1
|
Cong R, Lu C, Li X, Xu Z, Wang Y, Sun S. Tumor organoids in cancer medicine: from model systems to natural compound screening. PHARMACEUTICAL BIOLOGY 2025; 63:89-109. [PMID: 39893515 PMCID: PMC11789228 DOI: 10.1080/13880209.2025.2458149] [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: 08/26/2024] [Revised: 12/04/2024] [Accepted: 01/17/2025] [Indexed: 02/04/2025]
Abstract
CONTEXT The advent of tissue engineering and biomedical techniques has significantly advanced the development of three-dimensional (3D) cell culture systems, particularly tumor organoids. These self-assembled 3D cell clusters closely replicate the histopathological, genetic, and phenotypic characteristics of primary tissues, making them invaluable tools in cancer research and drug screening. OBJECTIVE This review addresses the challenges in developing in vitro models that accurately reflect tumor heterogeneity and explores the application of tumor organoids in cancer research, with a specific focus on the screening of natural products for antitumor therapies. METHODS This review synthesizes information from major databases, including Chemical Abstracts, Medicinal and Aromatic Plants Abstracts, ScienceDirect, Google Scholar, Scopus, PubMed and Springer Link. Publications were selected without date restrictions, using terms such as 'organoid', 'natural product', 'pharmacological', 'extract', 'nanomaterial' and 'traditional uses'. Articles related to agriculture, ecology, synthetic work or published in languages other than English were excluded. RESULTS AND CONCLUSIONS The review identifies key challenges related to the efficiency and variability of organoid generation and discusses ongoing efforts to enhance their predictive capabilities in drug screening and personalized medicine. Recent studies utilizing patient-derived organoid models for natural compound screening are highlighted, demonstrating the potential of these models in developing new classes of anticancer agents. The integration of natural products with patient-derived organoid models presents a promising approach for discovering novel anticancer compounds and elucidating their mechanisms of action.
Collapse
Affiliation(s)
- Rong Cong
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Can Lu
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xinying Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhijie Xu
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yaqin Wang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Shusen Sun
- College of Pharmacy and Health Sciences, Western New England University, Springfield, MA, USA
| |
Collapse
|
2
|
Kuhn MR, Wolcott EA, Langer EM. Developments in gastrointestinal organoid cultures to recapitulate tissue environments. Front Bioeng Biotechnol 2025; 13:1521044. [PMID: 40313639 PMCID: PMC12043594 DOI: 10.3389/fbioe.2025.1521044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/21/2025] [Indexed: 05/03/2025] Open
Abstract
Culture platforms that closely mimic the spatial architecture, cellular diversity, and extracellular matrix composition of native tissues can serve as invaluable tools for a range of scientific discovery and biomedical applications. Organoids have emerged as a promising alternative to both traditional 2D cell culture and animal models, offering a physiologically relevant 3D culture system for studying human cell biology. Organoids provide a manipulable platform to investigate organ development and function as well as to model patient-specific phenotypes. This mini review examines various methods used for culturing organoids to model normal and disease conditions in gastrointestinal tissues. We focus on how the matrix composition and media formulations can impact cell signaling, altering the baseline cellular phenotypes as well as response to perturbations. We discuss future directions for optimizing organoid culture conditions to improve basic and translational potential.
Collapse
Affiliation(s)
- Madeline R. Kuhn
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
- Division of Oncological Sciences, Oregon Health and Science University, Portland, OR, United States
| | - Emma A. Wolcott
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
- Division of Oncological Sciences, Oregon Health and Science University, Portland, OR, United States
| | - Ellen M. Langer
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
- Division of Oncological Sciences, Oregon Health and Science University, Portland, OR, United States
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
3
|
Lai W, Geliang H, Bin X, Wang W. Effects of hydrogel stiffness and viscoelasticity on organoid culture: a comprehensive review. Mol Med 2025; 31:83. [PMID: 40033190 PMCID: PMC11877758 DOI: 10.1186/s10020-025-01131-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 02/14/2025] [Indexed: 03/05/2025] Open
Abstract
As an emerging technology, organoids are promising new tools for basic and translational research in disease. Currently, the culture of organoids relies mainly on a type of unknown composition scaffold, namely Matrigel, which may pose problems in studying the effect of mechanical properties on organoids. Hydrogels, a new material with adjustable mechanical properties, can adapt to current studies. In this review, we summarized the synthesis of recent advance in developing definite hydrogel scaffolds for organoid culture and identified the critical parameters for regulating mechanical properties. In addition, classified by different mechanical properties like stiffness and viscoelasticity, we concluded the effect of mechanical properties on the development of organoids and tumor organoids. We hope this review enhances the understanding of the development of organoids by hydrogels and provides more practical approaches to investigating them.
Collapse
Affiliation(s)
- Wei Lai
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hu Geliang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xu Bin
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Wei Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
4
|
Zhao J, Zhi Y, Ren H, Wang J, Zhao Y. Emerging biotechnologies for engineering liver organoids. Bioact Mater 2025; 45:1-18. [PMID: 39588483 PMCID: PMC11585797 DOI: 10.1016/j.bioactmat.2024.11.002] [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: 08/22/2024] [Revised: 11/02/2024] [Accepted: 11/02/2024] [Indexed: 11/27/2024] Open
Abstract
The engineering construction of the liver has attracted enormous attention. Organoids, as emerging miniature three-dimensional cultivation units, hold significant potential in the biomimetic simulation of liver structure and function. Despite notable successes, organoids still face limitations such as high variability and low maturity. To overcome these challenges, engineering strategies have been established to maintain organoid stability and enhance their efficacy, laying the groundwork for the development of advanced liver organoids. The present review comprehensively summarizes the construction of engineered liver organoids and their prospective applications in biomedicine. Initially, we briefly present the latest research progress on matrix materials that maintain the three-dimensional morphology of organoids. Next, we discuss the manipulative role of engineering technologies in organoid assembly. Additionally, we outline the impact of gene-level regulation on organoid growth and development. Further, we introduce the applications of liver organoids in disease modeling, drug screening and regenerative medicine. Lastly, we overview the current obstacles and forward-looking perspectives on the future of engineered liver organoids. We anticipate that ongoing innovations in engineered liver organoids will lead to significant advancements in medical applications.
Collapse
Affiliation(s)
- Junqi Zhao
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yue Zhi
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Haozhen Ren
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Jinglin Wang
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Yuanjin Zhao
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- Shenzhen Research Institute, Southeast University, Shenzhen, 518038, China
| |
Collapse
|
5
|
Li T, Bo RQ, Yan J, Johnson NL, Liao MT, Li Y, Chen Y, Lin J, Li J, Chu FH, Ding X. Global landscape of hepatic organoid research: A bibliometric and visual study. World J Hepatol 2025; 17:95624. [PMID: 40027550 PMCID: PMC11866153 DOI: 10.4254/wjh.v17.i2.95624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 10/11/2024] [Accepted: 11/12/2024] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Hepatic organoid-based modelling, through the elucidation of a range of in vivo biological processes and the recreation of the intricate liver microenvironment, is yielding groundbreaking insights into the pathophysiology and personalized medicine approaches for liver diseases. AIM This study was designed to analyse the global scientific output of hepatic organoid research and assess current achievements and future trends through bibliometric analysis. METHODS Articles were retrieved from the Web of Science Core Collection, and CiteSpace 6.3.R1 was employed to analyse the literature, including outputs, journals, and countries, among others. RESULTS Between 2010 and 2024, a total of 991 articles pertaining to hepatic organoid research were published. The journal Hepatology published the greatest number of papers, and journals with an impact factor greater than 10 constituted 60% of the top 10 journals. The United States and Utrecht University were identified as the most prolific country and institution, respectively. Clevers H emerged as the most prolific author, whereas Huch M had the highest number of cocitations, suggesting that both are ideal candidates for academic collaboration. Research on hepatic organoids has exhibited a progressive shift in focus, evolving from initial investigations into model building, differentiation research in stem cells, bile ducts, and progenitor cells, to a broader spectrum encompassing lipid metabolism, single-cell RNA sequencing, and therapeutic applications. The phrases exhibiting citation bursts from 2022 to 2024 include "drug resistance", "disease model", and "patient-derived tumor organoids". CONCLUSION Research on hepatic organoids has increased over the past decade and is expected to continue to grow. Key research areas include applications for liver diseases and drug development. Future trends likely to gain focus include patient-derived tumour organoids, disease modelling, and personalized medicine.
Collapse
Affiliation(s)
- Tao Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Rong-Qiang Bo
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Jun Yan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Nadia L Johnson
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Meng-Ting Liao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yuan Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yan Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Jie Lin
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Jian Li
- Department of Histology and Embryology, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Fu-Hao Chu
- Institute of Regulatory Science for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Xia Ding
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100700, China
| |
Collapse
|
6
|
van Sprang JF, Smits IPM, Nooten JCH, Fransen PPKH, Söntjens SHM, van Houtem MHCJ, Janssen HM, Rutten MGTA, Schotman MJG, Dankers PYW. From natural to synthetic hydrogels: how much biochemical complexity is required for mechanotransduction? J Mater Chem B 2025; 13:610-621. [PMID: 39601124 DOI: 10.1039/d4tb01774a] [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: 11/29/2024]
Abstract
The biochemical complexity of a material determines the biological response of cells triggered by a cell-material interaction. The degree in which this complexity influences basic cell-material interactions such as cell adhesion, spreading, and mechanotransduction is not entirely clear. To this end, we compared three different hydrogel systems, ranging from completely natural to synthetic, in their ability to induce mechanotransduction in kidney epithelial cells (HK-2). A natural hydrogel system was developed based on a decellularized kidney extracellular matrix (dECM). Supramolecular ureido-pyrimidinone (UPy)-glycinamide molecules, with self-associative behavior, were used for a hybrid and complete synthetic system. A hybrid system was engineered by co-assembling this monovalent UPy molecule with a hyaluronic acid, functionalized with ∼7 UPy-groups (UPy-HA), into a transient network. A similar approach was used for the synthetic hydrogel system, in which the multivalent UPy-HA was replaced with a bivalent UPy-PEG molecule with bioinert properties. Both hybrid and synthetic hydrogel systems were more mechanically tunable compared to the dECM hydrogel. The higher bulk stiffness in combination with the introduction of collagen type I mimicking UPy-additives allowed these materials to induce more nuclear yes-associated protein translocation in HK-2 cells compared to the biochemically complex dECM hydrogel. This demonstrated that minimal biochemical complexity is sufficient for inducing mechanotransduction.
Collapse
Affiliation(s)
- Johnick F van Sprang
- Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
- Laboratory of Cell and Tissue Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Imke P M Smits
- Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.
- Laboratory of Cell and Tissue Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Jasper C H Nooten
- Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Peter-Paul K H Fransen
- Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | | | | | - Henk M Janssen
- SyMO-Chem B.V., Den Dolech 2, 5612 AZ Eindhoven, The Netherlands
| | - Martin G T A Rutten
- Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Maaike J G Schotman
- Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - P Y W Dankers
- Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
- Laboratory of Cell and Tissue Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
7
|
Rabiei Dolatabadi Z, Saheli M, Solhjoo S, Hosseini M, Mousanejad E, Jafari M, Sheibani V, Nematollahi-Mahani SN. Amniotic membrane hydrogel as novel injectable platform in combination with metformin for treatment of sciatic nerve injury. J Appl Biomater Funct Mater 2025; 23:22808000251322865. [PMID: 40071878 DOI: 10.1177/22808000251322865] [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: 05/14/2025] Open
Abstract
Peripheral nerve tissue engineering is a field that uses cells, growth factors and biological scaffold material to provide a nutritional and physical support in the repair of nerve injuries. The specific properties of injectable human amniotic membrane-derived hydrogel including growth factors as well as anti-inflammatory and neuroprotective agents make it an ideal tool for nerve tissue repair, and metformin may also aid in nerve regeneration. The aim of this study was to investigate the effects of hydrogel derived from amniotic membrane (AM) along with metformin (MET) administration in the repair of sciatic nerve injury in male rats. We randomly divided 60 male rats into five groups. A control and four sciatic nerve compression groups including model; hydrogel; metformin and mix which received hydrogel and metformin. The recovery rate was assessed by Sciatic Functional Index (SFI), Static Sciatic Index (SSI) and von-frey test. Conduction velocity of the sciatic nerve was measured by Electrophysiological studies, and histological evaluations were performed 14 days after injury. SFI, SSI, latency time, remyelination rate and the expression of NF-200 and S-100β improved in hydrogel group. Response to mechanical stimulus, myelin density, axonal regeneration, and myelin sheath reconstruction improved in the mix group. The gastrocnemius muscle index was significantly reduced in the experimental groups while collagen fibers increased in these groups. These findings suggest that injection of hydrogel derived from decellularized amniotic membrane into the epineurium can be promoted reconstruction of peripheral nerve injury and improved functional nerve recovery. Also, metformin administration can reinforce the therapeutic effect of the hydrogel.
Collapse
Affiliation(s)
- Zahra Rabiei Dolatabadi
- Department of Anatomical Sciences, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mona Saheli
- Department of Anatomical Sciences, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Somayeh Solhjoo
- Department of Anatomical Sciences, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehran Hosseini
- Department of Anatomical Sciences, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Elahe Mousanejad
- Department of Anatomical Sciences, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mandana Jafari
- Pharmaceutics Research Center, Institute of Neuropharmacology, Afzalipour Faculty of Medicine, Kerman University Medical Sciences, Kerman, Iran
| | - Vahid Sheibani
- Kerman Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | | |
Collapse
|
8
|
Chen Z, Wang J, Kankala RK, Jiang M, Long L, Li W, Zou L, Chen A, Liu Y. Decellularized extracellular matrix-based disease models for drug screening. Mater Today Bio 2024; 29:101280. [PMID: 39399243 PMCID: PMC11470555 DOI: 10.1016/j.mtbio.2024.101280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/12/2024] [Accepted: 09/26/2024] [Indexed: 10/15/2024] Open
Abstract
In vitro drug screening endeavors to replicate cellular states closely resembling those encountered in vivo, thereby maximizing the fidelity of drug effects and responses within the body. Decellularized extracellular matrix (dECM)-based materials offer a more authentic milieu for crafting disease models, faithfully emulating the extracellular components and structural complexities encountered by cells in vivo. This review discusses recent advancements in leveraging dECM-based materials as biomaterials for crafting cell models tailored for drug screening. Initially, we delineate the biological functionalities of diverse ECM components, shedding light on their potential influences on disease model construction. Further, we elucidate the decellularization techniques and methodologies for fabricating cell models utilizing dECM substrates. Then, the article delves into the research strides made in employing dECM-based models for drug screening across a spectrum of ailments, including tumors, as well as heart, liver, lung, and bone diseases. Finally, the review summarizes the bottlenecks, hurdles, and promising research trajectories associated with the dECM materials for drug screening, alongside their prospective applications in personalized medicine. Together, by encapsulating the contemporary research landscape surrounding dECM materials in cell model construction and drug screening, this review underscores the vast potential of dECM materials in drug assessment and personalized therapy.
Collapse
Affiliation(s)
- Zhoujiang Chen
- Institute for Advanced Study, Chengdu University, Chengdu, 610106, Sichuan, PR China
| | - Ji Wang
- Affiliated Hospital & Clinical Medical College of Chengdu University, Chengdu University, Chengdu, 610106, Sichuan, PR China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, PR China
| | - Mingli Jiang
- School of Pharmacy, Zunyi Medical University, Zunyi, 563099, Guizhou, PR China
| | - Lianlin Long
- School of Pharmacy, Zunyi Medical University, Zunyi, 563099, Guizhou, PR China
| | - Wei Li
- Affiliated Hospital & Clinical Medical College of Chengdu University, Chengdu University, Chengdu, 610106, Sichuan, PR China
| | - Liang Zou
- Institute for Advanced Study, Chengdu University, Chengdu, 610106, Sichuan, PR China
| | - Aizheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, PR China
| | - Ya Liu
- Affiliated Hospital & Clinical Medical College of Chengdu University, Chengdu University, Chengdu, 610106, Sichuan, PR China
| |
Collapse
|
9
|
Chavez-Granados PA, Garcia-Contreras R, Reyes-Lopez CAS, Correa-Basurto J, Hernandez-Rojas IE, Hernandez-Gomez G, Jurado CA, Alhotan A. Green Synthesis of Silver Nanoparticles with Roasted Green Tea: Applications in Alginate-Gelatin Hydrogels for Bone Regeneration. Gels 2024; 10:706. [PMID: 39590062 PMCID: PMC11594056 DOI: 10.3390/gels10110706] [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: 09/30/2024] [Revised: 10/19/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024] Open
Abstract
The incorporation of silver nanoparticles (AgNPs) into alginate-gelatin (Alg-Gel) hydrogels can enhance the properties of these materials for bone regeneration applications, due to the antimicrobial properties of AgNPs and non-cytotoxic concentrations, osteoinductive properties, and regulation of stem cell proliferation and differentiation. Here, the hydrogel formulation included 2% (w/v) sodium alginate, 4 µg/mL AgNPs, and 2.5% (w/v) gelatin. AgNPs were synthesized using a 2% (w/v) aqueous extract of roasted green tea with silver nitrate. The aqueous extract of roasted green tea for AgNP synthesis was characterized using HPLC and UHPLC-ESI-QTOF-MS/MS, and antioxidant capacity was measured in Trolox equivalents (TE) from 4 to 20 nmol/well concentrations. Stem cells from human exfoliated deciduous tooth cells were used for differentiation assays including positive (SHEDs/hydrogel with AgNPs) and negative controls (hydrogel without AgNPs). FTIR was used for hydrogel chemical characterization. Statistical analysis (p < 0.05, ANOVA) confirmed significant findings. Roasted green tea extract contained caffeine (most abundant), (-)-Gallocatechin, gallic acid, and various catechins. XRD analysis revealed FCC structure, TEM showed quasispheroidal AgNPs (19.85 ± 3 nm), and UV-Vis indicated a plasmon surface of 418 nm. This integration of nanotechnology and biomaterials shows promise for addressing bone tissue loss in clinical and surgical settings.
Collapse
Affiliation(s)
- Patricia Alejandra Chavez-Granados
- Interdisciplinary Research Laboratory, Nanostructures, and Biomaterials Area, National School of Higher Studies (ENES) Leon, National Autonomous University of Mexico (UNAM), Leon 37684, Mexico
- Dental Science, Master’s and Doctoral Program in Medical, Dental, and Health Sciences, National Autonomous University of Mexico (UNAM), Coyoacán, Mexico City 04510, Mexico
| | - Rene Garcia-Contreras
- Interdisciplinary Research Laboratory, Nanostructures, and Biomaterials Area, National School of Higher Studies (ENES) Leon, National Autonomous University of Mexico (UNAM), Leon 37684, Mexico
| | - Cesar A. S. Reyes-Lopez
- National Polytechnic Institute, National School of Medicine and Homeopathy, Mexico City 07320, Mexico
| | - Jose Correa-Basurto
- Laboratory of Design and Development of New Drugs and Biotechnological Innovation, SEPI-Escuela Superior de Medicina, National Polytechnic Institute, Plan de San Luis and Díaz Mirón, Mexico City 11340, Mexico
| | - Irving E. Hernandez-Rojas
- Periodontics and Implantology Area, National School of Higher Studies (ENES) Leon, National Autonomous University of Mexico (UNAM), Leon 37684, Mexico (G.H.-G.)
| | - Gabriela Hernandez-Gomez
- Periodontics and Implantology Area, National School of Higher Studies (ENES) Leon, National Autonomous University of Mexico (UNAM), Leon 37684, Mexico (G.H.-G.)
| | - Carlos Alberto Jurado
- Operative Dentistry Division, Department of General Dentistry, College of Dentistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Abdulaziz Alhotan
- Department of Dental Health, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 12372, Saudi Arabia
| |
Collapse
|
10
|
Li C, An N, Song Q, Hu Y, Yin W, Wang Q, Le Y, Pan W, Yan X, Wang Y, Liu J. Enhancing organoid culture: harnessing the potential of decellularized extracellular matrix hydrogels for mimicking microenvironments. J Biomed Sci 2024; 31:96. [PMID: 39334251 PMCID: PMC11429032 DOI: 10.1186/s12929-024-01086-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Over the past decade, organoids have emerged as a prevalent and promising research tool, mirroring the physiological architecture of the human body. However, as the field advances, the traditional use of animal or tumor-derived extracellular matrix (ECM) as scaffolds has become increasingly inadequate. This shift has led to a focus on developing synthetic scaffolds, particularly hydrogels, that more accurately mimic three-dimensional (3D) tissue structures and dynamics in vitro. The ECM-cell interaction is crucial for organoid growth, necessitating hydrogels that meet organoid-specific requirements through modifiable physical and compositional properties. Advanced composite hydrogels have been engineered to more effectively replicate in vivo conditions, offering a more accurate representation of human organs compared to traditional matrices. This review explores the evolution and current uses of decellularized ECM scaffolds, emphasizing the application of decellularized ECM hydrogels in organoid culture. It also explores the fabrication of composite hydrogels and the prospects for their future use in organoid systems.
Collapse
Affiliation(s)
- Chen Li
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China
| | - Ni An
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Clinical Translational Science Center, Tsinghua University, Beijing, 102218, China
| | - Qingru Song
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Clinical Translational Science Center, Tsinghua University, Beijing, 102218, China
| | - Yuelei Hu
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China
| | - Wenzhen Yin
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Clinical Translational Science Center, Tsinghua University, Beijing, 102218, China
| | - Qi Wang
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China
| | - Yinpeng Le
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China
- School of Materials Science and Engineering, Institute of Smart Biomedical Materials, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Wenting Pan
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China
| | - Xinlong Yan
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China.
| | - Yunfang Wang
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China.
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Clinical Translational Science Center, Tsinghua University, Beijing, 102218, China.
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China.
| | - Juan Liu
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China.
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
11
|
Berger C, Glaser M, Ziegler AL, Neukel V, Walz F, Zdzieblo D. Generation of a pancreas derived hydrogel for the culture of hiPSC derived pancreatic endocrine cells. Sci Rep 2024; 14:20653. [PMID: 39232042 PMCID: PMC11375036 DOI: 10.1038/s41598-024-67327-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 07/10/2024] [Indexed: 09/06/2024] Open
Abstract
Stem cell-derived β-cells (SC-BCs) represent a potential source for curing diabetes. To date, in vitro generated SC-BCs display an immature phenotype and lack important features in comparison to their bona-fide counterparts. Transplantation into a living animal promotes SC-BCs maturation, indicating that components of the in vivo microenvironment trigger final SC-BCs development. Here, we investigated whether cues of the pancreas specific extracellular matrix (ECM) can improve the differentiation of human induced pluripotent stem cells (hiPSCs) towards β-cells in vitro. To this aim, a pancreas specific ECM (PanMa) hydrogel was generated from decellularized porcine pancreas and its effect on the differentiation of hiPSC-derived pancreatic hormone expressing cells (HECs) was tested. The hydrogel solidified upon neutralization at 37 °C with gelation kinetics similar to Matrigel. Cytocompatibility of the PanMa hydrogel was demonstrated for a culture duration of 21 days. Encapsulation and culture of HECs in the PanMa hydrogel over 7 days resulted in a stable gene and protein expression of most β-cell markers, but did not improve β-cell identity. In conclusion, the study describes the production of a PanMa hydrogel, which provides the basis for the development of ECM hydrogels that are more adapted to the demands of SC-BCs.
Collapse
Affiliation(s)
- Constantin Berger
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany.
| | - Markus Glaser
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Anna-Lena Ziegler
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Valentina Neukel
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Fabiola Walz
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Daniela Zdzieblo
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
- Project Center for Stem Cell Process Engineering, Fraunhofer Institute for Silicate Research, Würzburg, Germany
- Translational Center Regenerative Therapies, Fraunhofer Institute for Silicate Research, Würzburg, Germany
| |
Collapse
|
12
|
Milton LA, Davern JW, Hipwood L, Chaves JCS, McGovern J, Broszczak D, Hutmacher DW, Meinert C, Toh YC. Liver click dECM hydrogels for engineering hepatic microenvironments. Acta Biomater 2024; 185:144-160. [PMID: 38960110 DOI: 10.1016/j.actbio.2024.06.037] [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: 01/22/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Decellularized extracellular matrix (dECM) hydrogels provide tissue-specific microenvironments which accommodate physiological cellular phenotypes in 3D in vitro cell cultures. However, their formation hinges on collagen fibrillogenesis, a complex process which limits regulation of physicochemical properties. Hence, achieving reproducible results with dECM hydrogels poses as a challenge. Here, we demonstrate that thiolation of solubilized liver dECM enables rapid formation of covalently crosslinked hydrogels via Michael-type addition, allowing for precise control over mechanical properties and superior organotypic biological activity. Investigation of various decellularization methodologies revealed that treatment of liver tissue with Triton X-100 and ammonium hydroxide resulted in near complete DNA removal with significant retention of the native liver proteome. Chemical functionalization of pepsin-solubilized liver dECMs via 1-ethyl-3(3-dimethylamino)propyl carbodiimide (EDC)/N-hydroxysuccinimide (NHS) coupling of l-Cysteine created thiolated liver dECM (dECM-SH), which rapidly reacted with 4-arm polyethylene glycol (PEG)-maleimide to form optically clear hydrogels under controlled conditions. Importantly, Young's moduli could be precisely tuned between 1 - 7 kPa by varying polymer concentrations, enabling close replication of healthy and fibrotic liver conditions in in vitro cell cultures. Click dECM-SH hydrogels were cytocompatible, supported growth of HepG2 and HepaRG liver cells, and promoted liver-specific functional phenotypes as evidenced by increased metabolic activity, as well CYP1A2 and CYP3A4 activity and excretory function when compared to monolayer culture and collagen-based hydrogels. Our findings demonstrate that click-functionalized dECM hydrogels offer a highly controlled, reproducible alternative to conventional tissue-derived hydrogels for in vitro cell culture applications. STATEMENT OF SIGNIFICANCE: Traditional dECM hydrogels face challenges in reproducibility and mechanical property control due to variable crosslinking processes. We introduce a click hydrogel based on porcine liver decellularized extracellular matrix (dECM) that circumnavigates these challenges. After optimizing liver decellularization for ECM retention, we integrated thiol-functionalized liver dECM with polyethylene-glycol derivatives through Michael-type addition click chemistry, enabling rapid, room-temperature gelation. This offers enhanced control over the hydrogel's mechanical and biochemical properties. The resultant click dECM hydrogels mimic the liver's natural ECM and exhibit greater mechanical tunability and handling ease, facilitating their application in high-throughput and industrial settings. Moreover, these hydrogels significantly improve the function of HepaRG-derived hepatocytes in 3D culture, presenting an advancement for liver tissue cell culture models for drug testing applications.
Collapse
Affiliation(s)
- Laura A Milton
- Faculty of Engineering, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia; Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia; Gelomics Pty Ltd, Brisbane, Australia
| | - Jordan W Davern
- Faculty of Engineering, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia; Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia; Gelomics Pty Ltd, Brisbane, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology, Brisbane, Australia
| | - Luke Hipwood
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia; Gelomics Pty Ltd, Brisbane, Australia; Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Juliana C S Chaves
- Cell & Molecular Biology Department, Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Jacqui McGovern
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology, Brisbane, Australia; Max Planck Queensland Centre (MPQC) for the Materials Science of Extracellular Matrices, Queensland University of Technology, Brisbane, Australia
| | - Daniel Broszczak
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Dietmar W Hutmacher
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology, Brisbane, Australia; Max Planck Queensland Centre (MPQC) for the Materials Science of Extracellular Matrices, Queensland University of Technology, Brisbane, Australia; Australian Research Council (ARC) Training Centre for Multiscale 3D Imaging, Modelling and Manufacturing (M3D Innovation), Queensland University of Technology, Brisbane, Australia
| | - Christoph Meinert
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia; Gelomics Pty Ltd, Brisbane, Australia.
| | - Yi-Chin Toh
- Faculty of Engineering, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia; Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology, Brisbane, Australia; Max Planck Queensland Centre (MPQC) for the Materials Science of Extracellular Matrices, Queensland University of Technology, Brisbane, Australia; Centre for Microbiome Research, Queensland University of Technology, Brisbane, Australia.
| |
Collapse
|
13
|
Shi W, Zhang Z, Wang X. The Prospect of Hepatic Decellularized Extracellular Matrix as a Bioink for Liver 3D Bioprinting. Biomolecules 2024; 14:1019. [PMID: 39199406 PMCID: PMC11352484 DOI: 10.3390/biom14081019] [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: 05/27/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 09/01/2024] Open
Abstract
The incidence of liver diseases is high worldwide. Many factors can cause liver fibrosis, which in turn can lead to liver cirrhosis and even liver cancer. Due to the shortage of donor organs, immunosuppression, and other factors, only a few patients are able to undergo liver transplantation. Therefore, how to construct a bioartificial liver that can be transplanted has become a global research hotspot. With the rapid development of three-dimensional (3D) bioprinting in the field of tissue engineering and regenerative medicine, researchers have tried to use various 3D bioprinting technologies to construct bioartificial livers in vitro. In terms of the choice of bioinks, liver decellularized extracellular matrix (dECM) has many advantages over other materials for cell-laden hydrogel in 3D bioprinting. This review mainly summarizes the acquisition of liver dECM and its application in liver 3D bioprinting as a bioink with respect to availability, printability, and biocompatibility in many aspects and puts forward the current challenges and prospects.
Collapse
Affiliation(s)
- Wen Shi
- Center of 3D Printing & Organ Manufacturing, School of Intelligent Medicine, China Medical University, Shenyang 110122, China;
- Department of Ultrasound, The First Hospital of China Medical University, Shenyang 110001, China
| | - Zhe Zhang
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang 110001, China;
| | - Xiaohong Wang
- Center of 3D Printing & Organ Manufacturing, School of Intelligent Medicine, China Medical University, Shenyang 110122, China;
| |
Collapse
|
14
|
Li Y, Zhang Y, Zhang G. Comparative Analysis of Decellularization Methods for the Production of Decellularized Umbilical Cord Matrix. Curr Issues Mol Biol 2024; 46:7686-7701. [PMID: 39057096 PMCID: PMC11276046 DOI: 10.3390/cimb46070455] [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: 06/13/2024] [Revised: 07/10/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
The importance of decellularized extracellular matrix (dECM) as a natural biomaterial in tissue engineering and regenerative medicine is rapidly growing. The core objective of the decellularization process is to eliminate cellular components while maximizing the preservation of the ECM's primary structure and components. Establishing a rapid, effective, and minimally destructive decellularization technique is essential for obtaining high-quality dECM to construct regenerative organs. This study focused on human umbilical cord tissue, designing different reagent combinations for decellularization protocols while maintaining a consistent processing time. The impact of these protocols on the decellularization efficiency of human umbilical cord tissue was evaluated. The results suggested that the composite decellularization strategy utilizing trypsin/EDTA + Triton X-100 + sodium deoxycholate was the optimal approach in this study for preparing decellularized human umbilical cord dECM. After 5 h of decellularization treatment, most cellular components were eliminated, confirmed through dsDNA quantitative detection, hematoxylin and eosin (HE) staining, and DAPI staining. Meanwhile, Masson staining, periodic acid-silver methenamine (PASM) staining, periodic acid-Schiff (PAS) staining, and immunofluorescent tissue section staining results revealed that the decellularized scaffold retained extracellular matrix components, including collagen and glycosaminoglycans (GAGs). Compared to native umbilical cord tissue, electron microscopy results demonstrated that the microstructure of the extracellular matrix was well preserved after decellularization. Furthermore, Fourier-transform infrared spectroscopy (FTIR) findings indicated that the decellularization process successfully retained the main functional group structures of extracellular matrix (ECM) components. The quantitative analysis of collagen, elastin, and GAG content validated the advantages of this decellularization process in preserving and purifying ECM components. Additionally, it was confirmed that this decellularized matrix exhibited no cytotoxicity in vitro. This study achieved short-term decellularization preparation for umbilical cord tissue through a combined decellularization strategy.
Collapse
Affiliation(s)
- Yang Li
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; (Y.L.); (Y.Z.)
- School of Chemical and Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Zhang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; (Y.L.); (Y.Z.)
| | - Guifeng Zhang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; (Y.L.); (Y.Z.)
- School of Chemical and Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
15
|
Roudaut M, Caillaud A, Souguir Z, Bray L, Girardeau A, Rimbert A, Croyal M, Lambert G, Patitucci M, Delpouve G, Vandenhaute É, Le May C, Maubon N, Cariou B, Si‐Tayeb K. Human induced pluripotent stem cells-derived liver organoids grown on a Biomimesys® hyaluronic acid-based hydroscaffold as a new model for studying human lipoprotein metabolism. Bioeng Transl Med 2024; 9:e10659. [PMID: 39036087 PMCID: PMC11256179 DOI: 10.1002/btm2.10659] [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: 07/27/2023] [Revised: 01/29/2024] [Accepted: 02/12/2024] [Indexed: 07/23/2024] Open
Abstract
The liver plays a key role in the metabolism of lipoproteins, controlling both production and catabolism. To accelerate the development of new lipid-lowering therapies in humans, it is essential to have a relevant in vitro study model available. The current hepatocyte-like cells (HLCs) models derived from hiPSC can be used to model many genetically driven diseases but require further improvement to better recapitulate the complexity of liver functions. Here, we aimed to improve the maturation of HLCs using a three-dimensional (3D) approach using Biomimesys®, a hyaluronic acid-based hydroscaffold in which hiPSCs may directly form aggregates and differentiate toward a functional liver organoid model. After a 28-day differentiation 3D protocol, we showed that many hepatic genes were upregulated in the 3D model (liver organoids) in comparison with the 2D model (HLCs). Liver organoids, grown on Biomimesys®, exhibited an autonomous cell organization, were composed of different cell types and displayed enhanced cytochromes P450 activities compared to HLCs. Regarding the functional capacities of these organoids, we showed that they were able to accumulate lipids (hepatic steatosis), internalize low-density lipoprotein and secrete apolipoprotein B. Interestingly, we showed for the first time that this model was also able to produce apolipoprotein (a), the apolipoprotein (a) specific of Lp(a). This innovative hiPSC-derived liver organoid model may serve as a relevant model for studying human lipopoprotein metabolism, including Lp(a).
Collapse
Affiliation(s)
- Meryl Roudaut
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thoraxNantesFrance
- HCS PharmaLilleFrance
| | - Amandine Caillaud
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thoraxNantesFrance
| | | | - Lise Bray
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thoraxNantesFrance
| | - Aurore Girardeau
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thoraxNantesFrance
| | - Antoine Rimbert
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thoraxNantesFrance
| | - Mikaël Croyal
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thoraxNantesFrance
- CRNH‐Ouest Mass Spectrometry Core FacilityNantesFrance
| | - Gilles Lambert
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI)Université de La RéunionSaint‐Denisde La RéunionFrance
| | - Murielle Patitucci
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thoraxNantesFrance
| | | | | | - Cédric Le May
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thoraxNantesFrance
| | | | - Bertrand Cariou
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thoraxNantesFrance
| | - Karim Si‐Tayeb
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thoraxNantesFrance
| |
Collapse
|
16
|
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
|
17
|
Wu L, Vllasaliu D, Cui Q, Raimi-Abraham BT. In Situ Self-Assembling Liver Spheroids with Synthetic Nanoscaffolds for Preclinical Drug Screening Applications. ACS APPLIED MATERIALS & INTERFACES 2024; 16:25610-25621. [PMID: 38741479 PMCID: PMC11129140 DOI: 10.1021/acsami.3c17384] [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: 11/20/2023] [Revised: 04/16/2024] [Accepted: 05/01/2024] [Indexed: 05/16/2024]
Abstract
Drug-induced liver injury (DILI) is one of the most common reasons for acute liver failure and a major reason for the withdrawal of medications from the market. There is a growing need for advanced in vitro liver models that can effectively recapitulate hepatic function, offering a robust platform for preclinical drug screening applications. Here, we explore the potential of self-assembling liver spheroids in the presence of electrospun and cryomilled poly(caprolactone) (PCL) nanoscaffolds for use as a new preclinical drug screening tool. This study investigated the extent to which nanoscaffold concentration may have on spheroid size and viability and liver-specific biofunctionality. The efficacy of our model was further validated using a comprehensive dose-dependent acetaminophen toxicity protocol. Our findings show the strong potential of PCL-based nanoscaffolds to facilitate in situ self-assembly of liver spheroids with sizes under 350 μm. The presence of the PCL-based nanoscaffolds (0.005 and 0.01% w/v) improved spheroid viability and the secretion of critical liver-specific biomarkers, namely, albumin and urea. Liver spheroids with nanoscaffolds showed improved drug-metabolizing enzyme activity and greater sensitivity to acetaminophen compared to two-dimensional monolayer cultures and scaffold-free liver spheroids. These promising findings highlight the potential of our nanoscaffold-based liver spheroids as an in vitro liver model for drug-induced hepatotoxicity and drug screening.
Collapse
Affiliation(s)
- Lina Wu
- King’s College London,
Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical
Sciences, Institute of Pharmaceutical Science, Franklin-Wilkins Building, 150 Stamford
Street, London SE1 9NH, U.K.
| | - Driton Vllasaliu
- King’s College London,
Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical
Sciences, Institute of Pharmaceutical Science, Franklin-Wilkins Building, 150 Stamford
Street, London SE1 9NH, U.K.
| | - Qi Cui
- King’s College London,
Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical
Sciences, Institute of Pharmaceutical Science, Franklin-Wilkins Building, 150 Stamford
Street, London SE1 9NH, U.K.
| | - Bahijja Tolulope Raimi-Abraham
- King’s College London,
Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical
Sciences, Institute of Pharmaceutical Science, Franklin-Wilkins Building, 150 Stamford
Street, London SE1 9NH, U.K.
| |
Collapse
|
18
|
Song H, Jiang H, Hu W, Hai Y, Cai Y, Li H, Liao Y, Huang Y, Lv X, Zhang Y, Zhang J, Huang Y, Liang X, Huang H, Lin X, Wang Y, Yi X. Cervical extracellular matrix hydrogel optimizes tumor heterogeneity of cervical squamous cell carcinoma organoids. SCIENCE ADVANCES 2024; 10:eadl3511. [PMID: 38748808 PMCID: PMC11095500 DOI: 10.1126/sciadv.adl3511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 04/10/2024] [Indexed: 05/19/2024]
Abstract
Cervical cancer, primarily squamous cell carcinoma, is the most prevalent gynecologic malignancy. Organoids can mimic tumor development in vitro, but current Matrigel inaccurately replicates the tissue-specific microenvironment. This limitation compromises the accurate representation of tumor heterogeneity. We collected para-cancerous cervical tissues from patients diagnosed with cervical squamous cell carcinoma (CSCC) and prepared uterine cervix extracellular matrix (UCEM) hydrogels. Proteomic analysis of UCEM identified several tissue-specific signaling pathways including human papillomavirus, phosphatidylinositol 3-kinase-AKT, and extracellular matrix receptor. Secreted proteins like FLNA, MYH9, HSPA8, and EEF1A1 were present, indicating UCEM successfully maintained cervical proteins. UCEM provided a tailored microenvironment for CSCC organoids, enabling formation and growth while preserving tumorigenic potential. RNA sequencing showed UCEM-organoids exhibited greater similarity to native CSCC and reflected tumor heterogeneity by exhibiting CSCC-associated signaling pathways including virus protein-cytokine, nuclear factor κB, tumor necrosis factor, and oncogenes EGR1, FPR1, and IFI6. Moreover, UCEM-organoids developed chemotherapy resistance. Our research provides insights into advanced organoid technology through native matrix hydrogels.
Collapse
Affiliation(s)
- Haonan Song
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Haoyuan Jiang
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Weichu Hu
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yan Hai
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yihuan Cai
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Hu Li
- The First Affiliated Hospital, Jinan University, Guangzhou 510280, China
| | - Yuru Liao
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yi Huang
- Department of Gynecology, The Sixth Affiliated Hospital, South China University of Technology, Foshan 528200, China
| | - Xiaogang Lv
- Department of Gynecologic Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 510030, China
| | - Yefei Zhang
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jiping Zhang
- Department of Gynecology, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China
| | - Yan Huang
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xiaomei Liang
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Hao Huang
- Department of Gynecology, The Sixth Affiliated Hospital, South China University of Technology, Foshan 528200, China
| | - Xinhua Lin
- Greater Bay Area Institute of Precision Medicine, Guangzhou 510280, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University Shanghai, Shanghai 200438, China
| | - Yifeng Wang
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xiao Yi
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Greater Bay Area Institute of Precision Medicine, Guangzhou 510280, China
| |
Collapse
|
19
|
Wu W, Liu Y, Liu R, Wang Y, Zhao Y, Li H, Lu B, Ju C, Gao X, Xu H, Cao Y, Cheng S, Wang Z, Jia S, Hu C, Zhu L, Hao D. Decellularized Brain Extracellular Matrix Hydrogel Aids the Formation of Human Spinal-Cord Organoids Recapitulating the Complex Three-Dimensional Organization. ACS Biomater Sci Eng 2024; 10:3203-3217. [PMID: 38557027 DOI: 10.1021/acsbiomaterials.4c00029] [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: 04/04/2024]
Abstract
The intricate electrophysiological functions and anatomical structures of spinal cord tissue render the establishment of in vitro models for spinal cord-related diseases highly challenging. Currently, both in vivo and in vitro models for spinal cord-related diseases are still underdeveloped, complicating the exploration and development of effective therapeutic drugs or strategies. Organoids cultured from human induced pluripotent stem cells (hiPSCs) hold promise as suitable in vitro models for spinal cord-related diseases. However, the cultivation of spinal cord organoids predominantly relies on Matrigel, a matrix derived from murine sarcoma tissue. Tissue-specific extracellular matrices are key drivers of complex organ development, thus underscoring the urgent need to research safer and more physiologically relevant organoid culture materials. Herein, we have prepared a rat decellularized brain extracellular matrix hydrogel (DBECMH), which supports the formation of hiPSC-derived spinal cord organoids. Compared with Matrigel, organoids cultured in DBECMH exhibited higher expression levels of markers from multiple compartments of the natural spinal cord, facilitating the development and maturation of spinal cord organoid tissues. Our study suggests that DBECMH holds potential to replace Matrigel as the standard culture medium for human spinal cord organoids, thereby advancing the development of spinal cord organoid culture protocols and their application in in vitro modeling of spinal cord-related diseases.
Collapse
Affiliation(s)
- Weidong Wu
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
| | - Youjun Liu
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
| | - Renfeng Liu
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
| | - Yuhao Wang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
| | - Yuqi Zhao
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
| | - Hui Li
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
| | - Botao Lu
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
| | - Cheng Ju
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
| | - Xinlin Gao
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
| | - Hailiang Xu
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
| | - Yulin Cao
- Healthina Academy of Cellular Intelligence Manufacturing & Neurotrauma Repair of Tianjin Economic-Technological Development Area, No. 220 DongTing Road, TEDA District, Tianjin 300457, China
- TANGYI Biomedicine (Tianjin) Co., Ltd. (TBMed), No. 286 Anshan West Road, Nankai District, Tianjin 300190, China
| | - Shixiang Cheng
- Healthina Academy of Cellular Intelligence Manufacturing & Neurotrauma Repair of Tianjin Economic-Technological Development Area, No. 220 DongTing Road, TEDA District, Tianjin 300457, China
- TANGYI Biomedicine (Tianjin) Co., Ltd. (TBMed), No. 286 Anshan West Road, Nankai District, Tianjin 300190, China
| | - Zhiyuan Wang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
| | - Shuaijun Jia
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
| | - Chunping Hu
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
| | - Lei Zhu
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
| | - Dingjun Hao
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No. 555, Beilin District, Xi'an, Shaanxi, China 710001
| |
Collapse
|
20
|
Asadollahi N, Hajari MA, Alipour Choshali M, Ajoudanian M, Ziai SA, Vosough M, Piryaei A. Bioengineering scalable and drug-responsive in vitro human multicellular non-alcoholic fatty liver disease microtissues encapsulated in the liver extracellular matrix-derived hydrogel. EXCLI JOURNAL 2024; 23:421-440. [PMID: 38741724 PMCID: PMC11089098 DOI: 10.17179/excli2023-6878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/06/2024] [Indexed: 05/16/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a high-prevalence and progressive disorder. Due to lack of reliable in vitro models to recapitulate the consecutive phases, the exact pathogenesis mechanism of this disease and approved therapeutic medications have not been revealed yet. It has been proven that the interplay between multiple hepatic cell types and liver extracellular matrix (ECM) are critical in NAFLD initiation and progression. Herein, a liver microtissue (LMT) consisting of Huh-7, THP-1, and LX-2 cell lines and human umbilical vein endothelial cells (HUVEC), which could be substituted for the main hepatic cells (hepatocyte, Kupffer, stellate, and sinusoidal endothelium, respectively), encapsulated in liver derived ECM-Alginate composite, was bioengineered. When the microtissues were treated with free fatty acids (FFAs) including Oleic acid (6.6×10-4M) and Palmitic acid (3.3×10-4M), they displayed the key features of NAFLD, including similar pattern of transcripts for genes involved in lipid metabolism, inflammation, insulin-resistance, and fibrosis, as well as pro-inflammatory and pro-fibrotic cytokines' secretions and intracellular lipid accumulation. Continuing FFAs supplementation, we demonstrated that the NAFLD phenomenon was established on day 3 and progressed to the initial fibrosis stage by day 8. Furthermore, this model was stable until day 12 post FFAs withdrawal on day 3. Moreover, administration of an anti-steatotic drug candidate, Liraglutide (15 μM), on the NAFLD microtissues significantly ameliorated the NAFLD phenomenon. Overall, we bioengineered a drug-responsive, cost-benefit liver microtissues which can simulate the initiation and progression of NAFLD. It is expected that this platform could potentially be used for studying molecular pathogenesis of NAFLD and high-throughput drug screening. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Negar Asadollahi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Mohammad Amin Hajari
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahmoud Alipour Choshali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Ajoudanian
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Ziai
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Huddinge, Sweden
| | - Abbas Piryaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Smandri A, Al-Masawa ME, Hwei NM, Fauzi MB. ECM-derived biomaterials for regulating tissue multicellularity and maturation. iScience 2024; 27:109141. [PMID: 38405613 PMCID: PMC10884934 DOI: 10.1016/j.isci.2024.109141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2024] Open
Abstract
Recent breakthroughs in developing human-relevant organotypic models led to the building of highly resemblant tissue constructs that hold immense potential for transplantation, drug screening, and disease modeling. Despite the progress in fine-tuning stem cell multilineage differentiation in highly controlled spatiotemporal conditions and hosting microenvironments, 3D models still experience naive and incomplete morphogenesis. In particular, existing systems and induction protocols fail to maintain stem cell long-term potency, induce high tissue-level multicellularity, or drive the maturity of stem cell-derived 3D models to levels seen in their in vivo counterparts. In this review, we highlight the use of extracellular matrix (ECM)-derived biomaterials in providing stem cell niche-mimicking microenvironment capable of preserving stem cell long-term potency and inducing spatial and region-specific differentiation. We also examine the maturation of different 3D models, including organoids, encapsulated in ECM biomaterials and provide looking-forward perspectives on employing ECM biomaterials in building more innovative, transplantable, and functional organs.
Collapse
Affiliation(s)
- Ali Smandri
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Maimonah Eissa Al-Masawa
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Ng Min Hwei
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Mh Busra Fauzi
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
22
|
Carpentier N, Ye S, Delemarre MD, Van der Meeren L, Skirtach AG, van der Laan LJW, Schneeberger K, Spee B, Dubruel P, Van Vlierberghe S. Gelatin-Based Hybrid Hydrogels as Matrices for Organoid Culture. Biomacromolecules 2024; 25:590-604. [PMID: 38174962 DOI: 10.1021/acs.biomac.2c01496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
The application of liver organoids is very promising in the field of liver tissue engineering; however, it is still facing some limitations. One of the current major limitations is the matrix in which they are cultured. The mainly undefined and murine-originated tumor matrices derived from Engelbreth-Holm-Swarm (EHS) sarcoma, such as Matrigel, are still the standard culturing matrices for expansion and differentiation of organoids toward hepatocyte-like cells, which will obstruct its future clinical application potential. In this study, we exploited the use of newly developed highly defined hydrogels as potential matrices for the culture of liver organoids and compared them to Matrigel and two hydrogels that were already researched in the field of organoid research [i.e., polyisocyanopeptides, enriched with laminin-entactin complex (PIC-LEC) and gelatin methacryloyl (GelMA)]. The newly developed hydrogels are materials that have a physicochemical resemblance with native liver tissue. Norbornene-modified dextran cross-linked with thiolated gelatin (DexNB-GelSH) has a swelling ratio and macro- and microscale properties that highly mimic liver tissue. Norbornene-modified chondroitin sulfate cross-linked with thiolated gelatin (CSNB-GelSH) contains chondroitin sulfate, which is a glycosaminoglycan (GAG) that is present in the liver ECM. Furthermore, CSNB-GelSH hydrogels with different mechanical properties were evaluated. Bipotent intrahepatic cholangiocyte organoids (ICOs) were applied in this work and encapsulated in these materials. This research revealed that the newly developed materials outperformed Matrigel, PIC-LEC, and GelMA in the differentiation of ICOs toward hepatocyte-like cells. Furthermore, some trends indicate that an interplay of both the chemical composition and the mechanical properties has an influence on the relative expression of certain hepatocyte markers. Both DexNB-GelSH and CSNB-GelSH showed promising results for the expansion and differentiation of intrahepatic cholangiocyte organoids. The stiffest CSNB-GelSH hydrogel even significantly outperformed Matrigel based on ALB, BSEP, and CYP3A4 gene expression, being three important hepatocyte markers.
Collapse
Affiliation(s)
- Nathan Carpentier
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent 9000, Belgium
| | - Shicheng Ye
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CT, The Netherlands
| | - Maarten D Delemarre
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CT, The Netherlands
| | - Louis Van der Meeren
- Nano-Biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent 9000, Belgium
| | - André G Skirtach
- Nano-Biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent 9000, Belgium
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam 3000 CA, The Netherlands
| | - Kerstin Schneeberger
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CT, The Netherlands
| | - Bart Spee
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CT, The Netherlands
| | - Peter Dubruel
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent 9000, Belgium
| | - Sandra Van Vlierberghe
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent 9000, Belgium
| |
Collapse
|
23
|
Ansari M, Darvishi A, Sabzevari A. A review of advanced hydrogels for cartilage tissue engineering. Front Bioeng Biotechnol 2024; 12:1340893. [PMID: 38390359 PMCID: PMC10881834 DOI: 10.3389/fbioe.2024.1340893] [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: 11/19/2023] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
With the increase in weight and age of the population, the consumption of tobacco, inappropriate foods, and the reduction of sports activities in recent years, bone and joint diseases such as osteoarthritis (OA) have become more common in the world. From the past until now, various treatment strategies (e.g., microfracture treatment, Autologous Chondrocyte Implantation (ACI), and Mosaicplasty) have been investigated and studied for the prevention and treatment of this disease. However, these methods face problems such as being invasive, not fully repairing the tissue, and damaging the surrounding tissues. Tissue engineering, including cartilage tissue engineering, is one of the minimally invasive, innovative, and effective methods for the treatment and regeneration of damaged cartilage, which has attracted the attention of scientists in the fields of medicine and biomaterials engineering in the past several years. Hydrogels of different types with diverse properties have become desirable candidates for engineering and treating cartilage tissue. They can cover most of the shortcomings of other treatment methods and cause the least secondary damage to the patient. Besides using hydrogels as an ideal strategy, new drug delivery and treatment methods, such as targeted drug delivery and treatment through mechanical signaling, have been studied as interesting strategies. In this study, we review and discuss various types of hydrogels, biomaterials used for hydrogel manufacturing, cartilage-targeting drug delivery, and mechanosignaling as modern strategies for cartilage treatment.
Collapse
Affiliation(s)
- Mojtaba Ansari
- Department of Biomedical Engineering, Meybod University, Meybod, Iran
| | - Ahmad Darvishi
- Department of Biomedical Engineering, Meybod University, Meybod, Iran
| | - Alireza Sabzevari
- Department of Biomedical Engineering, Meybod University, Meybod, Iran
| |
Collapse
|
24
|
Xiu Z, Yang Q, Xie F, Han F, He W, Liao W. Revolutionizing digestive system tumor organoids research: Exploring the potential of tumor organoids. J Tissue Eng 2024; 15:20417314241255470. [PMID: 38808253 PMCID: PMC11131411 DOI: 10.1177/20417314241255470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/02/2024] [Indexed: 05/30/2024] Open
Abstract
Digestive system tumors are the leading cause of cancer-related deaths worldwide. Despite ongoing research, our understanding of their mechanisms and treatment remain inadequate. One promising tool for clinical applications is the use of gastrointestinal tract tumor organoids, which serve as an important in vitro model. Tumor organoids exhibit a genotype similar to the patient's tumor and effectively mimic various biological processes, including tissue renewal, stem cell, and ecological niche functions, and tissue response to drugs, mutations, or injury. As such, they are valuable for drug screening, developing novel drugs, assessing patient outcomes, and supporting immunotherapy. In addition, innovative materials and techniques can be used to optimize tumor organoid culture systems. Several applications of digestive system tumor organoids have been described and have shown promising results in related aspects. In this review, we discuss the current progress, limitations, and prospects of this model for digestive system tumors.
Collapse
Affiliation(s)
- Zhian Xiu
- Department of Medical Laboratory, Clinical Medical College, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fusheng Xie
- Department of Medical Laboratory, Clinical Medical College, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Feng Han
- Department of Medical Laboratory, Clinical Medical College, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Weiwei He
- Department of Medical Laboratory, Clinical Medical College, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Weifang Liao
- Department of Medical Laboratory, Clinical Medical College, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| |
Collapse
|
25
|
Udagawa D, Nagata S, Yagi H, Nishi K, Morisaku T, Adachi S, Nakano Y, Tanaka M, Hori S, Hasegawa Y, Abe Y, Kitago M, Kitagawa Y. A Novel Approach to Orthotopic Hepatocyte Transplantation Engineered With Liver Hydrogel for Fibrotic Livers, Enhancing Cell-Cell Interaction and Angiogenesis. Cell Transplant 2024; 33:9636897241253700. [PMID: 38770981 PMCID: PMC11110510 DOI: 10.1177/09636897241253700] [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: 10/08/2023] [Revised: 04/22/2024] [Accepted: 04/22/2024] [Indexed: 05/22/2024] Open
Abstract
Hepatocyte transplantation (HCT) is a potential bridging therapy or an alternative to liver transplantation. Conventionally, single-cell hepatocytes are injected via the portal vein. This strategy, however, has yet to overcome poor cell engraftment and function. Therefore, we developed an orthotopic HCT method using a liver-derived extracellular matrix (L-ECM) gel. PXB cells (flesh mature human hepatocytes) were dispersed into the hydrogel solution in vitro, and the gel solution was immediately gelated in 37°C incubators to investigate the affinity between mature human hepatocyte and the L-ECM gel. During the 3-day cultivation in hepatocyte medium, PXB cells formed cell aggregates via cell-cell interactions. Quantitative analysis revealed human albumin production in culture supernatants. For the in vivo assay, PXB cells were encapsulated in the L-ECM gel and transplanted between the liver lobes of normal rats. Pathologically, the L-ECM gel was localized at the transplant site and retained PXB cells. Cell survival and hepatic function marker expression were verified in another rat model wherein thioacetamide was administered to induce liver fibrosis. Moreover, cell-cell interactions and angiogenesis were enhanced in the L-ECM gel compared with that in the collagen gel. Our results indicate that L-ECM gels can help engraft transplanted hepatocytes and express hepatic function as a scaffold for cell transplantation.
Collapse
Affiliation(s)
- Daisuke Udagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Shogo Nagata
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Yagi
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Kotaro Nishi
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | | | - Shungo Adachi
- Fundamental Innovative Oncology Core, National Cancer Center Research Institute, Tokyo, Japan
| | - Yutaka Nakano
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masayuki Tanaka
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Shutaro Hori
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yasushi Hasegawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yuta Abe
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Minoru Kitago
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
26
|
Estridge RC, O’Neill JE, Keung AJ. Matrigel Tunes H9 Stem Cell-Derived Human Cerebral Organoid Development. ORGANOIDS 2023; 2:165-176. [PMID: 38196836 PMCID: PMC10776236 DOI: 10.3390/organoids2040013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Human cerebral organoids are readily generated from human embryonic stem cells and human induced pluripotent stem cells and are useful in studying human neurodevelopment. Recent work with human cerebral organoids have explored the creation of different brain regions and the impacts of soluble and mechanical cues. Matrigel is a gelatinous, heterogenous mixture of extracellular matrix proteins, morphogens, and growth factors secreted by Engelbreth-Holm-Swarm mouse sarcoma cells. It is a core component of almost all cerebral organoid protocols, generally supporting neuroepithelial budding and tissue polarization; yet, its roles and effects beyond its general requirement in organoid protocols are not well understood, and its mode of delivery is variable, including the embedding of organoids within it or its delivery in soluble form. Given its widespread usage, we asked how H9 stem cell-derived hCO development and composition are affected by Matrigel dosage and delivery method. We found Matrigel exposure influences organoid size, morphology, and cell type composition. We also showed that greater amounts of Matrigel promote an increase in the number of choroid plexus (ChP) cells, and this increase is regulated by the BMP4 pathway. These results illuminate the effects of Matrigel on human cerebral organoid development and the importance of delivery mode and amount on organoid phenotype and composition.
Collapse
Affiliation(s)
- R. Chris Estridge
- Department of Chemical & Biomolecular Engineering, North Carolina State University, Raleigh, NC 27606, USA
| | - Jennifer E. O’Neill
- Genetics Program, Department of Biological Sciences, North Carolina State University, Raleigh, NC 27606, USA;
| | - Albert J. Keung
- Department of Chemical & Biomolecular Engineering, North Carolina State University, Raleigh, NC 27606, USA
| |
Collapse
|
27
|
Vasudevan A, Majumder N, Sharma I, Kaur I, Sundarrajan S, Venugopal JR, Vijayaraghavan P, Singh N, Ramakrishna S, Ghosh S, M Tripathi D, Kaur S. Liver Extracellular Matrix-Based Nanofiber Scaffolds for the Culture of Primary Hepatocytes and Drug Screening. ACS Biomater Sci Eng 2023; 9:6357-6368. [PMID: 37847169 DOI: 10.1021/acsbiomaterials.3c01216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
Immortalized liver cell lines and primary hepatocytes are currently used as in vitro models for hepatotoxic drug screening. However, a decline in the viability and functionality of hepatocytes with time is an important limitation of these culture models. Advancements in tissue engineering techniques have allowed us to overcome this challenge by designing suitable scaffolds for maintaining viable and functional primary hepatocytes for a longer period of time in culture. In the current study, we fabricated liver-specific nanofiber scaffolds with polylactic acid (PLA) along with a decellularized liver extracellular matrix (LEM) by the electrospinning technique. The fabricated hybrid PLA-LEM scaffolds were more hydrophilic and had better swelling properties than the PLA scaffolds. The hybrid scaffolds had a pore size of 38 ± 8 μm and supported primary rat hepatocyte cultures for 10 days. Increased viability (2-fold increase in the number of live cells) and functionality (5-fold increase in albumin secretion) were observed in primary hepatocytes cultured on the PLA-LEM scaffolds as compared to those on conventional collagen-coated plates on day 10 of culture. A significant increase in CYP1A2 enzyme activity was observed in hepatocytes cultured on PLA-LEM hybrid scaffolds in comparison to those on collagen upon induction with phenobarbital. Drugs like acetaminophen and rifampicin showed the highest toxicity in hepatocytes cultured on hybrid scaffolds. Also, the lethal dose of these drugs in rodents was accurately predicted as 1.6 g/kg and 594 mg/kg, respectively, from the corresponding IC50 values obtained from drug-treated hepatocytes on hybrid scaffolds. Thus, the fabricated liver-specific electrospun scaffolds maintained primary hepatocyte viability and functionality for an extended period in culture and served as an effective ex vivo drug screening platform to predict an accurate in vivo drug-induced hepatotoxicity.
Collapse
Affiliation(s)
- Ashwini Vasudevan
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
- Amity Institute of Biotechnology, Sector-125, Amity University Uttar Pradesh, Noida 201301, India
| | - Nilotpal Majumder
- Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Indu Sharma
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Impreet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Subramanian Sundarrajan
- Department of Mechanical Engineering, National University of Singapore, Singapore 117581, Singapore
- Department of Prosthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India
| | - Jayarama Reddy Venugopal
- Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang, Kuantan 26600, Malaysia
| | - Pooja Vijayaraghavan
- Amity Institute of Biotechnology, Sector-125, Amity University Uttar Pradesh, Noida 201301, India
| | - Neetu Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Seeram Ramakrishna
- Department of Mechanical Engineering, National University of Singapore, Singapore 117581, Singapore
| | - Sourabh Ghosh
- Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Dinesh M Tripathi
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| |
Collapse
|
28
|
Mir TA, Alzhrani A, Nakamura M, Iwanaga S, Wani SI, Altuhami A, Kazmi S, Arai K, Shamma T, Obeid DA, Assiri AM, Broering DC. Whole Liver Derived Acellular Extracellular Matrix for Bioengineering of Liver Constructs: An Updated Review. Bioengineering (Basel) 2023; 10:1126. [PMID: 37892856 PMCID: PMC10604736 DOI: 10.3390/bioengineering10101126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 10/29/2023] Open
Abstract
Biomaterial templates play a critical role in establishing and bioinstructing three-dimensional cellular growth, proliferation and spatial morphogenetic processes that culminate in the development of physiologically relevant in vitro liver models. Various natural and synthetic polymeric biomaterials are currently available to construct biomimetic cell culture environments to investigate hepatic cell-matrix interactions, drug response assessment, toxicity, and disease mechanisms. One specific class of natural biomaterials consists of the decellularized liver extracellular matrix (dECM) derived from xenogeneic or allogeneic sources, which is rich in bioconstituents essential for the ultrastructural stability, function, repair, and regeneration of tissues/organs. Considering the significance of the key design blueprints of organ-specific acellular substrates for physiologically active graft reconstruction, herein we showcased the latest updates in the field of liver decellularization-recellularization technologies. Overall, this review highlights the potential of acellular matrix as a promising biomaterial in light of recent advances in the preparation of liver-specific whole organ scaffolds. The review concludes with a discussion of the challenges and future prospects of liver-specific decellularized materials in the direction of translational research.
Collapse
Affiliation(s)
- Tanveer Ahmed Mir
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
| | - Alaa Alzhrani
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21423, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia
| | - Makoto Nakamura
- Division of Biomedical System Engineering, Graduate School of Science and Engineering for Education, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan; (M.N.); (S.I.)
| | - Shintaroh Iwanaga
- Division of Biomedical System Engineering, Graduate School of Science and Engineering for Education, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan; (M.N.); (S.I.)
| | - Shadil Ibrahim Wani
- Division of Biomedical System Engineering, Graduate School of Science and Engineering for Education, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan; (M.N.); (S.I.)
| | - Abdullah Altuhami
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
| | - Shadab Kazmi
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
- Department of Child Health, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Kenchi Arai
- Department of Clinical Biomaterial Applied Science, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Talal Shamma
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
| | - Dalia A. Obeid
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
| | - Abdullah M. Assiri
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia
| | - Dieter C. Broering
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia (T.S.)
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia
| |
Collapse
|
29
|
Li C, Zhang Y, Du Y, Hou Z, Zhang Y, Cui W, Chen W. A Review of Advanced Biomaterials and Cells for the Production of Bone Organoid. SMALL SCIENCE 2023; 3:2300027. [PMID: 40213606 PMCID: PMC11935846 DOI: 10.1002/smsc.202300027] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/25/2023] [Indexed: 02/18/2025] Open
Abstract
Rapid advancements in traditional bone tissue engineering have led to innovation in bone repair models and the resolution of insurmountable clinical issues like graft scarcity. The pathophysiological process of treating bone disease, however, is a multidimensional and multimodal regenerative regulatory mechanism that includes numerous immune, inflammatory, or metabolic responses related to the graft or the organism itself. Based on a 3D in vitro cell culture system that is remarkably identical to the body's bone tissue, the bone organoid is a biomimicking bone organ environment. It can accurately mimic the actual repair and regeneration condition in vivo because it shares the same physiological function, structure, morphology, and metabolic process as endogenous bone tissue. As a disruptive regenerative medicine technology, it has wide application prospects in the fields of organ development, gene editing, disease modeling, and precision therapy. Herein, the development process and physiological basis of different cell-based bone organoids are reviewed, the current status of the application of different materials, cells, and construction methods for building bone organoids is described, and the prospects and challenges for the development of bone organoids in future medical fields is discussed.
Collapse
Affiliation(s)
- Chao Li
- Department of Orthopaedic SurgeryKey Laboratory of Biomechanics of Hebei ProvinceOrthopaedic Research Institution of Hebei ProvinceNHC Key Laboratory of Intelligent Orthopaedic EquipmentThe Third Hospital of Hebei Medical UniversityNo.139 Ziqiang RoadShijiazhuang050051P. R. China
| | - Yipu Zhang
- Department of Orthopaedic SurgeryKey Laboratory of Biomechanics of Hebei ProvinceOrthopaedic Research Institution of Hebei ProvinceNHC Key Laboratory of Intelligent Orthopaedic EquipmentThe Third Hospital of Hebei Medical UniversityNo.139 Ziqiang RoadShijiazhuang050051P. R. China
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Yawei Du
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Zhiyong Hou
- Department of Orthopaedic SurgeryKey Laboratory of Biomechanics of Hebei ProvinceOrthopaedic Research Institution of Hebei ProvinceNHC Key Laboratory of Intelligent Orthopaedic EquipmentThe Third Hospital of Hebei Medical UniversityNo.139 Ziqiang RoadShijiazhuang050051P. R. China
| | - Yingze Zhang
- Department of Orthopaedic SurgeryKey Laboratory of Biomechanics of Hebei ProvinceOrthopaedic Research Institution of Hebei ProvinceNHC Key Laboratory of Intelligent Orthopaedic EquipmentThe Third Hospital of Hebei Medical UniversityNo.139 Ziqiang RoadShijiazhuang050051P. R. China
| | - Wenguo Cui
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Wei Chen
- Department of Orthopaedic SurgeryKey Laboratory of Biomechanics of Hebei ProvinceOrthopaedic Research Institution of Hebei ProvinceNHC Key Laboratory of Intelligent Orthopaedic EquipmentThe Third Hospital of Hebei Medical UniversityNo.139 Ziqiang RoadShijiazhuang050051P. R. China
| |
Collapse
|
30
|
Zamudio-Ceja RB, Garcia-Contreras R, Chavez-Granados PA, Aranda-Herrera B, Alvarado-Garnica H, Jurado CA, Fischer NG. Decellularized Scaffolds of Nopal ( Opuntia Ficus-indica) for Bioengineering in Regenerative Dentistry. J Funct Biomater 2023; 14:jfb14050252. [PMID: 37233362 DOI: 10.3390/jfb14050252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/23/2023] [Accepted: 04/27/2023] [Indexed: 05/27/2023] Open
Abstract
Opuntia Ficus-indica, or nopal, is traditionally used for its medicinal properties in Mexico. This study aims to decellularize and characterize nopal (Opuntia Ficus-indica) scaffolds, assess their degradation and the proliferation of hDPSC, and determine potential pro-inflammatory effects by assessing the expression of cyclooxygenase 1 and 2 (COX-1 and 2). The scaffolds were decellularized using a 0.5% sodium dodecyl sulfate (SDS) solution and confirmed by color, optical microscopy, and SEM. The degradation rates and mechanical properties of the scaffolds were determined by weight and solution absorbances using trypsin and PBS and tensile strength testing. Human dental pulp stem cells (hDPSCs) primary cells were used for scaffold-cell interaction and proliferation assays, as well as an MTT assay to determine proliferation. Proinflammatory protein expression of COX-I and -II was discovered by Western blot assay, and the cultures were induced into a pro-inflammatory state with interleukin 1-β. The nopal scaffolds exhibited a porous structure with an average pore size of 252 ± 77 μm. The decellularized scaffolds showed a 57% reduction in weight loss during hydrolytic degradation and a 70% reduction during enzymatic degradation. There was no difference in tensile strengths between native and decellularized scaffolds (12.5 ± 1 and 11.8 ± 0.5 MPa). Furthermore, hDPSCs showed a significant increase in cell viability of 95% and 106% at 168 h for native and decellularized scaffolds, respectively. The combination of the scaffold and hDPSCs did not cause an increase in the expression of COX-1 and COX-2 proteins. However, when the combination was exposed to IL-1β, there was an increase in the expression of COX-2. This study demonstrates the potential application of nopal scaffolds in tissue engineering and regenerative medicine or dentistry, owing to their structural characteristics, degradation properties, mechanical properties, ability to induce cell proliferation, and lack of enhancement of pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Ruth Betsabe Zamudio-Ceja
- Interdisciplinary Research Laboratory, Nanostructures, and Biomaterials Area, National School of Higher Studies (ENES) Leon, National Autonomous University of Mexico (UNAM), Leon 37684, Gto, Mexico
| | - Rene Garcia-Contreras
- Interdisciplinary Research Laboratory, Nanostructures, and Biomaterials Area, National School of Higher Studies (ENES) Leon, National Autonomous University of Mexico (UNAM), Leon 37684, Gto, Mexico
| | - Patricia Alejandra Chavez-Granados
- Interdisciplinary Research Laboratory, Nanostructures, and Biomaterials Area, National School of Higher Studies (ENES) Leon, National Autonomous University of Mexico (UNAM), Leon 37684, Gto, Mexico
| | - Benjamin Aranda-Herrera
- Interdisciplinary Research Laboratory, Nanostructures, and Biomaterials Area, National School of Higher Studies (ENES) Leon, National Autonomous University of Mexico (UNAM), Leon 37684, Gto, Mexico
| | - Hugo Alvarado-Garnica
- Interdisciplinary Research Laboratory, Nanostructures, and Biomaterials Area, National School of Higher Studies (ENES) Leon, National Autonomous University of Mexico (UNAM), Leon 37684, Gto, Mexico
| | - Carlos A Jurado
- Department of Prosthodontics, The University of Iowa College of Dentistry and Dental Clinics, Iowa City, IA 52242, USA
| | - Nicholas G Fischer
- Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
31
|
Silva-Pedrosa R, Salgado AJ, Ferreira PE. Revolutionizing Disease Modeling: The Emergence of Organoids in Cellular Systems. Cells 2023; 12:930. [PMID: 36980271 PMCID: PMC10047824 DOI: 10.3390/cells12060930] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/03/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Cellular models have created opportunities to explore the characteristics of human diseases through well-established protocols, while avoiding the ethical restrictions associated with post-mortem studies and the costs associated with researching animal models. The capability of cell reprogramming, such as induced pluripotent stem cells (iPSCs) technology, solved the complications associated with human embryonic stem cells (hESC) usage. Moreover, iPSCs made significant contributions for human medicine, such as in diagnosis, therapeutic and regenerative medicine. The two-dimensional (2D) models allowed for monolayer cellular culture in vitro; however, they were surpassed by the three-dimensional (3D) cell culture system. The 3D cell culture provides higher cell-cell contact and a multi-layered cell culture, which more closely respects cellular morphology and polarity. It is more tightly able to resemble conditions in vivo and a closer approach to the architecture of human tissues, such as human organoids. Organoids are 3D cellular structures that mimic the architecture and function of native tissues. They are generated in vitro from stem cells or differentiated cells, such as epithelial or neural cells, and are used to study organ development, disease modeling, and drug discovery. Organoids have become a powerful tool for understanding the cellular and molecular mechanisms underlying human physiology, providing new insights into the pathogenesis of cancer, metabolic diseases, and brain disorders. Although organoid technology is up-and-coming, it also has some limitations that require improvements.
Collapse
Affiliation(s)
- Rita Silva-Pedrosa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; (A.J.S.); (P.E.F.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
- Centre of Biological Engineering (CEB), Department of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| | - António José Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; (A.J.S.); (P.E.F.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Pedro Eduardo Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; (A.J.S.); (P.E.F.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| |
Collapse
|
32
|
Zhu L, Yuhan J, Yu H, Zhang B, Huang K, Zhu L. Decellularized Extracellular Matrix for Remodeling Bioengineering Organoid's Microenvironment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023:e2207752. [PMID: 36929582 DOI: 10.1002/smll.202207752] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/17/2023] [Indexed: 06/18/2023]
Abstract
Over the past decade, stem cell- and tumor-derived organoids are the most promising models in developmental biology and disease modeling, respectively. The matrix is one of three main elements in the construction of an organoid and the most important module of its extracellular microenvironment. However, the source of the currently available commercial matrix, Matrigel, limits the application of organoids in clinical medicine. It is worth investigating whether the original decellularized extracellular matrix (dECM) can be exploited as the matrix of organoids and improving organoid construction are very important. In this review, tissue decellularization protocols and the characteristics of decellularization methods, the mechanical support and biological cues of extraccellular matrix (ECM), methods for construction of multifunctional dECM and responsive dECM hydrogel, and the potential applications of functional dECM are summarized. In addition, some expectations are provided for dECM as the matrix of organoids in clinical applications.
Collapse
Affiliation(s)
- Liye Zhu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, P. R. China
- College of Veterinary Medicine, China Agricultural University, Beijing, 100094, P. R. China
| | - Jieyu Yuhan
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, P. R. China
| | - Hao Yu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, P. R. China
| | - Boyang Zhang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, P. R. China
| | - Kunlun Huang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, P. R. China
| | - Longjiao Zhu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, P. R. China
| |
Collapse
|
33
|
Peng B, Du L, Zhang T, Chen J, Xu B. Research progress in decellularized extracellular matrix hydrogels for intervertebral disc degeneration. Biomater Sci 2023; 11:1981-1993. [PMID: 36734099 DOI: 10.1039/d2bm01862d] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
As one of the most common clinical disorders, low back pain (LBP) influences patient quality of life and causes substantial social and economic burdens. Many factors can result in LBP, the most common of which is intervertebral disc degeneration (IDD). The progression of IDD cannot be alleviated by conservative or surgical treatments, and gene therapy, growth factor therapy, and cell therapy have their own limitations. Recently, research on the use of hydrogel biomaterials for the treatment of IDD has garnered great interest, and satisfactory treatment results have been achieved. This article describes the classification of hydrogels, the methods of decellularized extracellular matrix (dECM) production and the various types of gel formation. The current research on dECM hydrogels for the treatment of IDD is described in detail in this article. First, an overview of the material sources, decellularization methods, and gel formation methods is given. The focus is on research performed over the last three years, which mainly consists of bovine and porcine NP tissues, while for decellularization methods, combinations of several approaches are primarily used. dECM hydrogels have significantly improved mechanical properties after the polymers are cross-linked. The main effects of these gels include induction of stem cell differentiation to intervertebral disc (IVD) cells, good mechanical properties to restore IVD height after polymer cross-linking, and slow release of exosomes. Finally, the challenges and problems still faced by dECM hydrogels for the treatment of IDD are summarised, and potential solutions are proposed. This paper is the first to summarise the research on dECM hydrogels for the treatment of IDD and aims to provide a theoretical reference for subsequent studies.
Collapse
Affiliation(s)
- Bing Peng
- Tianjin University of Traditional Chinese Medicine, No.10, Poyang Lake Road, Jinghai District, Tianjin, 301617, China
| | - Lilong Du
- Tianjin Hospital, Tianjin, No.406, Jiefang South Road, Hexi District, Tianjin, 301617, China.
| | - Tongxing Zhang
- Tianjin Hospital, Tianjin, No.406, Jiefang South Road, Hexi District, Tianjin, 301617, China.
| | - Jiangping Chen
- Liuyang Hospital of Traditional Chinese Medicine, Beizhengzhong Road, Hunan, 410399, China.
| | - Baoshan Xu
- Tianjin Hospital, Tianjin, No.406, Jiefang South Road, Hexi District, Tianjin, 301617, China.
| |
Collapse
|
34
|
Kort-Mascort J, Flores-Torres S, Peza-Chavez O, Jang JH, Pardo LA, Tran SD, Kinsella J. Decellularized ECM hydrogels: prior use considerations, applications, and opportunities in tissue engineering and biofabrication. Biomater Sci 2023; 11:400-431. [PMID: 36484344 DOI: 10.1039/d2bm01273a] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Tissue development, wound healing, pathogenesis, regeneration, and homeostasis rely upon coordinated and dynamic spatial and temporal remodeling of extracellular matrix (ECM) molecules. ECM reorganization and normal physiological tissue function, require the establishment and maintenance of biological, chemical, and mechanical feedback mechanisms directed by cell-matrix interactions. To replicate the physical and biological environment provided by the ECM in vivo, methods have been developed to decellularize and solubilize tissues which yield organ and tissue-specific bioactive hydrogels. While these biomaterials retain several important traits of the native ECM, the decellularizing process, and subsequent sterilization, and solubilization result in fragmented, cleaved, or partially denatured macromolecules. The final product has decreased viscosity, moduli, and yield strength, when compared to the source tissue, limiting the compatibility of isolated decellularized ECM (dECM) hydrogels with fabrication methods such as extrusion bioprinting. This review describes the physical and bioactive characteristics of dECM hydrogels and their role as biomaterials for biofabrication. In this work, critical variables when selecting the appropriate tissue source and extraction methods are identified. Common manual and automated fabrication techniques compatible with dECM hydrogels are described and compared. Fabrication and post-manufacturing challenges presented by the dECM hydrogels decreased mechanical and structural stability are discussed as well as circumvention strategies. We further highlight and provide examples of the use of dECM hydrogels in tissue engineering and their role in fabricating complex in vitro 3D microenvironments.
Collapse
Affiliation(s)
| | | | - Omar Peza-Chavez
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada.
| | - Joyce H Jang
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada.
| | | | - Simon D Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Joseph Kinsella
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
35
|
Gholami M, Tajabadi M, Khavandi A, Azarpira N. Synthesis, optimization, and cell response investigations of natural-based, thermoresponsive, injectable hydrogel: An attitude for 3D hepatocyte encapsulation and cell therapy. Front Bioeng Biotechnol 2023; 10:1075166. [PMID: 36686232 PMCID: PMC9853065 DOI: 10.3389/fbioe.2022.1075166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/19/2022] [Indexed: 01/09/2023] Open
Abstract
For the purpose of developing a 3D vehicle for the delivery of hepatocytes in cell therapy, the improved system of crosslinker and new gelling agent combinations consisting of glycerophosphate and sodium hydrogen carbonate have been employed to produce injectable, thermoresponsive hydrogels based on chitosan and silk fibroin. Adjusting the polymer-to-gelling agent ratio and utilizing a chemical crosslinker developed hydrogel scaffolds with optimal gelling time and pH. Applying sodium hydrogen carbonate neutralizes chitosan while keeping its thermoresponsive characteristics and decreases glycerophosphate from 60% to 30%. Genipin boosts the mechanical properties of hydrogel without affecting the gel time. Due to their stable microstructure and lower amine availability, genipin-containing materials have a low swelling ratio, around six compared to eight for those without genipin. Hydrogels that are crosslinked degrade about half as fast as those that are not. The slowerr degradation of Silk fibroin compared to chitosan makes it an efficient degradation inhibitor in silk-containing formulations. All of the optimized samples showed less than 5% hemolytic activity, indicating that they lacked hemolytic characteristics. The acceptable cell viability in crosslinked hydrogels ranges from 72% to 91% due to the decreasing total salt concentration, which protects cells from hyperosmolality. The pH of hydrogels and their interstitial pores kept most encapsulated cells alive and functioning for 24 h. Urea levels are higher in the encapsulation condition compared to HepG2 cultivated alone, and this may be due to cell-matrix interactions that boost liver-specific activity. Urea synthesis in genipin crosslinked hydrogels increased dramatically from day 1 (about 4 mg dl-1) to day 3 (approximately 6 mg dl-1), suggesting the enormous potential of these hydrogels for cell milieu preparation. All mentioned findings represent that the optimized system may be a promising candidate for liver regeneration.
Collapse
Affiliation(s)
- Mahnaz Gholami
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology (IUST), Tehran, Iran
| | - Maryam Tajabadi
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology (IUST), Tehran, Iran,*Correspondence: Maryam Tajabadi,
| | - Alireza Khavandi
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology (IUST), Tehran, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Science, Shiraz, Iran
| |
Collapse
|
36
|
Bioengineering Liver Organoids for Diseases Modelling and Transplantation. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9120796. [PMID: 36551002 PMCID: PMC9774794 DOI: 10.3390/bioengineering9120796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
Organoids as three-dimension (3D) cellular organizations partially mimic the physiological functions and micro-architecture of native tissues and organs, holding great potential for clinical applications. Advances in the identification of essential factors including physical cues and biochemical signals for controlling organoid development have contributed to the success of growing liver organoids from liver tissue and stem/progenitor cells. However, to recapitulate the physiological properties and the architecture of a native liver, one has to generate liver organoids that contain all the major liver cell types in correct proportions and relative 3D locations as found in a native liver. Recent advances in stem-cell-, biomaterial- and engineering-based approaches have been incorporated into conventional organoid culture methods to facilitate the development of a more sophisticated liver organoid culture resembling a near to native mini-liver in a dish. However, a comprehensive review on the recent advancement in the bioengineering liver organoid is still lacking. Here, we review the current liver organoid systems, focusing on the construction of the liver organoid system with various cell sources, the roles of growth factors for engineering liver organoids, as well as the recent advances in the bioengineering liver organoid disease models and their biomedical applications.
Collapse
|
37
|
Li G, Liu S, Chen W, Jiang Z, Luo Y, Wang D, Zheng Y, Liu Y. Acellularized Uvea Hydrogel as Novel Injectable Platform for Cell-Based Delivering Treatment of Retinal Degeneration and Optimizing Retinal Organoids Inducible System. Adv Healthc Mater 2022; 11:e2202114. [PMID: 36189847 DOI: 10.1002/adhm.202202114] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Indexed: 01/28/2023]
Abstract
Replenishing the retina with retinal pigment epithelial (RPE) cells derived from pluripotent stem cells (PSCs) has great promise for treating retinal degenerative diseases, but it is limited by poor cell survival and integration in vivo. Herein, porcine acellular sclera and uvea extracellular matrix (ECM) and their counterpart hydrogels are developed, and their effects on the biological behavior of human induced pluripotent stem cell (hiPSC)-derived RPE cells (hiPSC-RPE) and embryoid body (hiPSC-EB) differentiation are investigated. Both acellular ECM hydrogels have excellent biocompatibility and suitable biodegradability without evoking an obvious immune response. Most importantly, the decellularized uvea hydrogel-delivered cells' injection remarkably promotes the hiPSC-RPE cells' survival and integration in the subretinal space, rescues the photoreceptor cells' death and retinal gliosis, and restores vision in rats with retinal degeneration for a long duration. In addition, medium supplementation with decellularized uvea peptides promotes hiPSC-EBs onset morphogenesis and neural/retinal differentiation, forming layered retinal organoids. This study demonstrates that ECM hydrogel-delivered hiPSC-RPE cells' injection may be a useful approach for treating retinal degeneration disease, combined with an optimized retinal seeding cells' induction program, which has potential for clinical application.
Collapse
Affiliation(s)
- Guilan Li
- Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, #7 Jinsui Road, Tianhe District, Guangzhou, 510060, China.,Research Unit of Ocular Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Sheng Liu
- Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, #7 Jinsui Road, Tianhe District, Guangzhou, 510060, China.,Guangzhou Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou, 510005, China
| | - Wenfei Chen
- Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, #7 Jinsui Road, Tianhe District, Guangzhou, 510060, China
| | - Zhijian Jiang
- Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, #7 Jinsui Road, Tianhe District, Guangzhou, 510060, China
| | - Yuanting Luo
- Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, #7 Jinsui Road, Tianhe District, Guangzhou, 510060, China
| | - Dongliang Wang
- Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, #7 Jinsui Road, Tianhe District, Guangzhou, 510060, China
| | - Yingfeng Zheng
- Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, #7 Jinsui Road, Tianhe District, Guangzhou, 510060, China.,Research Unit of Ocular Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yizhi Liu
- Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, #7 Jinsui Road, Tianhe District, Guangzhou, 510060, China.,Research Unit of Ocular Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, 100730, China
| |
Collapse
|
38
|
Qian S, Mao J, Liu Z, Zhao B, Zhao Q, Lu B, Zhang L, Mao X, Cheng L, Cui W, Zhang Y, Sun X. Stem cells for organoids. SMART MEDICINE 2022; 1:e20220007. [PMID: 39188738 PMCID: PMC11235201 DOI: 10.1002/smmd.20220007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 08/23/2022] [Indexed: 08/28/2024]
Abstract
Organoids are three-dimensional (3D) cell culture systems that simulate the structures and functions of organs, involving applications in disease modeling, drug screening, and cellular developmental biology. The material matrix in organoids can provide a 3D environment for stem cells to differentiate into different cell types and continuously self-renew, thereby realizing the in vitro culture of organs, which has received extensive attention in recent years. However, some challenges still exist in organoids, including low maturity, high heterogeneity, and lack of spatiotemporal regulation. Therefore, in this review, we summarized the culturing protocols and various applications of stem cell-derived organoids and proposed insightful thoughts for engineering stem cells into organoids in view of the current shortcomings, to achieve the further application and clinical translation of stem cells and engineered stem cells in organoid research.
Collapse
Affiliation(s)
- Shutong Qian
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jiayi Mao
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhimo Liu
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Binfan Zhao
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qiuyu Zhao
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bolun Lu
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Liucheng Zhang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiyuan Mao
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Liying Cheng
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wenguo Cui
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yuguang Zhang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiaoming Sun
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
39
|
Kołakowski A, Dziemitko S, Chmielecka A, Żywno H, Bzdęga W, Charytoniuk T, Chabowski A, Konstantynowicz-Nowicka K. Molecular Advances in MAFLD—A Link between Sphingolipids and Extracellular Matrix in Development and Progression to Fibrosis. Int J Mol Sci 2022; 23:ijms231911380. [PMID: 36232681 PMCID: PMC9569877 DOI: 10.3390/ijms231911380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/18/2022] [Accepted: 09/23/2022] [Indexed: 11/25/2022] Open
Abstract
Metabolic-Associated Fatty Liver Disease (MAFLD) is a major cause of liver diseases globally and its prevalence is expected to grow in the coming decades. The main cause of MAFLD development is changed in the composition of the extracellular matrix (ECM). Increased production of matrix molecules and inflammatory processes lead to progressive fibrosis, cirrhosis, and ultimately liver failure. In addition, increased accumulation of sphingolipids accompanied by increased expression of pro-inflammatory cytokines in the ECM is closely related to lipogenesis, MAFLD development, and its progression to fibrosis. In our work, we will summarize all information regarding the role of sphingolipids e.g., ceramide and S1P in MAFLD development. These sphingolipids seem to have the most significant effect on macrophages and, consequently, HSCs which trigger the entire cascade of overproduction matrix molecules, especially type I and III collagen, proteoglycans, elastin, and also tissue inhibitors of metalloproteinases, which as a result cause the development of liver fibrosis.
Collapse
Affiliation(s)
- Adrian Kołakowski
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Sylwia Dziemitko
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | | | - Hubert Żywno
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Wiktor Bzdęga
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Tomasz Charytoniuk
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland
- Department of Ophthalmology, Antoni Jurasz University Hospital No. 1, 85-094 Bydgoszcz, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | | |
Collapse
|
40
|
Brown M, Li J, Moraes C, Tabrizian M, Li-Jessen NY. Decellularized extracellular matrix: New promising and challenging biomaterials for regenerative medicine. Biomaterials 2022; 289:121786. [DOI: 10.1016/j.biomaterials.2022.121786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 08/22/2022] [Accepted: 08/29/2022] [Indexed: 11/28/2022]
|
41
|
Rahaman MS, Park S, Kang HJ, Sultana T, Gwon JG, Lee BT. Liver tissue-derived ECM loaded nanocellulose-alginate-TCP composite beads for accelerated bone regeneration. Biomed Mater 2022; 17. [PMID: 35952638 DOI: 10.1088/1748-605x/ac8901] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 08/11/2022] [Indexed: 11/11/2022]
Abstract
Guided bone regeneration with osteoinductive scaffolds is a competitive edge of tissue engineering due to faster and more consistent healing. In the present study, we developed such composite beads with nanocellulose reinforced alginate hydrogel that carried β-tricalcium phosphate (β-TCP) nano-powder and liver-derived extracellular matrix (ECM) from porcine. Interestingly, it was observed that the beads' group containing ECM-βTCP-alginate-nanocellulose (ETAC) was more biocompatible with higher cellular affinity than the others comprised of βTCP-alginate-nanocellulose (TAC) and alginate-nanocellulose (AC). Cell attachment on ETAC beads was dramatically increased with time. In parallel with in vitro results, ETAC beads produced uniform cortical and cancellous bone in the femur defect model of rabbits within two months. Although the group TAC also produced noticeable bone in the defect site, the healing quality was improved and regeneration was faster after adding ECM. This conclusion was not only confirmed by micro-anatomical analysis but also demonstrated with X-ray microtomography. In addition, the characteristic moldable and injectable properties made ETAC a promising scaffold for clinical applications.
Collapse
Affiliation(s)
- Md Sohanur Rahaman
- Department of Regenerative Medicine, Soonchunhyang University, 366-1, Ssangyougndong, Cheonan, Chungcheongnam-do, 31538, Korea (the Republic of)
| | - Seongsu Park
- Soonchunhyang University College of Medicine, 366-1, Ssangyougndong, Cheonan, Chungcheongnam-do, 31204, Korea (the Republic of)
| | - Hoe-Jin Kang
- Soonchunhyang University, 366-1, Ssangyougndong, Cheonan, Chungcheongnam-do, 31538, Korea (the Republic of)
| | - Tamanna Sultana
- Soonchunhyang University, 366-1, Ssangyougndong, Cheonan, Chungcheongnam-do, 31538, Korea (the Republic of)
| | - Jae-Gyoung Gwon
- Department of Forest Products, Korea Forest Research Institute, 57, Hoegi-ro, Dongdaemun-gu, Seoul, 02455, Korea (the Republic of)
| | - Byong-Taek Lee
- Department of Biomedical Engineering and Materials, Soonchunhyang University College of Medicine, 366-1, Ssangyougndong, Cheonan, Chungcheongnam-do, 31204, Korea (the Republic of)
| |
Collapse
|
42
|
Lee IS, Takebe T. Narrative engineering of the liver. Curr Opin Genet Dev 2022; 75:101925. [PMID: 35700688 PMCID: PMC10118678 DOI: 10.1016/j.gde.2022.101925] [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: 03/13/2022] [Revised: 05/06/2022] [Accepted: 05/08/2022] [Indexed: 11/30/2022]
Abstract
Liver organoids are primary or pluripotent stem cell-derived three-dimensional structures that recapitulate regenerative or ontogenetic processes in vitro towards biomedical applications including disease modelling and diagnostics, drug safety and efficacy prediction, and therapeutic use. The cellular composition and structural organization of liver organoids may vary depending on the goal at hand, and the key challenge in general is to direct their development in a rational and controlled fashion for gaining targeted maturity, reproducibility, and scalability. Such endeavor begins with a detailed understanding of the biological processes in space and time behind hepatogenesis, followed by precise translation of these narrative processes through a bioengineering approach. Here, we discuss advancements in liver organoid technology through the lens of 'narrative engineering' in an attempt to synergize evolving understanding around molecular and cellular landscape governing hepatogenesis with engineering-inspired approaches for organoidgenesis.
Collapse
Affiliation(s)
- Inkyu S Lee
- Division of Gastroenterology, Hepatology & Nutrition, Developmental Biology, Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039, USA
| | - Takanori Takebe
- Division of Gastroenterology, Hepatology & Nutrition, Developmental Biology, Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Institute of Research, Tokyo Medical and Dental University (TMDU), Tokyo, Japan; Communication Design Center, Advanced Medical Research Center, Yokohama City University Graduate School of Medicine, Japan.
| |
Collapse
|
43
|
Decellularized normal and cancer tissues as tools for cancer research. Cancer Gene Ther 2022; 29:879-888. [PMID: 34785762 DOI: 10.1038/s41417-021-00398-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/23/2021] [Accepted: 10/08/2021] [Indexed: 12/11/2022]
Abstract
Today it is widely accepted that molecular mechanisms triggering cancer initiate with a genetic modification. However, a genetic alteration providing the aberrant clone with a growing advantage over neighboring cells is not sufficient to develop cancer. Currently, tumors are considered a heterogeneous population of cells and an extracellular matrix (ECM) that make up a characteristic microenvironment. Interactions between tumor cells and cancer microenvironment define cancer progression and therapeutic response. To investigate and clarify the role of ECM in the regulation of cancer cell behavior and response to therapy, the decellularization of ECM, a widely used technique in tissue engineering, has been recently employed to develop 3D culture model of disease. In this review, we briefly explore the different components of healthy and pathological ECM and the methods to obtain and characterize the ECM from native bioptic tissue. Finally, we highlight the most relevant applications of ECM in translational cancer research strategies: decellularized ECM, ECM-hydrogel and 3D bioprinting.
Collapse
|
44
|
Wang L, Zhao C, Zheng T, Zhang Y, Liu H, Wang X, Tang X, Zhao B, Liu P. Torin 1 alleviates impairment of TFEB-mediated lysosomal biogenesis and autophagy in TGFBI (p.G623_H626del)-linked Thiel-Behnke corneal dystrophy. Autophagy 2022; 18:765-782. [PMID: 34403298 PMCID: PMC9037417 DOI: 10.1080/15548627.2021.1955469] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 01/02/2023] Open
Abstract
Thiel-Behnke corneal dystrophy (TBCD) is an epithelial-stromal TGFBI dystrophy caused by mutations in the TGFBI (transforming growth factor beta induced) gene, though the underlying mechanisms and pathogenesis of TBCD are still obscure. The study identifies a novel mutation in the TGFBI gene (p.Gly623_His626del) in a TBCD pedigree. Characteristics of the typical vacuole formation, irregular corneal epithelial thickening and thinning, deposition of eosinophilic substances beneath the epithelium, and involvement of the anterior stroma were observed in this pedigree via transmission electron microscopy (TEM) and histological staining. Tgfbi-p.Gly623_Tyr626del mouse models of TBCD were subsequently generated via CRISPR/Cas9 technology, and the above characteristics were further verified via TEM and histological staining. Lysosomal dysfunction and downregulation of differential expression protein CTSD (cathepsin D) were observed using LysoTracker Green DND-26 and proteomic analysis, respectively. Hence, lysosomal dysfunction probably leads to autophagic flux obstruction in TBCD; this was supported by enhanced LC3-II and SQSTM1 levels and decreased CTSD. TFEB (transcription factor EB) was prominently decreased in TBCD corneal fibroblasts and administration of ATP-competitive MTOR inhibitor torin 1 reversed this decline, resulting in the degradation of accumulated mut-TGFBI (mutant TGFBI protein) via the ameliorative lysosomal function and autophagic flux owing to elevated TFEB activity as measured by western blot, confocal microscopy, and flow cytometry. Transfected HEK 293 cells overexpressing human full-length WT-TGFBI and mut-TGFBI were generated to further verify the results obtained in human corneal fibroblasts. Amelioration of lysosome dysfunction may therefore have therapeutic efficacy in the treatment of TBCD.Abbreviations AS-OCT: anterior segment optical coherence tomography; ATP: adenosine triphosphate; Cas9: CRISPR-associated protein 9; CLEAR: coordinated lysosomal expression and regulation; CRISPR: clustered regularly interspaced short palindromic repeats; CTSB: cathepsin B; CTSD: cathepsin D; CTSF: cathepsin F; CTSL: cathepsin L; DNA: deoxyribonucleic acid; ECM: extracellular matrix; Fas1: fasciclin 1; FC: flow cytometry; GAPDH: glyceraldeyde-3-phosphate dehydrogenase; GCD2: granular corneal dystrophy type 2; HE: hematoxylin and eosin; LAMP2: lysosomal-associated membrane protein; MT: mutation type; MTOR: mechanistic target of rapamycin kinase; MTORC1: MTOR complex 1; mut-TGFBI: mutant TGFBI protein; SD: standard deviation; TBCD: Thiel-Behnke corneal dystrophy; TEM: transmission electron microscopy; TFEB: transcription factor EB; TGFBI: transforming growth factor beta induced; WT: wild type.
Collapse
Affiliation(s)
- Liyuan Wang
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chuchu Zhao
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tao Zheng
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yi Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hanruo Liu
- The Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xi Wang
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xianling Tang
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Baowen Zhao
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ping Liu
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
45
|
Francés-Herrero E, Rodríguez-Eguren A, Gómez-Álvarez M, de Miguel-Gómez L, Ferrero H, Cervelló I. Future Challenges and Opportunities of Extracellular Matrix Hydrogels in Female Reproductive Medicine. Int J Mol Sci 2022; 23:3765. [PMID: 35409119 PMCID: PMC8998701 DOI: 10.3390/ijms23073765] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 02/01/2023] Open
Abstract
Bioengineering and reproductive medicine have progressed shoulder to shoulder for several decades. A key point of overlap is the development and clinical translation of technologies to support reproductive health, e.g., scaffold-free constructs, polymeric scaffolds, bioprinting or microfluidics, and hydrogels. Hydrogels are the focus of intense study, and those that are derived from the extracellular matrix (ECM) of reproductive tissues and organs are emerging as promising new players given their results in pre-clinical models. This literature review addresses the recent advances in the use of organ-specific ECM hydrogels in reproductive medicine, considering the entire female reproductive tract. We discuss in-depth papers describing the development of ECM hydrogels, their use in in vitro models, and their in vivo application in preclinical studies. We also summarize the functions of hydrogels, including as grafts, carriers for cell transplantation, or drug depots, and present the potential and possible scope for use of ECM hydrogels in the near future based on recent scientific advances.
Collapse
Affiliation(s)
- Emilio Francés-Herrero
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, 46010 Valencia, Spain;
- Fundación IVI, IVI-RMA Global, 46026 Valencia, Spain; (A.R.-E.); (M.G.-Á.); (L.d.M.-G.); (H.F.)
| | - Adolfo Rodríguez-Eguren
- Fundación IVI, IVI-RMA Global, 46026 Valencia, Spain; (A.R.-E.); (M.G.-Á.); (L.d.M.-G.); (H.F.)
- Reproductive Medicine Research Group, IIS La Fe, 46026 Valencia, Spain
| | - María Gómez-Álvarez
- Fundación IVI, IVI-RMA Global, 46026 Valencia, Spain; (A.R.-E.); (M.G.-Á.); (L.d.M.-G.); (H.F.)
- Reproductive Medicine Research Group, IIS La Fe, 46026 Valencia, Spain
| | - Lucía de Miguel-Gómez
- Fundación IVI, IVI-RMA Global, 46026 Valencia, Spain; (A.R.-E.); (M.G.-Á.); (L.d.M.-G.); (H.F.)
| | - Hortensia Ferrero
- Fundación IVI, IVI-RMA Global, 46026 Valencia, Spain; (A.R.-E.); (M.G.-Á.); (L.d.M.-G.); (H.F.)
- Reproductive Medicine Research Group, IIS La Fe, 46026 Valencia, Spain
| | - Irene Cervelló
- Fundación IVI, IVI-RMA Global, 46026 Valencia, Spain; (A.R.-E.); (M.G.-Á.); (L.d.M.-G.); (H.F.)
- Reproductive Medicine Research Group, IIS La Fe, 46026 Valencia, Spain
| |
Collapse
|
46
|
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: 55] [Impact Index Per Article: 18.3] [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
|
47
|
Xu J, Pan D, Liao W, Jia Z, Pan M, Weng J, Han X, Li S, Li Y, Liang K, Zhou S, Peng Q, Gao Y. Application of 3D Hepatic Plate-Like Liver Model for Statin-Induced Hepatotoxicity Evaluation. Front Bioeng Biotechnol 2022; 10:826093. [PMID: 35372314 PMCID: PMC8968918 DOI: 10.3389/fbioe.2022.826093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/03/2022] [Indexed: 12/12/2022] Open
Abstract
Background: Drug-induced liver injury is one of the main reasons of withdrawals of drugs in postmarketing stages. However, an experimental model(s) which can accurately recapitulates liver functions and reflects the level of drug hepatotoxicity is lack. In this study, we assessed drug hepatotoxicity using a novel three-dimensional hepatic plate-like hydrogel fiber (3D-P) co-culture system. Methods: During the 28-days culture period, the liver-specific functions, hepatocyte polarity, sensitivity of drug-induced toxicity of 3D-P co-culture system were evaluated with 2D co-culture, collagen sandwich co-culture, 3D hybrid hydrogel fiber co-culture and human primary hepatocytes as controls. High-content imaging and analysis (HCA) methods were used to explore the hepatotoxicity mechanism of five statins. Results: The 3D-P co-culture system showed enhancing liver-specific functions, cytochrome P450 enzymes (CYPs) metabolic activity and bile excretion, which were considered to result from improved hepatocyte polarity. Three of the statins may cause acute or chronic hepatotoxicity by via different mechanisms, such as cholestatic liver injury. Conclusion: Our 3D-P co-culture system is characterized by its biomimetic hepatic plate-like structure, long-term stable liver specificity, and prominent bile secretion function, making it applicable for acute/chronic drug hepatotoxicity assessments.
Collapse
Affiliation(s)
- Jiecheng Xu
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Daogang Pan
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Wei Liao
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Zhidong Jia
- Guangzhou Overseas Chinese Hospital, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Mingxin Pan
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Jun Weng
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Xu Han
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Shao Li
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Yang Li
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Kangyan Liang
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Shuqin Zhou
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qing Peng
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
- *Correspondence: Qing Peng, ; Yi Gao,
| | - Yi Gao
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
- *Correspondence: Qing Peng, ; Yi Gao,
| |
Collapse
|
48
|
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
|
49
|
Wang Y, Kankala RK, Ou C, Chen A, Yang Z. Advances in hydrogel-based vascularized tissues for tissue repair and drug screening. Bioact Mater 2022; 9:198-220. [PMID: 34820566 PMCID: PMC8586021 DOI: 10.1016/j.bioactmat.2021.07.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
The construction of biomimetic vasculatures within the artificial tissue models or organs is highly required for conveying nutrients, oxygen, and waste products, for improving the survival of engineered tissues in vitro. In recent times, the remarkable progress in utilizing hydrogels and understanding vascular biology have enabled the creation of three-dimensional (3D) tissues and organs composed of highly complex vascular systems. In this review, we give an emphasis on the utilization of hydrogels and their advantages in the vascularization of tissues. Initially, the significance of vascular elements and the regeneration mechanisms of vascularization, including angiogenesis and vasculogenesis, are briefly introduced. Further, we highlight the importance and advantages of hydrogels as artificial microenvironments in fabricating vascularized tissues or organs, in terms of tunable physical properties, high similarity in physiological environments, and alternative shaping mechanisms, among others. Furthermore, we discuss the utilization of such hydrogels-based vascularized tissues in various applications, including tissue regeneration, drug screening, and organ-on-chips. Finally, we put forward the key challenges, including multifunctionalities of hydrogels, selection of suitable cell phenotype, sophisticated engineering techniques, and clinical translation behind the development of the tissues with complex vasculatures towards their future development.
Collapse
Affiliation(s)
- Ying Wang
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong, 523059, PR China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, Guangdong, 510080, PR China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian, 361021, PR China
| | - Caiwen Ou
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong, 523059, PR China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, Guangdong, 510080, PR China
| | - Aizheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian, 361021, PR China
| | - Zhilu Yang
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong, 523059, PR China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, Guangdong, 510080, PR China
| |
Collapse
|
50
|
Bioinspired Sandcastle Worm-Derived Peptide-Based Hybrid Hydrogel for Promoting the Formation of Liver Spheroids. Gels 2022; 8:gels8030149. [PMID: 35323262 PMCID: PMC8950079 DOI: 10.3390/gels8030149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 12/10/2022] Open
Abstract
The generation of hepatic spheroids is beneficial for a variety of potential applications, including drug development, disease modeling, transplantation, and regenerative medicine. Natural hydrogels are obtained from tissues and have been widely used to promote the growth, differentiation, and retention of specific functionalities of hepatocytes. However, relying on natural hydrogels for the generation of hepatic spheroids, which have batch to batch variations, may in turn limit the previously mentioned potential applications. For this reason, we researched a way to establish a three-dimensional (3D) culture system that more closely mimics the interaction between hepatocytes and their surrounding microenvironments, thereby potentially offering a more promising and suitable system for drug development, disease modeling, transplantation, and regenerative medicine. Here, we developed self-assembling and bioactive hybrid hydrogels to support the generation and growth of hepatic spheroids. Our hybrid hydrogels (PC4/Cultrex) inspired by the sandcastle worm, an Arg-Gly-Asp (RGD) cell adhesion sequence, and bioactive molecules derived from Cultrex BME (Basement Membrane Extract). By performing optimizations to the design, the PC4/Cultrex hybrid hydrogels can enhance HepG2 cells to form spheroids and express their molecular signatures (e.g., Cyp3A4, Cyp7a1, A1at, Afp, Ck7, Ck1, and E-cad). Our study demonstrated that this hybrid hydrogel system offers potential advantages for hepatocytes in proliferating, differentiating, and self-organizing to form hepatic spheroids in a more controllable and reproducible manner. In addition, it is a versatile and cost-effective method for 3D tissue cultures in mass quantities. Importantly, we demonstrate that it is feasible to adapt a bioinspired approach to design biomaterials for 3D culture systems, which accelerates the design of novel peptide structures and broadens our research choices on peptide-based hydrogels.
Collapse
|