
Le Guilcher C, Merlen G, Dellaquila A, Labour MN, Aid R, Tordjmann T, Letourneur D, Simon-Yarza T. Engineered human liver based on pullulan-dextran hydrogel promotes mice survival after liver failure. Mater Today Bio 2023; 19:100554. [PMID: 36756209 PMCID: PMC9900439 DOI: 10.1016/j.mtbio.2023.100554] [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: 09/21/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Liver tissue engineering approaches aim to support drug testing, assistance devices, or transplantation. However, their suitability for clinical application remains unsatisfactory. Herein, we demonstrate the beneficial and biocompatible use of porous pullulan-dextran hydrogel for the self-assembly of hepatocytes and biliary-like cells into functional 3D microtissues. Using HepaRG cells, we obtained 21 days maintenance of engineered liver polarity, functional detoxification and excretion systems, as well as glycogen storage in hydrogel. Implantation on two liver lobes in mice of hydrogels containing 3800 HepaRG 3D structures of 100 μm in diameter, indicated successful engraftment and no signs of liver toxicity after one month. Finally, after acetaminophen-induced liver failure, when mice were transplanted with engineered livers on left lobe and peritoneal cavity, the survival rate at 7 days significantly increased by 31.8% compared with mice without cell therapy. These findings support the clinical potential of pullulan-dextran hydrogel for liver failure management.
Collapse
Affiliation(s)
- Camille Le Guilcher
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, U1148, F-75018 Paris, France,Corresponding author.
| | - Grégory Merlen
- Université Paris-Saclay, INSERM U1193, F- 94800 Villejuif, France
| | - Alessandra Dellaquila
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, U1148, F-75018 Paris, France
| | - Marie-Noëlle Labour
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, U1148, F-75018 Paris, France,ICGM, Université de Montpellier, CNRS, ENSCM, F- 34293 Montpellier, France,École Pratique des Hautes Études, Université Paris Sciences et Lettres, F-75014 Paris, France
| | - Rachida Aid
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, U1148, F-75018 Paris, France
| | | | - Didier Letourneur
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, U1148, F-75018 Paris, France,Corresponding author.
| | - Teresa Simon-Yarza
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, U1148, F-75018 Paris, France,Corresponding author.
| |
Collapse
|
28
|
Jambar Nooshin B, Tayebi T, Babajani A, Khani MM, Niknejad H. Effects of Different Perfusing Routes through The Portal Vein, Hepatic Vein, and Biliary Duct on Whole Rat Liver Decellularization. CELL JOURNAL 2023; 25:35-44. [PMID: 36680482 PMCID: PMC9868438 DOI: 10.22074/cellj.2022.557600.1081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Organ transplantation is the last therapeutic choice for end-stage liver failure, which is limited by the lack of sufficient donors. Decellularized liver can be used as a suitable matrix for liver tissue engineering with clinical application potential. Optimizing the decellularization procedure would obtain a biological matrix with completely removed cellular components and preserved 3-dimensional structure. This study aimed to evaluate the decellularization efficacy through three anatomical routes. MATERIALS AND METHODS In this experimental study, rat liver decellularization was performed through biliary duct (BD), portal vein (PV), and hepatic vein (HV); using chemical detergents and enzymes. The decellularization efficacy was evaluated by measurement of DNA content, extracellular matrix (ECM) total proteins, and glycosaminoglycans (GAGs). ECM preservation was examined by histological and immunohistochemical (IHC) staining and scanning electron microscopy (SEM). Scaffold biocompatibility was tested by the MTT assay for HepG2 and HUVEC cell lines. RESULTS Decellularization through HV and PV resulted in a transparent scaffold by complete cell removal, while the BD route produced an opaque scaffold with incomplete decellularization. H and E staining confirmed these results. Maximum DNA loss was obtained using 1% and 0.5% sodium dodecyl sulfate (SDS) in the PV and HV groups and the DNA content decreased faster in the HV group. At the final stages, the proteins excreted in the HV and PV groups were significantly less than the BD group. The GAGs level was diminished after decellularization, especially in the PV and HV groups. In the HV and PV groups the collagen amount was significantly more than the BD group. The IHC and SEM images showed that the ECM structure was preserved and cellular components were entirely removed. MTT assay showed the biocompatibility of the decellularized scaffold. CONCLUSION The results revealed that the HV is a more suitable route for liver decellularization than the PV and BD.
Collapse
Affiliation(s)
- Bahram Jambar Nooshin
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahereh Tayebi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirhesam Babajani
- Department of Tissue Engineering, School of Medicine, Shahroud, University of Medical Sciences, Shahroud, Iran
| | - Mohammad-Mehdi Khani
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran,P.O.Box: 1985717446Department of Tissue Engineering and Applied Cell SciencesSchool of Advanced
Technologies in MedicineShahid Beheshti University of Medical SciencesTehranIranP.O.Box: 1985717446Department of PharmacologySchool of MedicineShahid Beheshti University of Medical SciencesTehranIran
Emails:,
| | - Hassan Niknejad
- Department of Tissue Engineering, School of Medicine, Shahroud, University of Medical Sciences, Shahroud, Iran,P.O.Box: 1985717446Department of Tissue Engineering and Applied Cell SciencesSchool of Advanced
Technologies in MedicineShahid Beheshti University of Medical SciencesTehranIranP.O.Box: 1985717446Department of PharmacologySchool of MedicineShahid Beheshti University of Medical SciencesTehranIran
Emails:,
| |
Collapse
|
29
|
Tam PKH, Wong KKY, Atala A, Giobbe GG, Booth C, Gruber PJ, Monone M, Rafii S, Rando TA, Vacanti J, Comer CD, Elvassore N, Grikscheit T, de Coppi P. Regenerative medicine: postnatal approaches. THE LANCET. CHILD & ADOLESCENT HEALTH 2022; 6:654-666. [PMID: 35963270 DOI: 10.1016/s2352-4642(22)00193-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 05/20/2022] [Accepted: 05/27/2022] [Indexed: 06/15/2023]
Abstract
Paper 2 of the paediatric regenerative medicine Series focuses on recent advances in postnatal approaches. New gene, cell, and niche-based technologies and their combinations allow structural and functional reconstitution and simulation of complex postnatal cell, tissue, and organ hierarchies. Organoid and tissue engineering advances provide human disease models and novel treatments for both rare paediatric diseases and common diseases affecting all ages, such as COVID-19. Preclinical studies for gastrointestinal disorders are directed towards oesophageal replacement, short bowel syndrome, enteric neuropathy, biliary atresia, and chronic end-stage liver failure. For respiratory diseases, beside the first human tracheal replacement, more complex tissue engineering represents a promising solution to generate transplantable lungs. Genitourinary tissue replacement and expansion usually involve application of biocompatible scaffolds seeded with patient-derived cells. Gene and cell therapy approaches seem appropriate for rare paediatric diseases of the musculoskeletal system such as spinal muscular dystrophy, whereas congenital diseases of complex organs, such as the heart, continue to challenge new frontiers of regenerative medicine.
Collapse
Affiliation(s)
- Paul Kwong Hang Tam
- Faculty of Medicine, Macau University of Science and Technology, Macau Special Administrative Region, China; Division of Paediatric Surgery, Department of Surgery, Queen Mary Hospital, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Kenneth Kak Yuen Wong
- Division of Paediatric Surgery, Department of Surgery, Queen Mary Hospital, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| | - Giovanni Giuseppe Giobbe
- Stem Cell and Regenerative Medicine Section, Developmental Biology and Cancer Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Claire Booth
- Stem Cell and Regenerative Medicine Section, Developmental Biology and Cancer Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Peter J Gruber
- Department of Surgery, Yale University, New Haven, CT, USA
| | - Mimmi Monone
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Shahin Rafii
- Ansary Stem Cell Institute, Department of Medicine, Division of Regenerative Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Thomas A Rando
- Paul F Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph Vacanti
- Department of Pediatric Surgery, Laboratory for Tissue Engineering and Organ Fabrication, Harvard Medical School, Massachusetts General Hospital, Mass General Hospital for Children, Boston, MA, USA
| | - Carly D Comer
- Department of Pediatric Surgery, Laboratory for Tissue Engineering and Organ Fabrication, Harvard Medical School, Massachusetts General Hospital, Mass General Hospital for Children, Boston, MA, USA
| | - Nicola Elvassore
- Stem Cell and Regenerative Medicine Section, Developmental Biology and Cancer Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK; Department of Industrial Engineering, University of Padova, Padova, Italy
| | - Tracy Grikscheit
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Paolo de Coppi
- Stem Cell and Regenerative Medicine Section, Developmental Biology and Cancer Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK; Department of Specialist Neonatal and Paediatric Surgery, Great Ormond Street Hospital, London, UK.
| |
Collapse
|
30
|
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
|
31
|
Willemse J, van Tienderen G, van Hengel E, Schurink I, van der Ven D, Kan Y, de Ruiter P, Rosmark O, Westergren-Thorsson G G, Schneeberger K, van der Eerden B, Roest H, Spee B, van der Laan L, de Jonge J, Verstegen M. Hydrogels derived from decellularized liver tissue support the growth and differentiation of cholangiocyte organoids. Biomaterials 2022; 284:121473. [PMID: 35344800 DOI: 10.1016/j.biomaterials.2022.121473] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 03/04/2022] [Accepted: 03/15/2022] [Indexed: 02/07/2023]
Abstract
Human cholangiocyte organoids are promising for regenerative medicine applications, such as repair of damaged bile ducts. However, organoids are typically cultured in mouse tumor-derived basement membrane extracts (BME), which is poorly defined, highly variable and limits the direct clinical applications of organoids in patients. Extracellular matrix (ECM)-derived hydrogels prepared from decellularized human or porcine livers are attractive alternative culture substrates. Here, the culture and expansion of human cholangiocyte organoids in liver ECM(LECM)-derived hydrogels is described. These hydrogels support proliferation of cholangiocyte organoids and maintain the cholangiocyte-like phenotype. The use of LECM hydrogels does not significantly alter the expression of selected genes or proteins, such as the cholangiocyte marker cytokeratin-7, and no species-specific effect is found between human or porcine LECM hydrogels. Proliferation rates of organoids cultured in LECM hydrogels are lower, but the differentiation capacity of the cholangiocyte organoids towards hepatocyte-like cells is not altered by the presence of tissue-specific ECM components. Moreover, human LECM extracts support the expansion of ICO in a dynamic culture set up without the need for laborious static culture of organoids in hydrogel domes. Liver ECM hydrogels can successfully replace tumor-derived BME and can potentially unlock the full clinical potential of human cholangiocyte organoids.
Collapse
Affiliation(s)
- Jorke Willemse
- Department of Surgery, Transplant Institute, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Gilles van Tienderen
- Department of Surgery, Transplant Institute, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Eline van Hengel
- Department of Surgery, Transplant Institute, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Ivo Schurink
- Department of Surgery, Transplant Institute, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Diana van der Ven
- Department of Surgery, Transplant Institute, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Yik Kan
- Department of Surgery, Transplant Institute, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Petra de Ruiter
- Department of Surgery, Transplant Institute, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Oskar Rosmark
- Lung Biology, Department Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Kerstin Schneeberger
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Bram van der Eerden
- Department of Internal Medicine, Calcium and Bone Metabolism, Erasmus MC-University, Rotterdam, the Netherlands
| | - Henk Roest
- Department of Surgery, Transplant Institute, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Bart Spee
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Luc van der Laan
- Department of Surgery, Transplant Institute, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Jeroen de Jonge
- Department of Surgery, Transplant Institute, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Monique Verstegen
- Department of Surgery, Transplant Institute, Erasmus MC, University Medical Center Rotterdam, the Netherlands.
| |
Collapse
|
32
|
Li K, Tharwat M, Larson EL, Felgendreff P, Hosseiniasl SM, Rmilah AA, Safwat K, Ross JJ, Nyberg SL. Re-Endothelialization of Decellularized Liver Scaffolds: A Step for Bioengineered Liver Transplantation. Front Bioeng Biotechnol 2022; 10:833163. [PMID: 35360393 PMCID: PMC8960611 DOI: 10.3389/fbioe.2022.833163] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/11/2022] [Indexed: 12/12/2022] Open
Abstract
Bioengineered livers (BELs) are an attractive therapeutic alternative to address the donor organ shortage for liver transplantation. The goal of BELs technology aims at replacement or regeneration of the native human liver. A variety of approaches have been proposed for tissue engineering of transplantable livers; the current review will highlight the decellularization-recellularization approach to BELs. For example, vascular patency and appropriate cell distribution and expansion are critical components in the production of successful BELs. Proper solutions to these components of BELs have challenged its development. Several strategies, such as heparin immobilization, heparin-gelatin, REDV peptide, and anti-CD31 aptamer have been developed to extend the vascular patency of revascularized bioengineered livers (rBELs). Other novel methods have been developed to enhance cell seeding of parenchymal cells and to increase graft functionality during both bench and in vivo perfusion. These enhanced methods have been associated with up to 15 days of survival in large animal (porcine) models of heterotopic transplantation but have not yet permitted extended survival after implantation of BELs in the orthotopic position. This review will highlight both the remaining challenges and the potential for clinical application of functional bioengineered grafts.
Collapse
Affiliation(s)
- Kewei Li
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- Department of Pediatric Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Mohammad Tharwat
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- General Surgery Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ellen L. Larson
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | - Philipp Felgendreff
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- Department for General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany
| | | | - Anan Abu Rmilah
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | - Khaled Safwat
- General Surgery Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | | | - Scott L. Nyberg
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
- *Correspondence: Scott L. Nyberg,
| |
Collapse
|
33
|
Design by Nature: Emerging Applications of Native Liver Extracellular Matrix for Cholangiocyte Organoid-Based Regenerative Medicine. Bioengineering (Basel) 2022; 9:bioengineering9030110. [PMID: 35324799 PMCID: PMC8945468 DOI: 10.3390/bioengineering9030110] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/25/2022] [Accepted: 03/04/2022] [Indexed: 12/14/2022] Open
Abstract
Organoid technology holds great promise for regenerative medicine. Recent studies show feasibility for bile duct tissue repair in humans by successfully transplanting cholangiocyte organoids in liver grafts during perfusion. Large-scale expansion of cholangiocytes is essential for extending these regenerative medicine applications. Human cholangiocyte organoids have a high and stable proliferation capacity, making them an attractive source of cholangiocytes. Commercially available basement membrane extract (BME) is used to expand the organoids. BME allows the cells to self-organize into 3D structures and stimulates cell proliferation. However, the use of BME is limiting the clinical applications of the organoids. There is a need for alternative tissue-specific and clinically relevant culture substrates capable of supporting organoid proliferation. Hydrogels prepared from decellularized and solubilized native livers are an attractive alternative for BME. These hydrogels can be used for the culture and expansion of cholangiocyte organoids in a clinically relevant manner. Moreover, the liver-derived hydrogels retain tissue-specific aspects of the extracellular microenvironment. They are composed of a complex mixture of bioactive and biodegradable extracellular matrix (ECM) components and can support the growth of various hepatobiliary cells. In this review, we provide an overview of the clinical potential of native liver ECM-based hydrogels for applications with human cholangiocyte organoids. We discuss the current limitations of BME for the clinical applications of organoids and how native ECM hydrogels can potentially overcome these problems in an effort to unlock the full regenerative clinical potential of the organoids.
Collapse
|
34
|
Three-dimensional scaffolds for tissue bioengineering cartilages. Biocybern Biomed Eng 2022. [DOI: 10.1016/j.bbe.2022.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
35
|
Main Applications and Recent Research Progresses of Additive Manufacturing in Dentistry. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5530188. [PMID: 35252451 PMCID: PMC8894006 DOI: 10.1155/2022/5530188] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 12/16/2021] [Accepted: 01/28/2022] [Indexed: 12/13/2022]
Abstract
In recent ten years, with the fast development of digital and engineering manufacturing technology, additive manufacturing has already been more and more widely used in the field of dentistry, from the first personalized surgical guides to the latest personalized restoration crowns and root implants. In particular, the bioprinting of teeth and tissue is of great potential to realize organ regeneration and finally improve the life quality. In this review paper, we firstly presented the workflow of additive manufacturing technology. Then, we summarized the main applications and recent research progresses of additive manufacturing in dentistry. Lastly, we sketched out some challenges and future directions of additive manufacturing technology in dentistry.
Collapse
|
36
|
Morales-Guerrero NA, Varela-Echavarría A, Lozano Flores C, Vázquez-Cuevas FG, Velázquez-Miranda E, Reyes-López JV, García-Solís P, Solís-S JC, Hernández-Montiel HL. A new strategy for the decellularization of whole organs by hydrostatic pressure. Biotechnol Prog 2022; 38:e3248. [PMID: 35201677 DOI: 10.1002/btpr.3248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 11/06/2022]
Abstract
Tissue engineering has been able to develop novel decellularization-recellularization techniques, which facilitates the research for the generation of functional organs. This is based in the initial obtention of the organ's extracellular matrix (ECM). Therefore, any improvement in the decellularization process would have a positive impact in the results of the recellularization process. Nevertheless, commonly the methods and equipment employed for this process are expensive and thus limit the access of this technique to various research groups globally. AIM To develop a decellularization technique with the exclusive use of hydrostatic pressure of detergent solutions, to have an easily accessible and low-cost technique that meets the basic requirements of acellularity and functionality of the ECM. METHODS This experimental study was performed in 10 male Wistar rats, obtaining the liver to carry out serial washes, with 1, 2 and 3% Triton X-100 solutions and 0.1% SDS. The washes were performed by using a Gravity Perfusion System (GPS), which assured us a continuous hydrostatic pressure of 7.5 mmHg. The obtained ECM was processed using stains and immunostaining to determine the residual cell content and preservation of its components. RESULTS The staining showed a removal of cellular and nuclear components of approximately 97% of the acellular ECM, with an adequate three-dimensional pattern of collagen and proteoglycans. Furthermore, the acellular ECM allowed the viability of a primary hepatocyte culture. CONCLUSIONS The use of the GPS decellularization technique allowed us to obtain an acellular and functional ECM, drastically reducing experimentation costs. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Nelly A Morales-Guerrero
- Department of Biomedical Research, School of Medicine, Autonomous University of Queretaro, Qro., Mexico
| | | | - Carlos Lozano Flores
- Institute of Neurobiology, National Autonomous University of Mexico, Qro., Mexico
| | | | | | - Julián V Reyes-López
- Laboratory of Neurobiology and Cellular Bioengineering, Neurodiagnostic and Rehabilitation Unit "Dr. Moisés López González ", Faculty of Natural Sciences, Autonomous University of Querétaro
| | - Pablo García-Solís
- Department of Biomedical Research, School of Medicine, Autonomous University of Queretaro, Qro., Mexico
| | - Juan Carlos Solís-S
- Department of Biomedical Research, School of Medicine, Autonomous University of Queretaro, Qro., Mexico
| | - Hebert Luis Hernández-Montiel
- Laboratory of Neurobiology and Cellular Bioengineering, Neurodiagnostic and Rehabilitation Unit "Dr. Moisés López González ", Faculty of Natural Sciences, Autonomous University of Querétaro
| |
Collapse
|
37
|
Dias ML, Paranhos BA, Ferreira JRP, Fonseca RJC, Batista CMP, Martins-Santos R, de Andrade CBV, Faccioli LAP, da Silva AC, Nogueira FCS, Domont GB, Dos Santos Goldenberg RC. Improving hemocompatibility of decellularized liver scaffold using Custodiol solution. BIOMATERIALS ADVANCES 2022; 133:112642. [PMID: 35034821 DOI: 10.1016/j.msec.2022.112642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/02/2021] [Accepted: 01/02/2022] [Indexed: 10/19/2022]
Abstract
Organ decellularization is one of the most promising approaches of tissue engineering to overcome the shortage of organs available for transplantation. However, there are key hurdles that still hinder its clinical application, and the lack of hemocompatibility of decellularized materials is a central one. In this work, we demonstrate that Custodiol (HTK solution), a common solution used in organ transplantation, increased the hemocompatibility of acellular scaffolds obtained from rat livers. We showed that Custodiol inhibited ex vivo, in vitro, and in vivo blood coagulation to such extent that allowed successful transplantation of whole-liver scaffolds into recipient animals. Scaffolds previously perfused with Custodiol showed no signs of platelet aggregation and maintained in vitro and in vivo cellular compatibility. Proteomic analysis revealed that proteins related to platelet aggregation were reduced in Custodiol samples while control samples were enriched with thrombogenicity-related proteins. We also identified distinct components that could potentially be involved with this anti-thrombogenic effect and thus require further investigation. Therefore, Custodiol perfusion emerge as a promising strategy to reduce the thrombogenicity of decellularized biomaterials and could benefit several applications of whole-organ tissue engineering.
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, UFRJ, 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, UFRJ, Rio de Janeiro, Brasil
| | - Juliana Ribeiro Pinheiro Ferreira
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil; Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Brasil
| | - Roberto José Castro Fonseca
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil; Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Cíntia Marina Paz Batista
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Ricardo Martins-Santos
- 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, UFRJ, Rio de Janeiro, Brasil
| | - Cherley Borba Vieira de Andrade
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil; Departamento de Histologia e Embriologia, Universidade do Estado do Rio de Janeiro, UERJ, Rio de Janeiro, RJ, Brasil
| | - Lanuza Alaby Pinheiro Faccioli
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil; Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | - Gilberto Barbosa Domont
- Laboratório de Proteômica /LADETEC, 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, UFRJ, Rio de Janeiro, Brasil.
| |
Collapse
|
38
|
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
|
39
|
Ladeira B, Custodio C, Mano J. Core-Shell Microcapsules: Biofabrication and Potential Applications in Tissue Engineering and Regenerative Medicine. Biomater Sci 2022; 10:2122-2153. [DOI: 10.1039/d1bm01974k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The construction of biomaterial scaffolds that accurately recreate the architecture of living tissues in vitro is a major challenge in the field of tissue engineering and regenerative medicine. Core-shell microcapsules...
Collapse
|
40
|
Steinle H, Weber J, Stoppelkamp S, Große-Berkenbusch K, Golombek S, Weber M, Canak-Ipek T, Trenz SM, Schlensak C, Avci-Adali M. Delivery of synthetic mRNAs for tissue regeneration. Adv Drug Deliv Rev 2021; 179:114007. [PMID: 34710530 DOI: 10.1016/j.addr.2021.114007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/03/2021] [Accepted: 10/12/2021] [Indexed: 02/06/2023]
Abstract
In recent years, nucleic acid-based therapeutics have gained increasing importance as novel treatment options for disease prevention and treatment. Synthetic messenger RNAs (mRNAs) are promising nucleic acid-based drugs to transiently express desired proteins that are missing or defective. Recently, synthetic mRNA-based vaccines encoding viral proteins have been approved for emergency use against COVID-19. Various types of vehicles, such as lipid nanoparticles (LNPs) and liposomes, are being investigated to enable the efficient uptake of mRNA molecules into desired cells. In addition, the introduction of novel chemical modifications into mRNAs increased the stability, enabled the modulation of nucleic acid-based drugs, and increased the efficiency of mRNA-based therapeutic approaches. In this review, novel and innovative strategies for the delivery of synthetic mRNA-based therapeutics for tissue regeneration are discussed. Moreover, with this review, we aim to highlight the versatility of synthetic mRNA molecules for various applications in the field of regenerative medicine and also discuss translational challenges and required improvements for mRNA-based drugs.
Collapse
Affiliation(s)
- Heidrun Steinle
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Josefin Weber
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Sandra Stoppelkamp
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Katharina Große-Berkenbusch
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Sonia Golombek
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Marbod Weber
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Tuba Canak-Ipek
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Sarah-Maria Trenz
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Christian Schlensak
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Meltem Avci-Adali
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany.
| |
Collapse
|
41
|
Ali S, Haque N, Azhar Z, Saeinasab M, Sefat F. Regenerative Medicine of Liver: Promises, Advances and Challenges. Biomimetics (Basel) 2021; 6:biomimetics6040062. [PMID: 34698078 PMCID: PMC8544204 DOI: 10.3390/biomimetics6040062] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/06/2021] [Accepted: 10/14/2021] [Indexed: 12/16/2022] Open
Abstract
Liver tissue engineering is a rapidly developing field which combines the novel use of liver cells, appropriate biochemical factors, and engineering principles, in order to replace or regenerate damaged liver tissue or the organ. The aim of this review paper is to critically investigate different possible methods to tackle issues related with liver diseases/disorders mainly using regenerative medicine. In this work the various regenerative treatment options are discussed, for improving the prognosis of chronic liver disorders. By reviewing existing literature, it is apparent that the current popular treatment option is liver transplantation, although the breakthroughs of stem cell-based therapy and bioartificial liver technology make them a promising alternative.
Collapse
Affiliation(s)
- Saiful Ali
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford BD7 1DP, UK; (S.A.); (N.H.); (Z.A.)
| | - Nasira Haque
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford BD7 1DP, UK; (S.A.); (N.H.); (Z.A.)
| | - Zohya Azhar
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford BD7 1DP, UK; (S.A.); (N.H.); (Z.A.)
| | - Morvarid Saeinasab
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad 9177948974, Iran;
| | - Farshid Sefat
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford BD7 1DP, UK; (S.A.); (N.H.); (Z.A.)
- Interdisciplinary Research Centre in Polymer Science & Technology (Polymer IRC), University of Bradford, Bradford BD7 1DP, UK
- Correspondence: ; Tel.: +44-(0)-1274-233679 or +44-(0)-781-381-7460
| |
Collapse
|
42
|
Gao Y, Callanan A. Influence of surface topography on PCL electrospun scaffolds for liver tissue engineering. J Mater Chem B 2021; 9:8081-8093. [PMID: 34491259 PMCID: PMC8493469 DOI: 10.1039/d1tb00789k] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 08/24/2021] [Indexed: 01/16/2023]
Abstract
Severe liver disease is one of the most common causes of death globally. Currently, whole organ transplantation is the only therapeutic method for end-stage liver disease treatment, however, the need for donor organs far outweighs demand. Recently liver tissue engineering is starting to show promise for alleviating part of this problem. Electrospinning is a well-known method to fabricate a nanofibre scaffold which mimics the natural extracellular matrix that can support cell growth. This study aims to investigate liver cell responses to topographical features on electrospun fibres. Scaffolds with large surface depression (2 μm) (LSD), small surface depression (0.37 μm) (SSD), and no surface depression (NSD) were fabricated by using a solvent-nonsolvent system. A liver cell line (HepG2) was seeded onto the scaffolds for up to 14 days. The SSD group exhibited higher levels of cell viability and DNA content compared to the other groups. Additionally, the scaffolds promoted gene expression of albumin, with all cases having similar levels, while the cell growth rate was altered. Furthermore, the scaffold with depressions showed 0.8 MPa higher ultimate tensile strength compared to the other groups. These results suggest that small depressions might be preferred by HepG2 cells over smooth and large depression fibres and highlight the potential for tailoring liver cell responses.
Collapse
Affiliation(s)
- Yunxi Gao
- Institute of Bioengineering, School of Engineering, The University of Edinburgh, Edinburgh, UK.
| | - Anthony Callanan
- Institute of Bioengineering, School of Engineering, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
43
|
Characterization of Indonesia Decellularized Liver Cubes Scaffold using Scanning Electron Microscopy. JOURNAL OF BIOMIMETICS BIOMATERIALS AND BIOMEDICAL ENGINEERING 2021. [DOI: 10.4028/www.scientific.net/jbbbe.52.38] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Liver biological scaffold was developed in order to resemble native liver tissue environment. It can be achieved by decellularizing native liver tissue that will remove cells and preserve extracellular matrix (ECM). Furthermore, ECM fibers are arranged in a special pattern, which affect liver cell polarity and topography that are important for cells’ implantation, proliferation and differentiation. Therefore, the aim of this study was to evaluate liver cube scaffold topography that was decellularized with fixed multiple sites syringe injection (Indonesia patent number: S00201907930).Rat liver cubes (n=3) underwent decellularization with Ethylene Glycol Tetraacetic Acid (EGTA) immersion and increased Sodium Dodecyl Sulfate (SDS) concentrations using previous multiple sites syringe injection protocol study. Deoxyribonucleic Acid (DNA) concentrations were measured to confirm less DNA materials remaining in scaffolds. Scanning Electron Microscope (SEM) analysis of scaffolds were conducted for topographic characterization compared to undecellularized liver control. Molecular analysis of DNA concentration showed complete removal of DNA material. SEM analysis gave appearance of intact liver cube scaffold microarchitecture. Liver cubes decellularization using multiple sites syringe injection showed good topographic liver scaffold characterization.
Collapse
|
44
|
Kim S, Han G, Hwang D, Won D, Shin Y, Kim C, Kang JM, Park J, Jung H, Park W, Yun J. Design and Usability Evaluations of a 3D-Printed Implantable Drug Delivery Device for Acute Liver Failure in Preclinical Settings. Adv Healthc Mater 2021; 10:e2100497. [PMID: 34160141 DOI: 10.1002/adhm.202100497] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/18/2021] [Indexed: 12/11/2022]
Abstract
Acute liver failure (ALF) requiring liver transplantation is a disease that occurs due to rapid hepatocellular dysfunction. As liver transplantation has various limitations, including donor scarcity, high cost, and immuno-incompatibility, continuous local delivery of biopharmaceuticals to the liver tissue can be a promising ALF treatment option. Here, the in vivo safety and usability of a 3D-printed implantable drug delivery device for effective ALF treatment is evaluated. The implantable reservoir consists of a 3D-printed container and a semipermeable membrane for repeated administrations of drugs, specifically to the liver tissue. The physical stability and function of the 3D-printed reservoir are confirmed by the mechanical properties and in vitro drug release test, respectively. In mice implanted with the reservoir system, mortality, weight changes, clinical signs, hematological and serum biochemical changes, and organ weight changes are not observed, suggesting no foreign body reaction. The usability of the reservoir system is further evaluated using an ALF model of 70% hepatectomized mice treated with N-acetylcysteine through the system, showing cell-specific regeneration and significant liver injury alleviation. Overall, the 3D-printed reservoir system is safe for studying the therapeutic potential of ALF treatment, and it can be used for the delivery of various active pharmaceutical ingredients.
Collapse
Affiliation(s)
- Shin‐Young Kim
- Department of Biotechnology The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
| | - Ginam Han
- Department of Biotechnology The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
- Department of Biomedical‐Chemical Engineering The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
| | - Da‐Bin Hwang
- Department of Biotechnology The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
| | - Dong‐Hoon Won
- Department of Biotechnology The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
| | - Yoo‐Sub Shin
- Department of Biotechnology The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
| | - Changuk Kim
- Department of Biotechnology The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
| | - Jeon Min Kang
- Biomedical Engineering Research Center Asan Institute for Life Sciences Asan Medical Center 88 Olympic‐ro 43‐gil Songpa‐gu Seoul 05505 Republic of Korea
| | - Jung‐Hoon Park
- Biomedical Engineering Research Center Asan Institute for Life Sciences Asan Medical Center 88 Olympic‐ro 43‐gil Songpa‐gu Seoul 05505 Republic of Korea
| | - Hyun‐Do Jung
- Department of Biotechnology The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
- Department of Biomedical‐Chemical Engineering The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
| | - Wooram Park
- Department of Biotechnology The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
- Department of Biomedical‐Chemical Engineering The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
| | - Jun‐Won Yun
- Department of Biotechnology The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
- Department of Medical and Biological Sciences The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
| |
Collapse
|
45
|
Ahmad Raus R, Wan Nawawi WMF, Nasaruddin RR. Alginate and alginate composites for biomedical applications. Asian J Pharm Sci 2021; 16:280-306. [PMID: 34276819 PMCID: PMC8261255 DOI: 10.1016/j.ajps.2020.10.001] [Citation(s) in RCA: 199] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/26/2020] [Accepted: 10/07/2020] [Indexed: 12/22/2022] Open
Abstract
Alginate is an edible heteropolysaccharide that abundantly available in the brown seaweed and the capsule of bacteria such as Azotobacter sp. and Pseudomonas sp. Owing to alginate gel forming capability, it is widely used in food, textile and paper industries; and to a lesser extent in biomedical applications as biomaterial to promote wound healing and tissue regeneration. This is evident from the rising use of alginate-based dressing for heavily exuding wound and their mass availability in the market nowadays. However, alginate also has limitation. When in contact with physiological environment, alginate could gelate into softer structure, consequently limits its potential in the soft tissue regeneration and becomes inappropriate for the usage related to load bearing body parts. To cater this problem, wide range of materials have been added to alginate structure, producing sturdy composite materials. For instance, the incorporation of adhesive peptide and natural polymer or synthetic polymer to alginate moieties creates an improved composite material, which not only possesses better mechanical properties compared to native alginate, but also grants additional healing capability and promote better tissue regeneration. In addition, drug release kinetic and cell viability can be further improved when alginate composite is used as encapsulating agent. In this review, preparation of alginate and alginate composite in various forms (fibre, bead, hydrogel, and 3D-printed matrices) used for biomedical application is described first, followed by the discussion of latest trend related to alginate composite utilization in wound dressing, drug delivery, and tissue engineering applications.
Collapse
Affiliation(s)
- Raha Ahmad Raus
- Department of Biotechnology Engineering, International Islamic University Malaysia, Kuala Lumpur 50728, Malaysia
| | - Wan Mohd Fazli Wan Nawawi
- Department of Biotechnology Engineering, International Islamic University Malaysia, Kuala Lumpur 50728, Malaysia
- Nanoscience and Nanotechnology Research Group (NanoRG), International Islamic University Malaysia, Kuala Lumpur 50728, Malaysia
| | - Ricca Rahman Nasaruddin
- Department of Biotechnology Engineering, International Islamic University Malaysia, Kuala Lumpur 50728, Malaysia
- Nanoscience and Nanotechnology Research Group (NanoRG), International Islamic University Malaysia, Kuala Lumpur 50728, Malaysia
| |
Collapse
|
46
|
Applicability of 3D-printed models in hepatobiliary surgey: results from "LIV3DPRINT" multicenter study. HPB (Oxford) 2021; 23:675-684. [PMID: 33071150 DOI: 10.1016/j.hpb.2020.09.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 05/26/2020] [Accepted: 09/25/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Hepatobiliary resections are challenging due to the complex liver anatomy. Three-dimensional printing (3DP) has gained popularity due to its ability to produce anatomical models based on the characteristics of each patient. METHODS A multicenter study was conducted on complex hepatobiliary tumours. The endpoint was to validate 3DP model accuracy from original image sources for application in the teaching, patient-communication, and planning of hepatobiliary surgery. RESULTS Thirty-five patients from eight centers were included. Process testing between 3DP and CT/MRI presented a considerable degree of similarity in vascular calibers (0.22 ± 1.8 mm), and distances between the tumour and vessel (0.31 ± 0.24 mm). The Dice Similarity Coefficient was 0.92, with a variation of 2%. Bland-Altman plots also demonstrated an agreement between 3DP and the surgical specimen with the distance of the resection margin (1.15 ± 1.52 mm). Professionals considered 3DP at a positive rate of 0.89 (95%CI; 0.73-0.95). According to student's distribution a higher success rate was reached with 3DP (median:0.9, IQR: 0.8-1) compared with CT/MRI or 3D digital imaging (P = 0.01). CONCLUSION 3DP hepatic models present a good correlation compared with CT/MRI and surgical pathology and they are useful for education, understanding, and surgical planning, but does not necessarily affect the surgical outcome.
Collapse
|
47
|
The triad of nanotechnology, cell signalling, and scaffold implantation for the successful repair of damaged organs: An overview on soft-tissue engineering. J Control Release 2021; 332:460-492. [DOI: 10.1016/j.jconrel.2021.02.036] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 02/26/2021] [Accepted: 02/28/2021] [Indexed: 12/11/2022]
|
48
|
Muttillo EM, Felli E. Can the mutational status of KRAS drive the treatment of colorectal liver metastases? HPB (Oxford) 2021; 23:643. [PMID: 33514493 DOI: 10.1016/j.hpb.2021.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 01/05/2021] [Indexed: 12/12/2022]
Affiliation(s)
- Edoardo M Muttillo
- Department of Surgical Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - Emanuele Felli
- HPB Unit, Digestive Surgery Department, Nouvel Hopital Civil, University of Strasbourg, Strasbourg, France.
| |
Collapse
|
49
|
Roy HS, Singh R, Ghosh D. SARS-CoV-2 and tissue damage: current insights and biomaterial-based therapeutic strategies. Biomater Sci 2021; 9:2804-2824. [PMID: 33666206 DOI: 10.1039/d0bm02077j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The effect of SARS-CoV-2 infection on humanity has gained worldwide attention and importance due to the rapid transmission, lack of treatment options and high mortality rate of the virus. While scientists across the world are searching for vaccines/drugs that can control the spread of the virus and/or reduce the risks associated with infection, patients infected with SARS-CoV-2 have been reported to have tissue/organ damage. With most tissues/organs having limited regenerative potential, interventions that prevent further damage or facilitate healing would be helpful. In the past few decades, biomaterials have gained prominence in the field of tissue engineering, in view of their major role in the regenerative process. Here we describe the effect of SARS-CoV-2 on multiple tissues/organs, and provide evidence for the positive role of biomaterials in aiding tissue repair. These findings are further extrapolated to explore their prospects as a therapeutic platform to address the tissue/organ damage that is frequently observed during this viral outbreak. This study suggests that the biomaterial-based approach could be an effective strategy for regenerating tissues/organs damaged by SARS-CoV-2.
Collapse
Affiliation(s)
- Himadri Shekhar Roy
- Department of Biological Science, Institute of Nanoscience and Technology (INST), Habitat Centre, Sector 64, Phase 10, Mohali-160062, Punjab, India.
| | - Rupali Singh
- Department of Biological Science, Institute of Nanoscience and Technology (INST), Habitat Centre, Sector 64, Phase 10, Mohali-160062, Punjab, India.
| | - Deepa Ghosh
- Department of Biological Science, Institute of Nanoscience and Technology (INST), Habitat Centre, Sector 64, Phase 10, Mohali-160062, Punjab, India.
| |
Collapse
|
50
|
Taymour R, Kilian D, Ahlfeld T, Gelinsky M, Lode A. 3D bioprinting of hepatocytes: core-shell structured co-cultures with fibroblasts for enhanced functionality. Sci Rep 2021; 11:5130. [PMID: 33664366 PMCID: PMC7933206 DOI: 10.1038/s41598-021-84384-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 02/16/2021] [Indexed: 01/31/2023] Open
Abstract
With the aim of understanding and recapitulating cellular interactions of hepatocytes in their physiological microenvironment and to generate an artificial 3D in vitro model, a co-culture system using 3D extrusion bioprinting was developed. A bioink based on alginate and methylcellulose (algMC) was first shown to be suitable for bioprinting of hepatocytes; the addition of Matrigel to algMC enhanced proliferation and morphology of them in monophasic scaffolds. Towards a more complex system that allows studying cellular interactions, we applied core-shell bioprinting to establish tailored 3D co-culture models for hepatocytes. The bioinks were specifically functionalized with natural matrix components (based on human plasma, fibrin or Matrigel) and used to co-print fibroblasts and hepatocytes in a spatially defined, coaxial manner. Fibroblasts acted as supportive cells for co-cultured hepatocytes, stimulating the expression of certain biomarkers of hepatocytes like albumin. Furthermore, matrix functionalization positively influenced both cell types in their respective compartments by enhancing their adhesion, viability, proliferation and function. In conclusion, we established a functional co-culture model with independently tunable compartments for different cell types via core-shell bioprinting. This provides the basis for more complex in vitro models allowing co-cultivation of hepatocytes with other liver-specific cell types to closely resemble the liver microenvironment.
Collapse
Affiliation(s)
- Rania Taymour
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - David Kilian
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Tilman Ahlfeld
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Anja Lode
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.
| |
Collapse
|