1
|
Vasudevan A, Ghosal D, Ram Sahu S, Kumar Jha N, Vijayaraghavan P, Kumar S, Kaur S. Injectable Hydrogels for Liver: Potential for Clinical Translation. Chem Asian J 2025; 20:e202401106. [PMID: 39552124 DOI: 10.1002/asia.202401106] [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: 08/31/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/19/2024]
Abstract
Injectable hydrogels are a sub-type of hydrogels which can be delivered into the host in a minimally invasive manner. They can act as carriers to encapsulate and deliver cells, drugs or active biomolecules across several disease conditions. Polymers, either synthetic or natural, or even a combination of the two, can be used to create injectable hydrogels. Clinically approved injectable hydrogels are being used as dressings for burn wounds, bone and cartilage reconstruction. Injectable hydrogels have recently gained tremendous attention for their delivery into the liver in pre-clinical models. However, their efficacy in clinical studies remains yet to be established. In this article, we describe principles for the design of these injectable hydrogels, delivery strategies and their potential applications in facilitating liver regeneration and ameliorating injury. We also discuss the several constraints related to translation of these hydrogels into clinical settings for liver diseases and deliberate some potential solutions to combat these challenges.
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, Amity University Uttar Pradesh, Sector-125, Noida, 201301, Uttar Pradesh, India
| | - Doyel Ghosal
- Centre for Biomedical Engineering, Indian Institute of Technology, New Delhi, 110016, India
| | - Sita Ram Sahu
- School of Interdisciplinary Research, Indian Institute of Technology, New Delhi, 110016, India
| | - Narsing Kumar Jha
- Department of Applied Mechanics, Indian Institute of Technology, New Delhi, 110016, India
| | - Pooja Vijayaraghavan
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Sector-125, Noida, 201301, Uttar Pradesh, India
| | - Sachin Kumar
- Centre for Biomedical Engineering, Indian Institute of Technology, New Delhi, 110016, India
- Department of Biomedical Engineering, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, 110070, India
| |
Collapse
|
2
|
Li G, Zeng M, Yan Z, Cai S, Ma Y, Wang Y, Li S, Li Y, Zhong K, Xiao M, Fu G, Weng J, Gao Y. HDAC inhibitors support long-term expansion of porcine hepatocytes in vitro. Biomed Pharmacother 2024; 177:116973. [PMID: 38908204 DOI: 10.1016/j.biopha.2024.116973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 06/03/2024] [Accepted: 06/15/2024] [Indexed: 06/24/2024] Open
Abstract
Hepatocyte transplantation is an effective treatment for end-stage liver disease. However, due to the limited supply of human hepatocytes, porcine hepatocytes have garnered attention as a potential alternative source. Nonetheless, traditional primary porcine hepatocytes exhibit certain limitations in function maintenance and in vitro proliferation. This study has discovered that by using histone deacetylase inhibitors (HDACi), primary porcine hepatocytes can be successfully reprogrammed into liver progenitor cells with high proliferative potential. This method enables porcine hepatocytes to proliferate over an extended period in vitro and exhibit increased susceptibility in lentivirus-mediated gene modification. These liver progenitor cells can readily differentiate into mature hepatocytes and, upon microencapsulation transplantation into mice with acute liver failure, significantly improve the survival rate. This research provides new possibilities for the application of porcine hepatocytes in the treatment of end-stage liver disease.
Collapse
Affiliation(s)
- Guanhong Li
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China; 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, Southern Medical University, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Southern Medical University, Guangzhou 510000, China
| | - Min Zeng
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China; 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, Southern Medical University, Guangzhou 510000, China
| | - Zhengming Yan
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China; 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, Southern Medical University, Guangzhou 510000, China
| | - Shaoru Cai
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Yi Ma
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China; 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, Southern Medical University, Guangzhou 510000, China
| | - Yuting Wang
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China; 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, Southern Medical University, Guangzhou 510000, China
| | - Shao Li
- 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, Southern Medical University, Guangzhou 510000, China
| | - Yang Li
- 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, Southern Medical University, Guangzhou 510000, China
| | - Kebo Zhong
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Mingjia Xiao
- Department of Hepatobiliary Surgery, Quzhou People's Hospital, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China.
| | - Gongbo Fu
- Department of Oncology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing 210000, China.
| | - Jun Weng
- Department of Endoscopy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou 510000, China.
| | - Yi Gao
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China; 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, Southern Medical University, Guangzhou 510000, China; State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Southern Medical University, Guangzhou 510000, China.
| |
Collapse
|
3
|
O'Sullivan D, Arora T, Durif C, Uriot O, Brun M, Riu M, Foguet-Romero E, Samarra I, Domingo-Almenara X, Gahan CGM, Etienne-Mesmin L, Blanquet-Diot S. Impact of Western Diet on Enterohemorrhagic Escherichia coli Colonization in the Human In Vitro Mucosal Artificial Colon as Mediated by Gut Microbiota. Nutrients 2024; 16:2046. [PMID: 38999794 PMCID: PMC11243482 DOI: 10.3390/nu16132046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) is a major food-borne pathogen that causes human disease ranging from diarrhea to life-threatening complications. Accumulating evidence demonstrates that the Western diet enhances the susceptibility to enteric infection in mice, but the effect of diet on EHEC colonization and the role of human gut microbiota remains unknown. Our research aimed to investigate the effects of a Standard versus a Western diet on EHEC colonization in the human in vitro Mucosal ARtificial COLon (M-ARCOL) and the associated changes in the gut microbiota composition and activities. After donor selection using simplified fecal batch experiments, two M-ARCOL bioreactors were inoculated with a human fecal sample (n = 4) and were run in parallel, one receiving a Standard diet, the other a Western diet and infected with EHEC O157:H7 strain EDL933. EHEC colonization was dependent on the donor and diet in the luminal samples, but was maintained in the mucosal compartment without elimination, suggesting a favorable niche for the pathogen, and may act as a reservoir. The Western diet also impacted the bacterial short-chain fatty acid and bile acid profiles, with a possible link between high butyrate concentrations and prolonged EHEC colonization. The work demonstrates the application of a complex in vitro model to provide insights into diet, microbiota, and pathogen interactions in the human gut.
Collapse
Affiliation(s)
- Deborah O'Sullivan
- UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Université Clermont Auvergne, 28 Place Henri Dunant, F-63000 Clermont-Ferrand, France
| | - Trisha Arora
- Centre for Omics Sciences (COS), Unique Scientific and Technical Infrastructures (ICTS), Eurecat-Technology Centre of Catalonia & Rovira i Virgili University Joint Unit, 43204 Reus, Spain
- Department of Electrical, Electronic and Control Engineering (DEEEA), Universitat Rovira i Virgili, 43007 Tarragona, Spain
- Computational Metabolomics for Systems Biology Lab, Eurecat-Technology Centre of Catalonia, 08005 Barcelona, Spain
| | - Claude Durif
- UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Université Clermont Auvergne, 28 Place Henri Dunant, F-63000 Clermont-Ferrand, France
| | - Ophélie Uriot
- UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Université Clermont Auvergne, 28 Place Henri Dunant, F-63000 Clermont-Ferrand, France
| | - Morgane Brun
- UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Université Clermont Auvergne, 28 Place Henri Dunant, F-63000 Clermont-Ferrand, France
| | - Marc Riu
- Centre for Omics Sciences (COS), Unique Scientific and Technical Infrastructures (ICTS), Eurecat-Technology Centre of Catalonia & Rovira i Virgili University Joint Unit, 43204 Reus, Spain
| | - Elisabet Foguet-Romero
- Centre for Omics Sciences (COS), Unique Scientific and Technical Infrastructures (ICTS), Eurecat-Technology Centre of Catalonia & Rovira i Virgili University Joint Unit, 43204 Reus, Spain
| | - Iris Samarra
- Centre for Omics Sciences (COS), Unique Scientific and Technical Infrastructures (ICTS), Eurecat-Technology Centre of Catalonia & Rovira i Virgili University Joint Unit, 43204 Reus, Spain
| | - Xavier Domingo-Almenara
- Centre for Omics Sciences (COS), Unique Scientific and Technical Infrastructures (ICTS), Eurecat-Technology Centre of Catalonia & Rovira i Virgili University Joint Unit, 43204 Reus, Spain
- Department of Electrical, Electronic and Control Engineering (DEEEA), Universitat Rovira i Virgili, 43007 Tarragona, Spain
- Computational Metabolomics for Systems Biology Lab, Eurecat-Technology Centre of Catalonia, 08005 Barcelona, Spain
| | - Cormac G M Gahan
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland
- School of Pharmacy, University College Cork, T12 K8AF Cork, Ireland
| | - Lucie Etienne-Mesmin
- UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Université Clermont Auvergne, 28 Place Henri Dunant, F-63000 Clermont-Ferrand, France
| | - Stéphanie Blanquet-Diot
- UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Université Clermont Auvergne, 28 Place Henri Dunant, F-63000 Clermont-Ferrand, France
| |
Collapse
|
4
|
Karaca MA, Kancagi DD, Ozbek U, Ovali E, Gok O. Betulin Stimulates Osteogenic Differentiation of Human Osteoblasts-Loaded Alginate-Gelatin Microbeads. Bioengineering (Basel) 2024; 11:553. [PMID: 38927789 PMCID: PMC11201098 DOI: 10.3390/bioengineering11060553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/06/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Osteoporosis, a terminal illness, has emerged as a global public health problem in recent years. The long-term use of bone anabolic drugs to treat osteoporosis causes multi-morbidity in elderly patients. Alternative therapies, such as allogenic and autogenic tissue grafts, face important issues, such as a limited source of allogenic grafts and tissue rejection in autogenic grafts. However, stem cell therapy has been shown to increase bone regeneration and decrease osteoporotic bone formation. Stem cell therapy combined with betulin (BET) supplementation might be adequate for bone remodeling and new bone tissue generation. In this study, the effect of BET on the viability and osteogenic differentiation of hFOB 1.19 cells was investigated. The cells were encapsulated in alginate-gelatin (AlGel) microbeads. In vitro tests were conducted during the 12 d of incubation. While BET showed cytotoxic activity (>1 µM) toward non-encapsulated hFOB 1.19 cells, encapsulated cells retained their functionality for up to 12 days, even at 5 µM BET. Moreover, the expression of osteogenic markers indicates an enhanced osteo-inductive effect of betulin on encapsulated hFOB 1.19, compared to the non-encapsulated cell culture. The 3D micro-environment of the AlGel microcapsules successfully protects the hFOB 1.19 cells against BET cytotoxicity, allowing BET to improve the mineralization and differentiation of osteoblast cells.
Collapse
Affiliation(s)
- Mehmet Ali Karaca
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey;
| | - Derya Dilek Kancagi
- Acibadem Labcell Cellular Therapy Laboratory, 34752 Istanbul, Turkey; (D.D.K.); (E.O.)
| | - Ugur Ozbek
- Medical Genetics Department, School of Medicine, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey;
| | - Ercument Ovali
- Acibadem Labcell Cellular Therapy Laboratory, 34752 Istanbul, Turkey; (D.D.K.); (E.O.)
| | - Ozgul Gok
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey
| |
Collapse
|
5
|
Wang H, Wen L, Jiang F, Ren P, Yang Y, Song S, Yang Z, Wang Y. A comprehensive review of advances in hepatocyte microencapsulation: selecting materials and preserving cell viability. Front Immunol 2024; 15:1385022. [PMID: 38694507 PMCID: PMC11061843 DOI: 10.3389/fimmu.2024.1385022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 03/28/2024] [Indexed: 05/04/2024] Open
Abstract
Liver failure represents a critical medical condition with a traditionally grim prognosis, where treatment options have been notably limited. Historically, liver transplantation has stood as the sole definitive cure, yet the stark disparity between the limited availability of liver donations and the high demand for such organs has significantly hampered its feasibility. This discrepancy has necessitated the exploration of hepatocyte transplantation as a temporary, supportive intervention. In light of this, our review delves into the burgeoning field of hepatocyte transplantation, with a focus on the latest advancements in maintaining hepatocyte function, co-microencapsulation techniques, xenogeneic hepatocyte transplantation, and the selection of materials for microencapsulation. Our examination of hepatocyte microencapsulation research highlights that, to date, most studies have been conducted in vitro or using liver failure mouse models, with a notable paucity of experiments on larger mammals. The functionality of microencapsulated hepatocytes is primarily inferred through indirect measures such as urea and albumin production and the rate of ammonia clearance. Furthermore, research on the mechanisms underlying hepatocyte co-microencapsulation remains limited, and the practicality of xenogeneic hepatocyte transplantation requires further validation. The potential of hepatocyte microencapsulation extends beyond the current scope of application, suggesting a promising horizon for liver failure treatment modalities. Innovations in encapsulation materials and techniques aim to enhance cell viability and function, indicating a need for comprehensive studies that bridge the gap between small-scale laboratory success and clinical applicability. Moreover, the integration of bioengineering and regenerative medicine offers novel pathways to refine hepatocyte transplantation, potentially overcoming the challenges of immune rejection and ensuring the long-term functionality of transplanted cells. In conclusion, while hepatocyte microencapsulation and transplantation herald a new era in liver failure therapy, significant strides must be made to translate these experimental approaches into viable clinical solutions. Future research should aim to expand the experimental models to include larger mammals, thereby providing a clearer understanding of the clinical potential of these therapies. Additionally, a deeper exploration into the mechanisms of cell survival and function within microcapsules, alongside the development of innovative encapsulation materials, will be critical in advancing the field and offering new hope to patients with liver failure.
Collapse
Affiliation(s)
- Hailian Wang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Center of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, China
| | - Lebin Wen
- Department of Thyroid, Sichuan Second Hospital of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Fengdi Jiang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Pengyu Ren
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yixin Yang
- Department of Clinical Medicine, The First Clinical Medical College of Norman Bethune University of Medical Sciences, Jilin, China
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Zhengteng Yang
- Department of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Yi Wang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
6
|
Deschamps C, Denis S, Humbert D, Priymenko N, Chalancon S, De Bodt J, Van de Wiele T, Ipharraguerre I, Alvarez-Acero I, Achard C, Apper E, Blanquet-Diot S. Canine Mucosal Artificial Colon: development of a new colonic in vitro model adapted to dog sizes. Appl Microbiol Biotechnol 2024; 108:166. [PMID: 38261090 PMCID: PMC10806056 DOI: 10.1007/s00253-023-12987-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/18/2023] [Accepted: 12/24/2023] [Indexed: 01/24/2024]
Abstract
Differences in dog breed sizes are an important determinant of variations in digestive physiology, mainly related to the large intestine. In vitro gut models are increasingly used as alternatives to animal experiments for technical, cost, societal, and regulatory reasons. Up to now, only one in vitro model of the canine colon incorporates the dynamics of different canine gut regions, yet no adaptations exist to reproduce size-related digestive parameters. To address this limitation, we developed a new model of the canine colon, the CANIne Mucosal ARtificial COLon (CANIM-ARCOL), simulating main physiochemical (pH, transit time, anaerobiosis), nutritional (ileal effluent composition), and microbial (lumen and mucus-associated microbiota) parameters of this ecosystem and adapted to three dog sizes (i.e., small under 10 kg, medium 10-30 kg, and large over 30 kg). To validate the new model regarding microbiota composition and activities, in vitro fermentations were performed in bioreactors inoculated with stools from 13 dogs (4 small, 5 medium, and 4 large). After a stabilization period, microbiota profiles clearly clustered depending on dog size. Bacteroidota and Firmicutes abundances were positively correlated with dog size both in vitro and in vivo, while opposite trends were observed for Actinobacteria and Proteobacteria. As observed in vivo, microbial activity also increased with dog size in vitro, as evidenced from gas production, short-chain fatty acids, ammonia, and bile acid dehydroxylation. In line with the 3R regulation, CANIM-ARCOL could be a relevant platform to assess bilateral interactions between food and pharma compounds and gut microbiota, capturing inter-individual or breed variabilities. KEY POINTS: • CANIM-ARCOL integrates main canine physicochemical and microbial colonic parameters • Gut microbiota associated to different dog sizes is accurately maintained in vitro • The model can help to move toward personalized approach considering dog body weight.
Collapse
Affiliation(s)
- Charlotte Deschamps
- UMR 454 MEDIS, Université Clermont Auvergne, INRAE, Clermont-Ferrand, France
- Lallemand Animal Nutrition, Blagnac, France
| | - Sylvain Denis
- UMR 454 MEDIS, Université Clermont Auvergne, INRAE, Clermont-Ferrand, France
| | | | - Nathalie Priymenko
- Toxalim (Research Center in Food Toxicology), University of Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31000, Toulouse, France
| | - Sandrine Chalancon
- UMR 454 MEDIS, Université Clermont Auvergne, INRAE, Clermont-Ferrand, France
| | - Jana De Bodt
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | | | - Inma Alvarez-Acero
- Institute of Food Science, Technology and Nutrition, Spanish National Research Council, ICTAN-CSIC), Madrid, Spain
| | | | | | | |
Collapse
|
7
|
Hussein M, Pasqua M, Pereira U, Benzoubir N, Duclos-Vallée JC, Dubart-Kupperschmitt A, Legallais C, Messina A. Microencapsulated Hepatocytes Differentiated from Human Induced Pluripotent Stem Cells: Optimizing 3D Culture for Tissue Engineering Applications. Cells 2023; 12:cells12060865. [PMID: 36980206 PMCID: PMC10047414 DOI: 10.3390/cells12060865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 03/14/2023] Open
Abstract
Liver cell therapy and in vitro models require functional human hepatocytes, the sources of which are considerably limited. Human induced pluripotent stem cells (hiPSCs) represent a promising and unlimited source of differentiated human hepatocytes. However, when obtained in two-dimensional (2D) cultures these hepatocytes are not fully mature and functional. As three-dimensional culture conditions offer advantageous strategies for differentiation, we describe here a combination of three-dimensional (3D) approaches enabling the successful differentiation of functional hepatocytes from hiPSCs by the encapsulation of hiPSC-derived hepatoblasts in alginate beads of preformed aggregates. The resulting encapsulated and differentiated hepatocytes (E-iHep-Orgs) displayed a high level of albumin synthesis associated with the disappearance of α-fetoprotein (AFP) synthesis, thus demonstrating that the E-iHep-Orgs had reached a high level of maturation, similar to that of adult hepatocytes. Gene expression analysis by RT-PCR and immunofluorescence confirmed this maturation. Further functional assessments demonstrated their enzymatic activities, including lactate and ammonia detoxification, as well as biotransformation activities of Phase I and Phase II enzymes. This study provides proof of concept regarding the benefits of combining three-dimensional techniques (guided aggregation and microencapsulation) with liver differentiation protocols as a robust approach to generate mature and functional hepatocytes that offer a permanent and unlimited source of hepatocytes. Based on these encouraging results, our combined conditions to produce mature hepatocytes from hiPSCs could be extended to liver tissue engineering and bioartificial liver (BAL) applications at the human scale for which large biomasses are mandatory.
Collapse
Affiliation(s)
- Marwa Hussein
- UMR_S 1193, INSERM/Université Paris Saclay, F-94800 Villejuif, France
- Fédération Hospitalo-Universitaire (FHU) Hépatinov, F-94800 Villejuif, France
| | - Mattia Pasqua
- Fédération Hospitalo-Universitaire (FHU) Hépatinov, F-94800 Villejuif, France
- UMR CNRS 7338 Biomechanics & Bioengineering, Université de Technologie de Compiègne, Sorbonne Universités, F-60203 Compiegne, France
| | - Ulysse Pereira
- Fédération Hospitalo-Universitaire (FHU) Hépatinov, F-94800 Villejuif, France
- UMR CNRS 7338 Biomechanics & Bioengineering, Université de Technologie de Compiègne, Sorbonne Universités, F-60203 Compiegne, France
| | - Nassima Benzoubir
- UMR_S 1193, INSERM/Université Paris Saclay, F-94800 Villejuif, France
- Fédération Hospitalo-Universitaire (FHU) Hépatinov, F-94800 Villejuif, France
| | - Jean-Charles Duclos-Vallée
- UMR_S 1193, INSERM/Université Paris Saclay, F-94800 Villejuif, France
- Fédération Hospitalo-Universitaire (FHU) Hépatinov, F-94800 Villejuif, France
| | - Anne Dubart-Kupperschmitt
- UMR_S 1193, INSERM/Université Paris Saclay, F-94800 Villejuif, France
- Fédération Hospitalo-Universitaire (FHU) Hépatinov, F-94800 Villejuif, France
| | - Cecile Legallais
- Fédération Hospitalo-Universitaire (FHU) Hépatinov, F-94800 Villejuif, France
- UMR CNRS 7338 Biomechanics & Bioengineering, Université de Technologie de Compiègne, Sorbonne Universités, F-60203 Compiegne, France
- Correspondence: (C.L.); (A.M.)
| | - Antonietta Messina
- UMR_S 1193, INSERM/Université Paris Saclay, F-94800 Villejuif, France
- Fédération Hospitalo-Universitaire (FHU) Hépatinov, F-94800 Villejuif, France
- Correspondence: (C.L.); (A.M.)
| |
Collapse
|
8
|
Fournier E, Ratel J, Denis S, Leveque M, Ruiz P, Mazal C, Amiard F, Edely M, Bezirard V, Gaultier E, Lamas B, Houdeau E, Engel E, Lagarde F, Etienne-Mesmin L, Mercier-Bonin M, Blanquet-Diot S. Exposure to polyethylene microplastics alters immature gut microbiome in an infant in vitro gut model. JOURNAL OF HAZARDOUS MATERIALS 2023; 443:130383. [PMID: 36444070 DOI: 10.1016/j.jhazmat.2022.130383] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 06/16/2023]
Abstract
Infants are characterized by an immaturity of the gut ecosystem and a high exposure to microplastics (MPs) through diet, dust and suckling. However, the bidirectional interactions between MPs and the immature infant intestinal microbiota remain unknown. Our study aims to investigate the impact of chronic exposure to polyethylene (PE) MPs on the gut microbiota and intestinal barrier of infants, using the new Toddler mucosal Artificial Colon coupled with a co-culture of epithelial and mucus-secreting cells. Gut microbiota composition was determined by 16S metabarcoding and microbial activities were evaluated by gas, short chain fatty acid and volatolomics analyses. Gut barrier integrity was assessed via evaluation of intestinal permeability, inflammation and mucus synthesis. Exposure to PE MPs induced gut microbial shifts increasing α-diversity and abundance of potentially harmful pathobionts, such as Dethiosulfovibrionaceae and Enterobacteriaceae. Those changes were associated to butyrate production decrease and major changes in volatile organic compounds profiles. In contrast, no significant impact of PE MPs on the gut barrier, as mediated by microbial metabolites, was reported. For the first time, this study indicates that ingestion of PE MPs can induce perturbations in the gut microbiome of infants. Next step would be to further investigate the potential vector effect of MPs.
Collapse
Affiliation(s)
- Elora Fournier
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, F-63000 Clermont-Ferrand, France; Toxalim, Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, F-31000 Toulouse, France
| | - Jeremy Ratel
- INRAE, UR QuaPA, MASS Team, F-63122 Saint-Genès-Champanelle, France
| | - Sylvain Denis
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, F-63000 Clermont-Ferrand, France
| | - Mathilde Leveque
- Toxalim, Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, F-31000 Toulouse, France
| | - Philippe Ruiz
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, F-63000 Clermont-Ferrand, France
| | - Carine Mazal
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, F-63000 Clermont-Ferrand, France
| | - Frederic Amiard
- Le Mans Université, IMMM UMR-CNRS 6283, Avenue Olivier Messiaen, F-72085 Cedex 9 Le Mans, France
| | - Mathieu Edely
- Le Mans Université, IMMM UMR-CNRS 6283, Avenue Olivier Messiaen, F-72085 Cedex 9 Le Mans, France
| | - Valerie Bezirard
- Toxalim, Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, F-31000 Toulouse, France
| | - Eric Gaultier
- Toxalim, Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, F-31000 Toulouse, France
| | - Bruno Lamas
- Toxalim, Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, F-31000 Toulouse, France
| | - Eric Houdeau
- Toxalim, Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, F-31000 Toulouse, France
| | - Erwan Engel
- INRAE, UR QuaPA, MASS Team, F-63122 Saint-Genès-Champanelle, France
| | - Fabienne Lagarde
- Le Mans Université, IMMM UMR-CNRS 6283, Avenue Olivier Messiaen, F-72085 Cedex 9 Le Mans, France
| | - Lucie Etienne-Mesmin
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, F-63000 Clermont-Ferrand, France
| | - Muriel Mercier-Bonin
- Toxalim, Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, F-31000 Toulouse, France.
| | | |
Collapse
|
9
|
Fournier E, Leveque M, Ruiz P, Ratel J, Durif C, Chalancon S, Amiard F, Edely M, Bezirard V, Gaultier E, Lamas B, Houdeau E, Lagarde F, Engel E, Etienne-Mesmin L, Blanquet-Diot S, Mercier-Bonin M. Microplastics: What happens in the human digestive tract? First evidences in adults using in vitro gut models. JOURNAL OF HAZARDOUS MATERIALS 2023; 442:130010. [PMID: 36182891 DOI: 10.1016/j.jhazmat.2022.130010] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/08/2022] [Accepted: 09/15/2022] [Indexed: 06/16/2023]
Abstract
Microplastics (MPs) are ubiquitous in the environment and humans are inevitably exposed to them. However, the effects of MPs in the human digestive environment are largely unknown. The aim of our study was to investigate the impact of repeated exposure to polyethylene (PE) MPs on the human gut microbiota and intestinal barrier using, under adult conditions, the Mucosal Artificial Colon (M-ARCOL) model, coupled with a co-culture of intestinal epithelial and mucus-secreting cells. The composition of the luminal and mucosal gut microbiota was determined by 16S metabarcoding and microbial activities were characterized by gas, short chain fatty acid, volatolomic and AhR activity analyses. Gut barrier integrity was assessed via intestinal permeability, inflammation and mucin synthesis. First, exposure to PE MPs induced donor-dependent effects. Second, an increase in abundances of potentially harmful pathobionts, Desulfovibrionaceae and Enterobacteriaceae, and a decrease in beneficial bacteria such as Christensenellaceae and Akkermansiaceae were observed. These bacterial shifts were associated with changes in volatile organic compounds profiles, notably characterized by increased indole 3-methyl- production. Finally, no significant impact of PE MPs mediated by changes in gut microbial metabolites was reported on the intestinal barrier. Given these adverse effects of repeated ingestion of PE MPs on the human gut microbiota, studying at-risk populations like infants would be a valuable advance.
Collapse
Affiliation(s)
- Elora Fournier
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, F-63000 Clermont-Ferrand, France; Toxalim, Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, F-31000 Toulouse, France
| | - Mathilde Leveque
- Toxalim, Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, F-31000 Toulouse, France
| | - Philippe Ruiz
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, F-63000 Clermont-Ferrand, France
| | - Jeremy Ratel
- INRAE, UR QuaPA, F-63122 Saint-Genès-Champanelle, France
| | - Claude Durif
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, F-63000 Clermont-Ferrand, France
| | - Sandrine Chalancon
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, F-63000 Clermont-Ferrand, France
| | - Frederic Amiard
- Le Mans Université, IMMM UMR-CNRS 6283, Avenue Olivier Messiaen, F-72085, Le Mans Cedex 9, France
| | - Mathieu Edely
- Le Mans Université, IMMM UMR-CNRS 6283, Avenue Olivier Messiaen, F-72085, Le Mans Cedex 9, France
| | - Valerie Bezirard
- Toxalim, Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, F-31000 Toulouse, France
| | - Eric Gaultier
- Toxalim, Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, F-31000 Toulouse, France
| | - Bruno Lamas
- Toxalim, Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, F-31000 Toulouse, France
| | - Eric Houdeau
- Toxalim, Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, F-31000 Toulouse, France
| | - Fabienne Lagarde
- Le Mans Université, IMMM UMR-CNRS 6283, Avenue Olivier Messiaen, F-72085, Le Mans Cedex 9, France
| | - Erwan Engel
- INRAE, UR QuaPA, F-63122 Saint-Genès-Champanelle, France
| | - Lucie Etienne-Mesmin
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, F-63000 Clermont-Ferrand, France
| | | | - Muriel Mercier-Bonin
- Toxalim, Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, F-31000 Toulouse, France.
| |
Collapse
|
10
|
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
|
11
|
Kalhori D, Zakeri N, Azarpira N, Fanian M, Solati-Hashjin M. Chitosan-Coated-Alginate encapsulation of HepG2 cells for enhanced cellular functions. JOURNAL OF MACROMOLECULAR SCIENCE PART A-PURE AND APPLIED CHEMISTRY 2023. [DOI: 10.1080/10601325.2022.2162414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Dianoosh Kalhori
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Nima Zakeri
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Fanian
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehran Solati-Hashjin
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| |
Collapse
|
12
|
A child is not an adult: development of a new in vitro model of the toddler colon. Appl Microbiol Biotechnol 2022; 106:7315-7336. [PMID: 36202936 DOI: 10.1007/s00253-022-12199-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 11/02/2022]
Abstract
Early life is a critical period where gut ecosystem and functions are being established with significant impact on health. For regulatory, technical, and cost reasons, in vitro gut models can be used as a relevant alternative to in vivo assays. An exhaustive literature review was conducted to adapt the Mucosal Artificial Colon (M-ARCOL) to specific physicochemical (pH, transit time, and nutritional composition of ileal effluents) and microbial parameters from toddlers in the age range of 6 months-3 years, resulting in the Tm-ARCOL. In vitro fermentations were performed to validate this newly developed colonic model compared to in vivo toddler data. Results were also compared to those obtained with the classical adult configuration. Fecal samples from 5 toddlers and 4 adults were used to inoculate bioreactors, and continuous fermentations were performed for 8 days. Gut microbiota structure (lumen and mucus-associated microbiota) and functions (gas and short-chain fatty acids) were monitored. Clearly distinct microbial signatures were obtained between the two in vitro conditions, with lower α-diversity indices and higher abundances of infant-related microbial populations (e.g., Bifidobacteriaceae, Enterobacteriaceae) in toddler versus adult conditions. In accordance with in vivo data, methane was found only in adult bioreactors, while higher percentage of acetate but lower proportions of propionate and butyrate was measured in toddlers compared to adults. This new in vitro model will provide a powerful platform for gut microbiome mechanistic studies in a pediatric context, both in nutritional- (e.g., nutrients, probiotics, prebiotics) and health-related (e.g., drugs, enteric pathogens) studies. KEY POINTS: • Development of a novel in vitro colonic model recapitulating the toddler environment. • Specific toddler versus adult digestive conditions are preserved in vitro. • The new model provides a powerful platform for microbiome mechanistic studies.
Collapse
|
13
|
Xie X, Zhou X, Liu T, Zhong Z, Zhou Q, Iqbal W, Xie Q, Wei C, Zhang X, Chang TMS, Sun P. Direct Differentiation of Human Embryonic Stem Cells to 3D Functional Hepatocyte-like Cells in Alginate Microencapsulation Sphere. Cells 2022; 11:3134. [PMID: 36231094 PMCID: PMC9562699 DOI: 10.3390/cells11193134] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/19/2022] [Accepted: 09/30/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The lack of a stable source of hepatocytes is one of major limitations in hepatocyte transplantation and clinical applications of a bioartificial liver. Human embryonic stem cells (hESCs) with a high degree of self-renewal and totipotency are a potentially limitless source of a variety of cell lineages, including hepatocytes. Many techniques have been developed for effective differentiation of hESCs into functional hepatocyte-like cells. However, the application of hESC-derived hepatocyte-like cells (hESC-Heps) in the clinic has been constrained by the low yield of fully differentiated cells, small-scale culture, difficulties in harvesting, and immunologic graft rejection. To resolve these shortcomings, we developed a novel 3D differentiation system involving alginate-microencapsulated spheres to improve current hepatic differentiation, providing ready-to-use hESC-Heps. METHODS In this study, we used alginate microencapsulation technology to differentiate human embryonic stem cells into hepatocyte-like cells (hESC-Heps). Hepatic markers of hESC-Heps were examined by qPCR and Western blotting, and hepatic functions of hESC-Heps were evaluated by indocyanine-green uptake and release, and ammonia removal. RESULTS The maturity and hepatic functions of the hESC-Heps derived from this 3D system were better than those derived from 2D culture. Hepatocyte-enriched genes, such as HNF4α, AFP, and ALB, were expressed at higher levels in 3D hESC-Heps than in 2D hESC-Heps. 3D hESC-Heps could metabolize indocyanine green and had better capacity to scavenge ammonia. In addition, the 3D sodium alginate hydrogel microspheres could block viral entry into the microspheres, and thus protect hESC-Heps in 3D microspheres from viral infection. CONCLUSION We developed a novel 3D differentiation system for differentiating hESCs into hepatocyte-like cells by using alginate microcapsules.
Collapse
Affiliation(s)
- Xiaoling Xie
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China
- Guangdong Chaozhou Health Vocational College, Chaozhou 521000, China
- The Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Xiaoling Zhou
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China
- The Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Tingdang Liu
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China
- The Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Zhiqian Zhong
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China
- The Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Qi Zhou
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China
- The Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Waqas Iqbal
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China
- The Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Qingdong Xie
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China
- The Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Chiju Wei
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China
| | - Xin Zhang
- Laboratory of Molecular Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Thomas Ming Swi Chang
- Artificial Cells & Organs Research Centre, Departments of Physiology, Medicine & Biomedical Engineering, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Pingnan Sun
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China
- The Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
14
|
Ahmad Raus R, Wan Nawawi WMF, Nasaruddin RR. Alginate and alginate composites for biomedical applications. Asian J Pharm Sci 2021; 16:280-306. [PMID: 34276819 PMCID: PMC8261255 DOI: 10.1016/j.ajps.2020.10.001] [Citation(s) in RCA: 235] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/26/2020] [Accepted: 10/07/2020] [Indexed: 12/22/2022] Open
Abstract
Alginate is an edible heteropolysaccharide that abundantly available in the brown seaweed and the capsule of bacteria such as Azotobacter sp. and Pseudomonas sp. Owing to alginate gel forming capability, it is widely used in food, textile and paper industries; and to a lesser extent in biomedical applications as biomaterial to promote wound healing and tissue regeneration. This is evident from the rising use of alginate-based dressing for heavily exuding wound and their mass availability in the market nowadays. However, alginate also has limitation. When in contact with physiological environment, alginate could gelate into softer structure, consequently limits its potential in the soft tissue regeneration and becomes inappropriate for the usage related to load bearing body parts. To cater this problem, wide range of materials have been added to alginate structure, producing sturdy composite materials. For instance, the incorporation of adhesive peptide and natural polymer or synthetic polymer to alginate moieties creates an improved composite material, which not only possesses better mechanical properties compared to native alginate, but also grants additional healing capability and promote better tissue regeneration. In addition, drug release kinetic and cell viability can be further improved when alginate composite is used as encapsulating agent. In this review, preparation of alginate and alginate composite in various forms (fibre, bead, hydrogel, and 3D-printed matrices) used for biomedical application is described first, followed by the discussion of latest trend related to alginate composite utilization in wound dressing, drug delivery, and tissue engineering applications.
Collapse
Affiliation(s)
- Raha Ahmad Raus
- Department of Biotechnology Engineering, International Islamic University Malaysia, Kuala Lumpur 50728, Malaysia
| | - Wan Mohd Fazli Wan Nawawi
- Department of Biotechnology Engineering, International Islamic University Malaysia, Kuala Lumpur 50728, Malaysia
- Nanoscience and Nanotechnology Research Group (NanoRG), International Islamic University Malaysia, Kuala Lumpur 50728, Malaysia
| | - Ricca Rahman Nasaruddin
- Department of Biotechnology Engineering, International Islamic University Malaysia, Kuala Lumpur 50728, Malaysia
- Nanoscience and Nanotechnology Research Group (NanoRG), International Islamic University Malaysia, Kuala Lumpur 50728, Malaysia
| |
Collapse
|
15
|
Khodabakhshaghdam S, Khoshfetrat AB, Rahbarghazi R. Alginate-chitosan core-shell microcapsule cultures of hepatic cells in a small scale stirred bioreactor: impact of shear forces and microcapsule core composition. J Biol Eng 2021; 15:14. [PMID: 33865460 PMCID: PMC8052835 DOI: 10.1186/s13036-021-00265-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 04/03/2021] [Indexed: 01/20/2023] Open
Abstract
A small scale stirred bioreactor was designed and the effect of different agitation rates (30, 60 and 100 rpm) was investigated on HepG2 cells cultured in alginate-chitosan (AC) core-shell microcapsule in terms of the cell proliferation and liver-specific function. The microencapsulated hepatic cells could proliferate well when they were cultured for 10 days at 30 rpm while the cell-laden microcapsules showed no cell proliferation at 100 rpm in the bioreactor system. Albumin production rate, as an important liver function, increased also 1.8- and 1.5- fold under stirring rate of 30 rpm compared to the static culture and 60 rpm of agitation, respectively. Moreover, In comparison with the static culture, about 1.5-fold increment in urea production was observed at 30 rpm. Similarly, the highest expressions of albumin and P450 genes were found at 30 rpm stirring rate, which were 4.9- and 19.2-fold of the static culture. Addition of collagen to the microcapsule core composition (ACol/C) could improve the cell proliferation and functionality at 60 rpm in comparison with the cell-laden microcapsules without collagen. The study demonstrated the hepatic cell-laden ACol/C microcapsule hydrogel cultured in the small scale stirred bioreactor at low mixing rate has a great potential for mass production of the hepatic cells while maintaining liver-specific functions.
Collapse
Affiliation(s)
- Shahla Khodabakhshaghdam
- Chemical Engineering Faculty, Sahand University of Technology, Tabriz, 51335-1996, Iran
- Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz, 51335-1996, Iran
| | - Ali Baradar Khoshfetrat
- Chemical Engineering Faculty, Sahand University of Technology, Tabriz, 51335-1996, Iran.
- Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz, 51335-1996, Iran.
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
16
|
Kupikowska-Stobba B, Lewińska D. Polymer microcapsules and microbeads as cell carriers for in vivo biomedical applications. Biomater Sci 2020; 8:1536-1574. [PMID: 32110789 DOI: 10.1039/c9bm01337g] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Polymer microcarriers are being extensively explored as cell delivery vehicles in cell-based therapies and hybrid tissue and organ engineering. Spherical microcarriers are of particular interest due to easy fabrication and injectability. They include microbeads, composed of a porous matrix, and microcapsules, where matrix core is additionally covered with a semipermeable membrane. Microcarriers provide cell containment at implantation site and protect the cells from host immunoresponse, degradation and shear stress. Immobilized cells may be genetically altered to release a specific therapeutic product directly at the target site, eliminating side effects of systemic therapies. Cell microcarriers need to fulfil a number of extremely high standards regarding their biocompatibility, cytocompatibility, immunoisolating capacity, transport, mechanical and chemical properties. To obtain cell microcarriers of specified parameters, a wide variety of polymers, both natural and synthetic, and immobilization methods can be applied. Yet so far, only a few approaches based on cell-laden microcarriers have reached clinical trials. The main issue that still impedes progress of these systems towards clinical application is limited cell survival in vivo. Herein, we review polymer biomaterials and methods used for fabrication of cell microcarriers for in vivo biomedical applications. We describe their key limitations and modifications aiming at improvement of microcarrier in vivo performance. We also present the main applications of polymer cell microcarriers in regenerative medicine, pancreatic islet and hepatocyte transplantation and in the treatment of cancer. Lastly, we outline the main challenges in cell microimmobilization for biomedical purposes, the strategies to overcome these issues and potential future improvements in this area.
Collapse
Affiliation(s)
- Barbara Kupikowska-Stobba
- Laboratory of Electrostatic Methods of Bioencapsulation, Department of Biomaterials and Biotechnological Systems, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland.
| | - Dorota Lewińska
- Laboratory of Electrostatic Methods of Bioencapsulation, Department of Biomaterials and Biotechnological Systems, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland.
| |
Collapse
|
17
|
Deschamps C, Fournier E, Uriot O, Lajoie F, Verdier C, Comtet-Marre S, Thomas M, Kapel N, Cherbuy C, Alric M, Almeida M, Etienne-Mesmin L, Blanquet-Diot S. Comparative methods for fecal sample storage to preserve gut microbial structure and function in an in vitro model of the human colon. Appl Microbiol Biotechnol 2020; 104:10233-10247. [PMID: 33085024 DOI: 10.1007/s00253-020-10959-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/28/2020] [Accepted: 10/12/2020] [Indexed: 12/13/2022]
Abstract
In vitro gut models, such as the mucosal artificial colon (M-ARCOL), provide timely and cost-efficient alternatives to in vivo assays allowing mechanistic studies to better understand the role of human microbiome in health and disease. Using such models inoculated with human fecal samples may require a critical step of stool storage. The effects of preservation methods on microbial structure and function in in vitro gut models have been poorly investigated. This study aimed to assess the impact of three commonly used preserving methods, compared with fresh fecal samples used as a control, on the kinetics of lumen and mucus-associated microbiota colonization in the M-ARCOL model. Feces from two healthy donors were frozen 48 h at - 80 °C with or without cryoprotectant (10% glycerol) or lyophilized with maltodextrin and trehalose prior to inoculation of four parallel bioreactors (e.g., fresh stool, raw stool stored at - 80 °C, stool stored at - 80 °C with glycerol and lyophilized stool). Microbiota composition and diversity (qPCR and 16S metabarcoding) as well as metabolic activity (gases and short chain fatty acids) were monitored throughout the fermentation process (9 days). All the preservative treatments allowed the maintaining inside the M-ARCOL of a complex and functional microbiota, but considering stabilization time of microbial profiles and activities (and not technical constraints associated with the supply of frozen material), our results highlighted 48 h freezing at - 80 °C without cryoprotectant as the most efficient method. These results will help scientists to determine the most accurate method for fecal storage prior to inoculation of in vitro gut microbiome models. KEY POINTS: • In vitro ARCOL model reproduces luminal and mucosal human microbiome. • Short-term storage of fecal sample influences microbial stabilization and activity. • 48 h freezing at - 80°C: most efficient method to preserve microbial ecosystem. • Scientific and technical requirements: influencers of preservation method.
Collapse
Affiliation(s)
- Charlotte Deschamps
- Université Clermont Auvergne, INRAe, UMR 454 MEDIS, 28 place Henri Dunant, F-63000, Clermont-Ferrand, France
| | - Elora Fournier
- Université Clermont Auvergne, INRAe, UMR 454 MEDIS, 28 place Henri Dunant, F-63000, Clermont-Ferrand, France
| | - Ophélie Uriot
- Université Clermont Auvergne, INRAe, UMR 454 MEDIS, 28 place Henri Dunant, F-63000, Clermont-Ferrand, France
| | - Frédérique Lajoie
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | - Cécile Verdier
- Université Clermont Auvergne, INRAe, UMR 454 MEDIS, 28 place Henri Dunant, F-63000, Clermont-Ferrand, France
| | - Sophie Comtet-Marre
- Université Clermont Auvergne, INRAe, UMR 454 MEDIS, 28 place Henri Dunant, F-63000, Clermont-Ferrand, France
| | - Muriel Thomas
- Micalis Institute, INRAe, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Nathalie Kapel
- Laboratoire de Coprologie Fonctionnelle, Hôpital Pitié-Salpêtrière, 75013, Paris, France.,INSERM UMR-S1139, Université de Paris, 75006, Paris, France
| | - Claire Cherbuy
- Micalis Institute, INRAe, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Monique Alric
- Université Clermont Auvergne, INRAe, UMR 454 MEDIS, 28 place Henri Dunant, F-63000, Clermont-Ferrand, France
| | - Mathieu Almeida
- MetaGénoPolis, INRAe, Université Paris-Saclay, Jouy-en-Josas, France
| | - Lucie Etienne-Mesmin
- Université Clermont Auvergne, INRAe, UMR 454 MEDIS, 28 place Henri Dunant, F-63000, Clermont-Ferrand, France
| | - Stéphanie Blanquet-Diot
- Université Clermont Auvergne, INRAe, UMR 454 MEDIS, 28 place Henri Dunant, F-63000, Clermont-Ferrand, France.
| |
Collapse
|
18
|
Pasqua M, Pereira U, de Lartigue C, Nicolas J, Vigneron P, Dermigny Q, Legallais C. Preclinical characterization of alginate-poly-L-lysine encapsulated HepaRG for extracorporeal liver supply. Biotechnol Bioeng 2020; 118:453-464. [PMID: 32997339 DOI: 10.1002/bit.27583] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 09/15/2020] [Accepted: 09/26/2020] [Indexed: 12/26/2022]
Abstract
We recently demonstrated that HepaRG cells encapsulated into 1.5% alginate beads are capable of self-assembling into spheroids. They adequately differentiate into hepatocyte-like cells, with hepatic features observed at Day 14 post-encapsulation required for external bioartificial liver applications. Preliminary investigations performed within a bioreactor under shear stress conditions and using a culture medium mimicking acute liver failure (ALF) highlighted the need to reinforce beads with a polymer coating. We demonstrated in a first step that a poly-l-lysine coating improved the mechanical stability, without altering the metabolic activities necessary for bioartificial liver applications (such as ammonia and lactate elimination). In a second step, we tested the optimized biomass in a newly designed perfused dynamic bioreactor, in the presence of the medium model for pathological plasma for 6 h. Performances of the biomass were enhanced as compared to the steady configuration, demonstrating its efficacy in decreasing the typical toxins of ALF. This type of bioreactor is easy to scale up as it relies on the number of micro-encapsulated cells, and could provide an adequate hepatic biomass for liver supply. Its design allows it to be integrated into a hybrid artificial/bioartificial liver setup for further clinical studies regarding its impact on ALF animal models.
Collapse
Affiliation(s)
- Mattia Pasqua
- Université de technologie de Compiègne, CNRS, Laboratoire de Biomécanique et Bioingénierie, Centre de recherche de Royallieu, Compiègne, France
| | - Ulysse Pereira
- Université de technologie de Compiègne, CNRS, Laboratoire de Biomécanique et Bioingénierie, Centre de recherche de Royallieu, Compiègne, France
| | - Claire de Lartigue
- Université de technologie de Compiègne, CNRS, Laboratoire de Biomécanique et Bioingénierie, Centre de recherche de Royallieu, Compiègne, France
| | - Jonathan Nicolas
- Université de technologie de Compiègne, CNRS, Laboratoire de Biomécanique et Bioingénierie, Centre de recherche de Royallieu, Compiègne, France
| | - Pascale Vigneron
- Université de technologie de Compiègne, CNRS, Laboratoire de Biomécanique et Bioingénierie, Centre de recherche de Royallieu, Compiègne, France
| | - Quentin Dermigny
- Université de technologie de Compiègne, CNRS, Laboratoire de Biomécanique et Bioingénierie, Centre de recherche de Royallieu, Compiègne, France
| | - Cécile Legallais
- Université de technologie de Compiègne, CNRS, Laboratoire de Biomécanique et Bioingénierie, Centre de recherche de Royallieu, Compiègne, France.,DHU Hépatinov, Villejuif, France
| |
Collapse
|
19
|
Labour MN, Le Guilcher C, Aid-Launais R, El Samad N, Lanouar S, Simon-Yarza T, Letourneur D. Development of 3D Hepatic Constructs Within Polysaccharide-Based Scaffolds with Tunable Properties. Int J Mol Sci 2020; 21:ijms21103644. [PMID: 32455711 PMCID: PMC7279349 DOI: 10.3390/ijms21103644] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/17/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022] Open
Abstract
Organoids production is a key tool for in vitro studies of physiopathological conditions, drug-induced toxicity assays, and for a potential use in regenerative medicine. Hence, it prompted studies on hepatic organoids and liver regeneration. Numerous attempts to produce hepatic constructs had often limited success due to a lack of viability or functionality. Moreover, most products could not be translated for clinical studies. The aim of this study was to develop functional and viable hepatic constructs using a 3D porous scaffold with an adjustable structure, devoid of any animal component, that could also be used as an in vivo implantable system. We used a combination of pharmaceutical grade pullulan and dextran with different porogen formulations to form crosslinked scaffolds with macroporosity ranging from 30 µm to several hundreds of microns. Polysaccharide scaffolds were easy to prepare and to handle, and allowed confocal observations thanks to their transparency. A simple seeding method allowed a rapid impregnation of the scaffolds with HepG2 cells and a homogeneous cell distribution within the scaffolds. Cells were viable over seven days and form spheroids of various geometries and sizes. Cells in 3D express hepatic markers albumin, HNF4α and CYP3A4, start to polarize and were sensitive to acetaminophen in a concentration-dependant manner. Therefore, this study depicts a proof of concept for organoid production in 3D scaffolds that could be prepared under GMP conditions for reliable drug-induced toxicity studies and for liver tissue engineering.
Collapse
Affiliation(s)
- Marie-Noëlle Labour
- INSERM U1148, LVTS, Université de Paris, X Bichat Hospital, 46 rue H Huchard, F-75018 Paris, France; (M.-N.L.); (C.L.G.); (R.A.-L.); (N.E.S.); (S.L.); (T.S.-Y.)
- INSERM U1148, LVTS, Université Sorbonne Paris Nord, 99 av JB Clément, 93430 Villetaneuse, France
- École Pratique des Hautes Études, Paris Sciences et Lettres (PSL) Research University, 4-14 rue Ferrus, 75014 Paris, France
| | - Camile Le Guilcher
- INSERM U1148, LVTS, Université de Paris, X Bichat Hospital, 46 rue H Huchard, F-75018 Paris, France; (M.-N.L.); (C.L.G.); (R.A.-L.); (N.E.S.); (S.L.); (T.S.-Y.)
- INSERM U1148, LVTS, Université Sorbonne Paris Nord, 99 av JB Clément, 93430 Villetaneuse, France
| | - Rachida Aid-Launais
- INSERM U1148, LVTS, Université de Paris, X Bichat Hospital, 46 rue H Huchard, F-75018 Paris, France; (M.-N.L.); (C.L.G.); (R.A.-L.); (N.E.S.); (S.L.); (T.S.-Y.)
- INSERM UMS-34, FRIM Université de Paris, X Bichat School of Medicine, F-75018 Paris, France
| | - Nour El Samad
- INSERM U1148, LVTS, Université de Paris, X Bichat Hospital, 46 rue H Huchard, F-75018 Paris, France; (M.-N.L.); (C.L.G.); (R.A.-L.); (N.E.S.); (S.L.); (T.S.-Y.)
- INSERM U1148, LVTS, Université Sorbonne Paris Nord, 99 av JB Clément, 93430 Villetaneuse, France
| | - Soraya Lanouar
- INSERM U1148, LVTS, Université de Paris, X Bichat Hospital, 46 rue H Huchard, F-75018 Paris, France; (M.-N.L.); (C.L.G.); (R.A.-L.); (N.E.S.); (S.L.); (T.S.-Y.)
- INSERM U1148, LVTS, Université Sorbonne Paris Nord, 99 av JB Clément, 93430 Villetaneuse, France
| | - Teresa Simon-Yarza
- INSERM U1148, LVTS, Université de Paris, X Bichat Hospital, 46 rue H Huchard, F-75018 Paris, France; (M.-N.L.); (C.L.G.); (R.A.-L.); (N.E.S.); (S.L.); (T.S.-Y.)
- INSERM U1148, LVTS, Université Sorbonne Paris Nord, 99 av JB Clément, 93430 Villetaneuse, France
| | - Didier Letourneur
- INSERM U1148, LVTS, Université de Paris, X Bichat Hospital, 46 rue H Huchard, F-75018 Paris, France; (M.-N.L.); (C.L.G.); (R.A.-L.); (N.E.S.); (S.L.); (T.S.-Y.)
- INSERM U1148, LVTS, Université Sorbonne Paris Nord, 99 av JB Clément, 93430 Villetaneuse, France
- Correspondence:
| |
Collapse
|
20
|
da Silva Morais A, Vieira S, Zhao X, Mao Z, Gao C, Oliveira JM, Reis RL. Advanced Biomaterials and Processing Methods for Liver Regeneration: State-of-the-Art and Future Trends. Adv Healthc Mater 2020; 9:e1901435. [PMID: 31977159 DOI: 10.1002/adhm.201901435] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/13/2019] [Indexed: 12/17/2022]
Abstract
Liver diseases contribute markedly to the global burden of mortality and disease. The limited organ disposal for orthotopic liver transplantation results in a continuing need for alternative strategies. Over the past years, important progress has been made in the field of tissue engineering (TE). Many of the early trials to improve the development of an engineered tissue construct are based on seeding cells onto biomaterial scaffolds. Nowadays, several TE approaches have been developed and are applied to one vital organ: the liver. Essential elements must be considered in liver TE-cells and culturing systems, bioactive agents or growth factors (GF), and biomaterials and processing methods. The potential of hepatocytes, mesenchymal stem cells, and others as cell sources is demonstrated. They need engineered biomaterial-based scaffolds with perfect biocompatibility and bioactivity to support cell proliferation and hepatic differentiation as well as allowing extracellular matrix deposition and vascularization. Moreover, they require a microenvironment provided using conventional or advanced processing technologies in order to supply oxygen, nutrients, and GF. Herein the biomaterials and the conventional and advanced processing technologies, including cell-sheets process, 3D bioprinting, and microfluidic systems, as well as the future trends in these major fields are discussed.
Collapse
Affiliation(s)
- Alain da Silva Morais
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
| | - Sílvia Vieira
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
| | - Xinlian Zhao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University Hangzhou 310027 China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University Hangzhou 310027 China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University Hangzhou 310027 China
| | - Joaquim M. Oliveira
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision MedicineUniversity of Minho 4805‐017 Barco Guimarães Portugal
| | - Rui L. Reis
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision MedicineUniversity of Minho 4805‐017 Barco Guimarães Portugal
| |
Collapse
|
21
|
Pasqua M, Pereira U, Messina A, de Lartigue C, Vigneron P, Dubart-Kupperschmitt A, Legallais C. HepaRG Self-Assembled Spheroids in Alginate Beads Meet the Clinical Needs for Bioartificial Liver. Tissue Eng Part A 2020; 26:613-622. [PMID: 31914890 DOI: 10.1089/ten.tea.2019.0262] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
In liver tissue engineering, cell culture in spheroids is now well recognized to promote the maintenance of hepatic functions. However, the process leading to spheroids formation is time consuming, costly, and not easy to scale-up for further use in human bioartificial liver (BAL) applications. In this study, we encapsulated HepaRG cells (precursors of hepatocyte-like cells) in 1.5% alginate beads without preforming spheroids. Starting from a given hepatic biomass, we analyzed cell differentiation and metabolic performance for further use in a fluidized-bed BAL. We observed that cells self-rearranged as aggregates within the beads and adequately differentiated over time, in the absence of any differentiating factors classically used. On day 14 postencapsulation, cells displayed a wide range of hepatic features necessary for the treatment of a patient in acute liver failure. These activities include albumin synthesis, ammonia and lactate detoxification, and the efficacy of the enzymes involved in the xenobiotic metabolism (such as CYP1A1/2). Impact statement It has been recognized that culturing cells in spheroids (SPHs) is advantageous as they better reproduce the three-dimensional physiological microenvironment. This approach can be exploited in bioartificial liver applications, where obtaining a functional hepatic biomass is the major challenge. Our study describes an original method for culturing hepatic cells in alginate beads that makes possible the autonomous formation of SPHs after 3 days of culture. In turn, the cells differentiate adequately and display a wide range of hepatic features. They are also capable of treating a pathological plasma model. Finally, this setup can easily be scaled-up to treat acute liver failure.
Collapse
Affiliation(s)
- Mattia Pasqua
- UMR CNRS 7338 Biomechanics & Bioengineering, Université de Technologie de Compiègne, Alliance Sorbonne Université, Compiègne, France
| | - Ulysse Pereira
- UMR CNRS 7338 Biomechanics & Bioengineering, Université de Technologie de Compiègne, Alliance Sorbonne Université, Compiègne, France
| | - Antonietta Messina
- DHU Hépatinov, Villejuif, France.,UMR_S1193 Inserm/Paris-Saclay University, Villejuif, France
| | - Claire de Lartigue
- UMR CNRS 7338 Biomechanics & Bioengineering, Université de Technologie de Compiègne, Alliance Sorbonne Université, Compiègne, France
| | - Pascale Vigneron
- UMR CNRS 7338 Biomechanics & Bioengineering, Université de Technologie de Compiègne, Alliance Sorbonne Université, Compiègne, France
| | | | - Cecile Legallais
- UMR CNRS 7338 Biomechanics & Bioengineering, Université de Technologie de Compiègne, Alliance Sorbonne Université, Compiègne, France.,DHU Hépatinov, Villejuif, France
| |
Collapse
|
22
|
Khodabakhsh Aghdam S, Khoshfetrat AB, Rahbarghazi R, Jafarizadeh-Malmiri H, Khaksar M. Collagen modulates functional activity of hepatic cells inside alginate-galactosylated chitosan hydrogel microcapsules. Int J Biol Macromol 2019; 156:1270-1278. [PMID: 31760032 DOI: 10.1016/j.ijbiomac.2019.11.164] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 11/12/2019] [Accepted: 11/19/2019] [Indexed: 02/07/2023]
Abstract
To provide comparable hepatic tissue microenvironment and induce functional behavior for hepatocytes, galctosylated-chitosan (GC) as well as collagen (Col) was added to alginate microcapsule coated with extra layer of chitosan. Four different hydrogel groups of alginate/chitosan (AC); alginate-galactosylated chitosan/chitosan (AGC/C); alginate-collagen/chitosan (ACol/C); and alginate-galactosylated chitosan-collagen/chitosan (AGCCol/C) were prepared and characterized for physical properties such as porosity, swelling, degradation rate, and stiffness. Introduction of GC as well as Col to alginate regulated significantly the physical properties of the resultant hydrogels. GC addition decreased dramatically swelling, degradation, pore size and mechanical properties of the resultant hydrogel. However, the influence of GC on the physical properties in the presence of Col (AGCCol/A) was in a reverse manner, as compared to the AGC/C hydrogel. The AGCCol/C microenvironment also promoted proliferation of microencapsulated HepG2 cells, as a model of hepatocyte, compared to the control-matched groups. Biochemical analysis after 10 days revealed a superior effect of AGCCol/C on the secretion of albumin and urea compared to other groups (P < 0.05). These features were coincided with the mRNA up-regulation of P450 and albumin in the AGCCol/C groups compared to the AGC/C and ACol/C groups (P < 0.05). The study demonstrated that enrichment of alginate-based hydrogels with Col and GC could be touted as an appropriate 3D platform for modular hepatic tissue engineering.
Collapse
Affiliation(s)
- Shahla Khodabakhsh Aghdam
- Chemical Engineering Faculty, Sahand University of Technology, Tabriz 51335-1996, Iran; Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz 51335-1996, Iran
| | - Ali Baradar Khoshfetrat
- Chemical Engineering Faculty, Sahand University of Technology, Tabriz 51335-1996, Iran; Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz 51335-1996, Iran.
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Majid Khaksar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
23
|
Talovic M, Patel K, Schwartz M, Madsen J, Garg K. Decellularized extracellular matrix gelloids support mesenchymal stem cell growth and function in vitro. J Tissue Eng Regen Med 2019; 13:1830-1842. [PMID: 31306568 DOI: 10.1002/term.2933] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/10/2019] [Accepted: 07/03/2019] [Indexed: 12/15/2022]
Abstract
Volumetric muscle loss (VML) injuries are irrecoverable due to a significant loss of regenerative elements, persistent inflammation, extensive fibrosis, and functional impairment. When used in isolation, previous stem cell and biomaterial-based therapies have failed to regenerate skeletal muscle at clinically relevant levels. The extracellular matrix (ECM) microenvironment is crucial for the viability, stemness, and differentiation of stem cells. Decellularized-ECM (D-ECM) scaffolds are at the forefront of ongoing research to develop a viable therapy for VML. Due to the retention of key ECM components, D-ECM scaffolds provide an excellent substrate for the adhesion and migration of several cell types. Mesenchymal stem cells (MSCs) possess regenerative and immunomodulatory properties and are currently under investigation in clinical trials for a wide range of medical conditions. However, a major limitation to the use of MSCs in clinical applications is their poor viability at the site of transplantation. In this study, we have fabricated spherical scaffolds composed of gelatin and skeletal muscle D-ECM for the adhesion and delivery of MSCs to the site of VML injury. These spherical scaffolds termed "gelloids" supported MSC survival, expansion, trophic factor secretion, immunomodulation, and myogenic protein expression in vitro. Future studies would determine the therapeutic efficacy of this approach in a murine model of VML injury.
Collapse
Affiliation(s)
- Muhamed Talovic
- Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, MO
| | - Krishna Patel
- Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, MO
| | - Mark Schwartz
- Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, MO
| | - Josh Madsen
- Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, MO
| | - Koyal Garg
- Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, MO
| |
Collapse
|
24
|
Ye S, Boeter JWB, Penning LC, Spee B, Schneeberger K. Hydrogels for Liver Tissue Engineering. Bioengineering (Basel) 2019; 6:E59. [PMID: 31284412 PMCID: PMC6784004 DOI: 10.3390/bioengineering6030059] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 06/28/2019] [Accepted: 07/03/2019] [Indexed: 12/14/2022] Open
Abstract
Bioengineered livers are promising in vitro models for drug testing, toxicological studies, and as disease models, and might in the future be an alternative for donor organs to treat end-stage liver diseases. Liver tissue engineering (LTE) aims to construct liver models that are physiologically relevant. To make bioengineered livers, the two most important ingredients are hepatic cells and supportive materials such as hydrogels. In the past decades, dozens of hydrogels have been developed to act as supportive materials, and some have been used for in vitro models and formed functional liver constructs. However, currently none of the used hydrogels are suitable for in vivo transplantation. Here, the histology of the human liver and its relationship with LTE is introduced. After that, significant characteristics of hydrogels are described focusing on LTE. Then, both natural and synthetic materials utilized in hydrogels for LTE are reviewed individually. Finally, a conclusion is drawn on a comparison of the different hydrogels and their characteristics and ideal hydrogels are proposed to promote LTE.
Collapse
Affiliation(s)
- Shicheng Ye
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Jochem W B Boeter
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Louis C Penning
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Bart Spee
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Kerstin Schneeberger
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands.
| |
Collapse
|
25
|
Injectable cell-encapsulating composite alginate-collagen platform with inducible termination switch for safer ocular drug delivery. Biomaterials 2019; 201:53-67. [DOI: 10.1016/j.biomaterials.2019.01.032] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 12/27/2018] [Accepted: 01/20/2019] [Indexed: 12/18/2022]
|
26
|
Application of a cyanobacterial extracellular polymeric substance in the microencapsulation of vitamin B12. POWDER TECHNOL 2019. [DOI: 10.1016/j.powtec.2018.11.079] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
27
|
Moroni L, Burdick JA, Highley C, Lee SJ, Morimoto Y, Takeuchi S, Yoo JJ. Biofabrication strategies for 3D in vitro models and regenerative medicine. NATURE REVIEWS. MATERIALS 2018; 3:21-37. [PMID: 31223488 PMCID: PMC6586020 DOI: 10.1038/s41578-018-0006-y] [Citation(s) in RCA: 411] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Organs are complex systems composed of different cells, proteins and signalling molecules that are arranged in a highly ordered structure to orchestrate a myriad of functions in our body. Biofabrication strategies can be applied to engineer 3D tissue models in vitro by mimicking the structure and function of native tissue through the precise deposition and assembly of materials and cells. This approach allows the spatiotemporal control over cell-cell and cell-extracellular matrix communication and thus the recreation of tissue-like structures. In this Review, we examine biofabrication strategies for the construction of functional tissue replacements and organ models, focusing on the development of biomaterials, such as supramolecular and photosensitive materials, that can be processed using biofabrication techniques. We highlight bioprinted and bioassembled tissue models and survey biofabrication techniques for their potential to recreate complex tissue properties, such as shape, vasculature and specific functionalities. Finally, we discuss challenges, such as scalability and the foreign body response, and opportunities in the field and provide an outlook to the future of biofabrication in regenerative medicine.
Collapse
Affiliation(s)
- Lorenzo Moroni
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Complex Tissue Regeneration, Maastricht University, Maastricht, Netherlands
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher Highley
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Yuya Morimoto
- Institute of Industrial Science, University of Tokyo, Tokyo, Japan
| | - Shoji Takeuchi
- Institute of Industrial Science, University of Tokyo, Tokyo, Japan
| | - James J. Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
28
|
Beneficial Effects of Human Mesenchymal Stromal Cells on Porcine Hepatocyte Viability and Albumin Secretion. J Immunol Res 2018; 2018:1078547. [PMID: 29577046 PMCID: PMC5822000 DOI: 10.1155/2018/1078547] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 10/18/2017] [Accepted: 11/01/2017] [Indexed: 12/30/2022] Open
Abstract
Porcine hepatocytes transplanted during acute liver failure might support metabolic functions until the diseased liver recovers its function. Here, we isolated high numbers of viable pig hepatocytes and evaluated hepatocyte functionality after encapsulation. We further investigated whether coculture and coencapsulation of hepatocytes with human multipotent mesenchymal stromal cells (MSC) are beneficial on hepatocyte function. Livers from 10 kg pigs (n = 9) were harvested, and hepatocytes were isolated from liver suspensions for microencapsulation using alginate and poly(ethylene-glycol)- (PEG-) grafted alginate hydrogels, either alone or in combination with MSC. Viability, albumin secretion, and diazepam catabolism of hepatocytes were measured for one week. 9.2 ± 3.6 × 109 hepatocytes with 95.2 ± 3.1% viability were obtained after isolation. At day 3, free hepatocytes displayed 99% viability, whereas microencapsulation in alginate and PEG-grafted alginate decreased viability to 62% and 48%, respectively. Albumin secretion and diazepam catabolism occurred in free and microencapsulated hepatocytes. Coencapsulation of hepatocytes with MSC significantly improved viability and albumin secretion at days 4 and 8 (p < 0.05). Coculture with MSC significantly increased and prolonged albumin secretion. In conclusion, we established a protocol for isolation and microencapsulation of high numbers of viable pig hepatocytes and demonstrated that the presence of MSC is beneficial for the viability and function of porcine hepatocytes.
Collapse
|
29
|
Lee H, Woo HM, Kang BJ. Impact of collagen-alginate composition from microbead morphological properties to microencapsulated canine adipose tissue-derived mesenchymal stem cell activities. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2017; 29:1042-1052. [PMID: 29082833 DOI: 10.1080/09205063.2017.1399002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The purpose of this study was to identify the effect of collagen-alginate composition on the size and shape of microbeads and the proliferation and osteogenic properties of microencapsulated canine adipose-derived mesenchymal stem cells (ASCs) in vitro. Canine ASCs were microencapsulated in mixtures of various collagen-alginate compositions using a vibrational technologic encapsulator. The size and shape of the resultant microbeads were measured using a light field microscope and the viability of the microencapsulated canine ASCs was evaluated using a live/dead viability/cytotoxicity kit. Proliferation and osteogenic potentials of microencapsulated canine ASCs were evaluated using an alamarBlue proliferation assay and an alkaline phosphatase assay, respectively. As the collagen ratio increased, the size and size variation of microbeads increased and the shape of microbeads became more irregular. Nonetheless, homogeneous microbeads were created with no significant difference in size and shape, in the range of 0.75% alginate mixed with 0.099% collagen solution in 1.2% alginate solution. There were no significant differences in viability of the ASCs in the various collagen-alginate compositions. Both proliferation and osteogenic properties, in vitro, increased with increasing collagen ratio. Microencapsulation of canine ASCs with appropriate collagen-alginate composition increases cell proliferation and osteogenic properties, in vitro, without significant effects on the shape and size of microbeads and cell viability. Microencapsulation with adequate collagen-alginate composition may produce injectable microbeads that could enhance the therapeutic efficacy of stem cells.
Collapse
Affiliation(s)
- Hyunkyu Lee
- a Department of Veterinary Surgery, College of Veterinary Medicine and Institute of Veterinary Science , Kangwon National University , Chuncheon , Republic of Korea
| | - Heung-Myong Woo
- a Department of Veterinary Surgery, College of Veterinary Medicine and Institute of Veterinary Science , Kangwon National University , Chuncheon , Republic of Korea
| | - Byung-Jae Kang
- a Department of Veterinary Surgery, College of Veterinary Medicine and Institute of Veterinary Science , Kangwon National University , Chuncheon , Republic of Korea
| |
Collapse
|
30
|
Bajpai M, Shukla P, Bajpai SK. Enhancement in the stability of alginate gels prepared with mixed solution of divalent ions using a diffusion through dialysis tube (DTDT) approach. JOURNAL OF MACROMOLECULAR SCIENCE PART A-PURE AND APPLIED CHEMISTRY 2017. [DOI: 10.1080/10601325.2017.1294452] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
31
|
Koh E, Jung YC, Woo HM, Kang BJ. Injectable alginate-microencapsulated canine adipose tissue-derived mesenchymal stem cells for enhanced viable cell retention. J Vet Med Sci 2017; 79:492-501. [PMID: 28070061 PMCID: PMC5383167 DOI: 10.1292/jvms.16-0456] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The purpose of this study was to establish an optimized protocol for the production of alginate-encapsulated canine adipose-derived mesenchymal stem cells
(cASCs) and evaluate their suitability for clinical use, including viability, proliferation and in vivo cell retention. Alginate microbeads
were formed by vibrational technology and the production of injectable microbeads was performed using various parameters with standard methodology. Microbead
toxicity was tested in an animal model. Encapsulated cASCs were evaluated for viability and proliferation in vitro. HEK-293 cells, with or
without microencapsulation, were injected into the subcutaneous tissue of mice and were tracked using in vivo bioluminescent imaging to
evaluate the retention of transplanted cells. The optimized injectable microbeads were of uniform size and approximately 250 µm in diameter.
There was no strong evidence of in vivo toxicity for the alginate beads. The cells remained viable after encapsulation, and there was evidence
of in vitro proliferation within the microcapsules. In vivo bioluminescent imaging showed that alginate encapsulation improved
the retention of transplanted cells and the encapsulated cells remained viable in vivo for 7 days. Encapsulation enhances the retention of
viable cells in vivo and might represent a potential strategy to increase the therapeutic potency and efficacy of stem cells.
Collapse
Affiliation(s)
- Eunji Koh
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | | | | | | |
Collapse
|
32
|
Ramos PE, Cerqueira MA, Cook MT, Bourbon AI, Khutoryanskiy VV, Charalampoulos D, Teixeira JA, Vicente AA. Development of an immobilization system for in situ micronutrients release. Food Res Int 2016; 90:121-132. [DOI: 10.1016/j.foodres.2016.10.050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 10/27/2016] [Accepted: 10/29/2016] [Indexed: 01/08/2023]
|
33
|
Wong FSY, Wong CCH, Chan BP, Lo ACY. Sustained Delivery of Bioactive GDNF from Collagen and Alginate-Based Cell-Encapsulating Gel Promoted Photoreceptor Survival in an Inherited Retinal Degeneration Model. PLoS One 2016; 11:e0159342. [PMID: 27441692 PMCID: PMC4956057 DOI: 10.1371/journal.pone.0159342] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 06/30/2016] [Indexed: 11/29/2022] Open
Abstract
Encapsulated-cell therapy (ECT) is an attractive approach for continuously delivering freshly synthesized therapeutics to treat sight-threatening posterior eye diseases, circumventing repeated invasive intravitreal injections and improving local drug availability clinically. Composite collagen-alginate (CAC) scaffold contains an interpenetrating network that integrates the physical and biological merits of its constituents, including biocompatibility, mild gelling properties and availability. However, CAC ECT properties and performance in the eye are not well-understood. Previously, we reported a cultured 3D CAC system that supported the growth of GDNF-secreting HEK293 cells with sustainable GDNF delivery. Here, the system was further developed into an intravitreally injectable gel with 1x104 or 2x105 cells encapsulated in 2mg/ml type I collagen and 1% alginate. Gels with lower alginate concentration yielded higher initial cell viability but faster spheroid formation while increasing initial cell density encouraged cell growth. Continuous GDNF delivery was detected in culture and in healthy rat eyes for at least 14 days. The gels were well-tolerated with no host tissue attachment and contained living cell colonies. Most importantly, gel-implanted in dystrophic Royal College of Surgeons rat eyes for 28 days retained photoreceptors while those containing higher initial cell number yielded better photoreceptor survival. CAC ECT gels offers flexible system design and is a potential treatment option for posterior eye diseases.
Collapse
Affiliation(s)
- Francisca S. Y. Wong
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Calvin C. H. Wong
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Barbara P. Chan
- Tissue Engineering Laboratory, Department of Mechanical Engineering, Faculty of Engineering, The University of Hong Kong, Hong Kong, China
| | - Amy C. Y. Lo
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Research Centre of Heart, Brain, Hormone and Healthy Aging, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- * E-mail:
| |
Collapse
|
34
|
Ibarra V, Appel AA, Anastasio MA, Opara EC, Brey EM. This paper is a winner in the Undergraduate category for the SFB awards: Evaluation of the tissue response to alginate encapsulated islets in an omentum pouch model. J Biomed Mater Res A 2016; 104:1581-90. [PMID: 27144389 PMCID: PMC5897127 DOI: 10.1002/jbm.a.35769] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 04/02/2016] [Accepted: 04/29/2016] [Indexed: 12/21/2022]
Abstract
Islet transplantation is currently in clinical use as a treatment for type I diabetes, but donor shortages and long-term immunosuppression limit broad application. Alginate microcapsules coated with poly-l-ornithine can be used to encapsulate islets in an environment that allows diffusion of glucose, insulin, nutrients, and waste products while inhibiting cells and antibodies. While clinical trials are ongoing using islets encapsulated in alginate microbeads, there are concerns in regards to long-term stability. Evaluation of the local tissue response following implantation provides insight into the underlying mechanisms contributing to biomaterial failure, which can be used to the design of new material strategies. Macrophages play an important role in driving the response. In this study, the stability of alginate microbeads coated with PLO containing islets transplanted in the omentum pouch model was investigated. Biomaterial structure and the inflammatory response were characterized by X-ray phase contrast (XPC) μCT imaging, histology, and immunostaining. XPC allowed evaluation of microbead 3D structure and identification of failed and stable microbeads. A robust inflammatory response characterized by high cell density and the presence of pro-inflammatory macrophages was found around the failed grafts. The results obtained provide insight into the local tissue response and possible failure mechanisms for alginate microbeads. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 1581-1590, 2016.
Collapse
Affiliation(s)
- Veronica Ibarra
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois
| | - Alyssa A Appel
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois
| | - Mark A Anastasio
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri
| | - Emmanuel C Opara
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, Illinois
| | - Eric M Brey
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois
- Research Services, Edward Hines Jr. VA Hospital, Ines, IL
| |
Collapse
|
35
|
Mahou R, Passemard S, Carvello M, Petrelli A, Noverraz F, Gerber-Lemaire S, Wandrey C. Contribution of polymeric materials to progress in xenotransplantation of microencapsulated cells: a review. Xenotransplantation 2016; 23:179-201. [PMID: 27250036 DOI: 10.1111/xen.12240] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/09/2016] [Indexed: 12/13/2022]
Abstract
Cell microencapsulation and subsequent transplantation of the microencapsulated cells require multidisciplinary approaches. Physical, chemical, biological, engineering, and medical expertise has to be combined. Several natural and synthetic polymeric materials and different technologies have been reported for the preparation of hydrogels, which are suitable to protect cells by microencapsulation. However, owing to the frequent lack of adequate characterization of the hydrogels and their components as well as incomplete description of the technology, many results of in vitro and in vivo studies appear contradictory or cannot reliably be reproduced. This review addresses the state of the art in cell microencapsulation with special focus on microencapsulated cells intended for xenotransplantation cell therapies. The choice of materials, the design and fabrication of the microspheres, as well as the conditions to be met during the cell microencapsulation process, are summarized and discussed prior to presenting research results of in vitro and in vivo studies. Overall, this review will serve to sensitize medically educated specialists for materials and technological aspects of cell microencapsulation.
Collapse
Affiliation(s)
- Redouan Mahou
- Interfaculty Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Solène Passemard
- Interfaculty Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Michele Carvello
- Department of Surgery, San Raffaele Scientific Institute, Milan, Italy
| | | | - François Noverraz
- Interfaculty Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Sandrine Gerber-Lemaire
- Interfaculty Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Christine Wandrey
- Interfaculty Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
36
|
Bygd HC, Bratlie KM. The effect of chemically modified alginates on macrophage phenotype and biomolecule transport. J Biomed Mater Res A 2016; 104:1707-19. [DOI: 10.1002/jbm.a.35700] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 01/29/2016] [Accepted: 02/23/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Hannah C. Bygd
- Department of Materials Science and EngineeringIowa State UniversityAmes Iowa50011
| | - Kaitlin M. Bratlie
- Department of Materials Science and EngineeringIowa State UniversityAmes Iowa50011
- Department of Chemical and Biological EngineeringIowa State UniversityAmes Iowa50011
- Division of Materials Science & EngineeringAmes National LaboratoryAmes Iowa50011
| |
Collapse
|
37
|
Cell-laden Polymeric Microspheres for Biomedical Applications. Trends Biotechnol 2015; 33:653-666. [DOI: 10.1016/j.tibtech.2015.09.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 08/10/2015] [Accepted: 09/08/2015] [Indexed: 01/16/2023]
|
38
|
Shen C, Zhang G, Wang Q, Meng Q. Fabrication of Collagen Gel Hollow Fibers by Covalent Cross-Linking for Construction of Bioengineering Renal Tubules. ACS APPLIED MATERIALS & INTERFACES 2015; 7:19789-19797. [PMID: 26280545 DOI: 10.1021/acsami.5b05809] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Collagen, the most used natural biomacromolecule, has been extensively utilized to make scaffolds for cell cultures in tissue engineering, but has never been fabricated into the configuration of a hollow fiber (HF) for cell culture due to its poor mechanical properties. In this study, renal tubular cell-laden collagen hollow fiber (Col HF) was fabricated by dissolving sacrificial Ca-alginate cores from collagen shells strengthened by carbodiimide cross-linking. The inner/outer diameters of the Col HF were precisely controlled by the flow rates of core alginate/shell collagen solution in the microfluidic device. As found, the renal tubular cells self-assembled into renal tubules with diameters of 50-200 μm post to the culture in Col HF for 10 days. According to the 3D reconstructed confocal images or HE staining, the renal cells appeared as a tight tubular monolayer on the Col HF inner surface, sustaining more 3D cell morphology than the cell layer on the 2D flat collagen gel surface. Moreover, compared with the cultures in either a Transwell or polymer HF membrane, the renal tubules in Col HF exhibited at least 1-fold higher activity on brush border enzymes of alkaline phosphatase and γ-glutamyltransferase, consistent with their gene expressions. The enhancement occurred similarly on multidrug resistance protein 2 and glucose uptake. Such bioengineered renal tubules in Col HF will present great potential as alternatives to synthetic HF in both clinical use and pharmaceutical investigation.
Collapse
Affiliation(s)
- Chong Shen
- Department of Chemical and Biological Engineering, Zhejiang University , Hangzhou 310027, China
| | - Guoliang Zhang
- College of Chemical Engineering and Materials Science, Zhejiang University of Technology , Hangzhou 310023, China
| | - Qichen Wang
- Department of Chemical Engineering and Materials Science, Stevens Institute of Technology , Hoboken, New Jersey 07030, United States
| | - Qin Meng
- Department of Chemical and Biological Engineering, Zhejiang University , Hangzhou 310027, China
| |
Collapse
|
39
|
Khalid N, Kobayashi I, Neves MA, Uemura K, Nakajima M, Nabetani H. Preparation of monodisperse aqueous microspheres containing high concentration ofl-ascorbic acid by microchannel emulsification. J Microencapsul 2015; 32:570-7. [DOI: 10.3109/02652048.2015.1065919] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
40
|
Wang K, Yu LY, Jiang LY, Wang HB, Wang CY, Luo Y. The paracrine effects of adipose-derived stem cells on neovascularization and biocompatibility of a macroencapsulation device. Acta Biomater 2015; 15:65-76. [PMID: 25575852 DOI: 10.1016/j.actbio.2014.12.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 12/11/2014] [Accepted: 12/24/2014] [Indexed: 12/12/2022]
Abstract
The foreign-body response to biomaterials compromises the performance of many biomedical devices by severe fibrosis and limited neovascularization. Mesenchymal stem cells are known to secrete cytokines for treating inflammatory conditions. In this study, we aim to investigate whether the paracrine products of adipose-derived mesenchymal stem cells (ADSCs) can affect the microenvironment of biomaterials and improve tissue responses to biomaterial implants. A model system was built by loading ADSC spheroids into a macroencapsulation device composed of polytetrafluoroethylene (PTFE) filtration membranes. Soluble ADSC factors that diffused out of the device in vitro promoted the angiogenetic activity of endothelial cells and affected the secretion pattern of macrophages. In vivo study was carried out by subcutaneously embedding blank or ADSC-laden devices in rats. Following a 4 week implantation, the ADSC-laden devices were better vascularized and induced significantly less fibrotic tissue formation in comparison to the non-cellular controls. This study may facilitate our understanding of foreign-body responses and suggest new ways to improve the tissue reaction of biomedical devices for cell-based therapy.
Collapse
|
41
|
Injectable PEGylated fibrinogen cell-laden microparticles made with a continuous solvent- and oil-free preparation method. Acta Biomater 2015; 13:78-87. [PMID: 25462849 DOI: 10.1016/j.actbio.2014.11.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Revised: 10/23/2014] [Accepted: 11/05/2014] [Indexed: 12/12/2022]
Abstract
A new methodology is reported for the continuous, solvent- and oil-free production of photopolymerizable microparticles containing encapsulated human dermal fibroblasts. A precursor solution of cells in photoreactive poly(ethylene glycol) (PEG)-fibrinogen (PF) polymer was transported through a transparent injector exposed to light irradiation before being atomized in a jet-in-air nozzle. Shear rheometry data revealed the crosslinking kinetics of the PF/cell solution, which was then used to determine the amount of irradiation required to partially polymerize the mixture just prior to atomization. The partially polymerized drops of PF/cells fell into a gelation bath for further crosslinking until fully polymerized hydrogel microparticles were formed. As the drops of solution exited the air-in-jet nozzle, their viscosity was designed to be sufficiently high so as to prevent rapid mixing and/or dilution in the gelation bath, but without undergoing complete gelation in the nozzle. Several parameters of this system were varied to control the size and polydispersity of the microparticles, including the cell density, the flow rate and the air pressure in the nozzle. The system was capable of producing cell-laden microparticles with an average diameter of between 88.1 to 347.1 μm, and a dispersity of between 1.1 and 2.4, depending on the parameters chosen. Varying the precursor flow rate and/or cell density was beneficial in controlling the size and polydispersity of the microparticles; all microparticles exhibited very high cell viability, which was not affected by these parameters. In conclusion, this dropwise photopolymerization methodology for preparing cell-laden microparticles is an attractive alternative to existing techniques that use harsh solvents/oils and offer limited control over particle size and polydispersity.
Collapse
|
42
|
Tran NM, Dufresne M, Helle F, Hoffmann TW, François C, Brochot E, Paullier P, Legallais C, Duverlie G, Castelain S. Alginate hydrogel protects encapsulated hepatic HuH-7 cells against hepatitis C virus and other viral infections. PLoS One 2014; 9:e109969. [PMID: 25310111 PMCID: PMC4195705 DOI: 10.1371/journal.pone.0109969] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 09/11/2014] [Indexed: 12/25/2022] Open
Abstract
Cell microencapsulation in alginate hydrogel has shown interesting applications in regenerative medicine and the biomedical field through implantation of encapsulated tissue or for bioartificial organ development. Although alginate solution is known to have low antiviral activity, the same property regarding alginate gel has not yet been studied. The aim of this work is to investigate the potential protective effect of alginate encapsulation against hepatitis C virus (HCV) infection for a hepatic cell line (HuH-7) normally permissive to the virus. Our results showed that alginate hydrogel protects HuH-7 cells against HCV when the supernatant was loaded with HCV. In addition, alginate hydrogel blocked HCV particle release out of the beads when the HuH-7 cells were previously infected and encapsulated. There was evidence of interaction between the molecules of alginate hydrogel and HCV, which was dose- and incubation time-dependent. The protective efficiency of alginate hydrogel towards HCV infection was confirmed against a variety of viruses, whether or not they were enveloped. This promising interaction between an alginate matrix and viruses, whose chemical mechanisms are discussed, is of great interest for further medical therapeutic applications based on tissue engineering.
Collapse
Affiliation(s)
- Nhu-Mai Tran
- UMR CNRS 7338 Biomechanics and Bioingineering, University of Technology, Compiègne, France
| | - Murielle Dufresne
- UMR CNRS 7338 Biomechanics and Bioingineering, University of Technology, Compiègne, France
- * E-mail:
| | - François Helle
- EA4294 Department of Fundamental and Clinical Virology, University of Picardie Jules Verne, Amiens, France
| | - Thomas Walter Hoffmann
- EA4294 Department of Fundamental and Clinical Virology, University of Picardie Jules Verne, Amiens, France
| | - Catherine François
- EA4294 Department of Fundamental and Clinical Virology, University of Picardie Jules Verne, Amiens, France
| | - Etienne Brochot
- EA4294 Department of Fundamental and Clinical Virology, University of Picardie Jules Verne, Amiens, France
| | - Patrick Paullier
- UMR CNRS 7338 Biomechanics and Bioingineering, University of Technology, Compiègne, France
| | - Cécile Legallais
- UMR CNRS 7338 Biomechanics and Bioingineering, University of Technology, Compiègne, France
| | - Gilles Duverlie
- EA4294 Department of Fundamental and Clinical Virology, University of Picardie Jules Verne, Amiens, France
| | - Sandrine Castelain
- EA4294 Department of Fundamental and Clinical Virology, University of Picardie Jules Verne, Amiens, France
| |
Collapse
|
43
|
Díaz-Barrera A, Martínez F, Guevara Pezoa F, Acevedo F. Evaluation of gene expression and alginate production in response to oxygen transfer in continuous culture of Azotobacter vinelandii. PLoS One 2014; 9:e105993. [PMID: 25162704 PMCID: PMC4146552 DOI: 10.1371/journal.pone.0105993] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 07/25/2014] [Indexed: 11/18/2022] Open
Abstract
Alginates are polysaccharides used as food additives and encapsulation agents in biotechnology, and their functional properties depend on its molecular weight. In this study, different steady-states in continuous cultures of A. vinelandii were established to determine the effect of the dilution rate (D) and the agitation rate on alginate production and expression of genes involved in alginate polymerization and depolymerization. Both, the agitation and dilution rates, determined the partitioning of the carbon utilization from sucrose into alginate and CO2 under oxygen-limiting conditions. A low D (0.07 h−1) and 500 rpm resulted in the highest carbon utilization into alginate (25%). Quantitative real-time polymerase chain reaction was used to determine the transcription level of six genes involved in alginate polymerization and depolymerization. In chemostat cultures at 0.07 h−1, the gene expression was affected by changes in the agitation rate. By increasing the agitation rate from 400 to 600 rpm, the algE7 gene expression decreased tenfold, whereas alyA1, algL and alyA2 gene expression increased between 1.5 and 2.8 times under similar conditions evaluated. Chemostat at 0.07 h−1 showed a highest alginate molecular weight (580 kDa) at 500 rpm whereas similar molecular weights (480 kDa) were obtained at 400 and 600 rpm. The highest molecular weight was not explained by changes in the expression of alg8 and alg44 (genes involved in alginate polymerization). Nonetheless, a different expression pattern observed for lyases could explain the highest alginate molecular weight obtained. Overall, the results suggest that the control of alginate molecular weight in A. vinelandii cells growing in continuous mode is determined by a balance between the gene expression of intracellular and extracellular lyases in response to oxygen availability. These findings better our understanding of the biosynthesis of bacterial alginate and help us progress toward obtain tailor-made alginates.
Collapse
Affiliation(s)
- Alvaro Díaz-Barrera
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Catolica de Valparaiso, Valparaiso, Chile
- * E-mail:
| | - Fabiola Martínez
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Catolica de Valparaiso, Valparaiso, Chile
| | - Felipe Guevara Pezoa
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Catolica de Valparaiso, Valparaiso, Chile
| | - Fernando Acevedo
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Catolica de Valparaiso, Valparaiso, Chile
| |
Collapse
|
44
|
Rebelo SP, Costa R, Estrada M, Shevchenko V, Brito C, Alves PM. HepaRG microencapsulated spheroids in DMSO-free culture: novel culturing approaches for enhanced xenobiotic and biosynthetic metabolism. Arch Toxicol 2014; 89:1347-58. [PMID: 25107451 DOI: 10.1007/s00204-014-1320-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 07/21/2014] [Indexed: 01/08/2023]
Abstract
The need for models that recapitulate liver physiology is perceived for drug development, study of liver disease and bioartificial liver support. The bipotent cell line HepaRG constitutes an efficient surrogate of liver function, yet its differentiated status relies on high concentrations of DMSO, which may compromise the study of drug metabolism and limit the applicability of this hepatic model. Herein, we present a three-dimensional (3D) strategy for the differentiation of HepaRG based on alginate microencapsulation of cell spheroids and culture in dimethyl sulfoxide (DMSO)-free conditions. A ratio of 2.9:1 hepatocyte-like to biliary-like cells was obtained in the 3D culture, with an improvement of 35.9 % in the hepatocyte differentiation when compared with two-dimensional (2D) cultures. The expression of the hepatic identity genes HNF4α and PXR in 3D cultures was comparable to 2D differentiated cultures, while the expression of homeostatic-associated genes albumin and carbamoyl phosphate synthase 1 was higher in 3D. Moreover, the spheroids presented a polarized organization, exhibiting an interconnected bile canalicular network and excretory functionality, assessed by specific activity of MRP2. Importantly, despite variability in basal gene expression levels, the activity of the phase I enzymes cytochrome P450 family 3, subfamily A, polypeptide 4 and cytochrome P450 family 1, subfamily A, polypeptide 2 upon induction was comparable to differentiated 2D cultures and albumin production and ammonia detoxification were enhanced in 3D. The presented model is suitable for toxicological applications, as it allows high throughput analysis of multiple compounds in a DMSO-free setting. Due to the high xenobiotic metabolism and maintenance of biosynthetic functions, the applicability of this model might be broadened to understand liver physiology and for bioartificial liver applications.
Collapse
Affiliation(s)
- Sofia P Rebelo
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901, Oeiras, Portugal
| | | | | | | | | | | |
Collapse
|
45
|
Tiernan AR, Sambanis A. Bioluminescence tracking of alginate micro-encapsulated cell transplants. J Tissue Eng Regen Med 2014; 11:501-508. [PMID: 25047413 DOI: 10.1002/term.1946] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 05/08/2014] [Accepted: 06/16/2014] [Indexed: 01/07/2023]
Abstract
Cell-based therapies to treat loss-of-function hormonal disorders such as diabetes and Parkinson's disease are routinely coupled with encapsulation strategies, but an understanding of when and why grafts fail in vivo is lacking. Consequently, investigators cannot clearly define the key factors that influence graft success. Although bioluminescence is a popular method to track the survival of free cells transplanted in preclinical models, little is known of the ability to use bioluminescence for real-time tracking of microencapsulated cells. Furthermore, the impact that dynamic imaging distances may have, due to freely-floating microcapsules in vivo, on cell survival monitoring is unknown. This work addresses these questions by applying bioluminescence to a pancreatic substitute based on microencapsulated cells. Recombinant insulin-secreting cells were transduced with a luciferase lentivirus and microencapsulated in Ba2+ crosslinked alginate for in vitro and in vivo studies. In vitro quantitative bioluminescence monitoring was possible and viable microencapsulated cells were followed in real time under both normoxic and anoxic conditions. Although in vivo dispersion of freely-floating microcapsules in the peritoneal cavity limited the analysis to a qualitative bioluminescence evaluation, signals consistently four orders of magnitude above background were clear indicators of temporal cell survival. Strong agreement between in vivo and in vitro cell proliferation over time was discovered by making direct bioluminescence comparisons between explanted microcapsules and parallel in vitro cultures. Broader application of this bioluminescence approach to retrievable transplants, in supplement to currently used end-point physiological tests, could improve understanding and accelerate development of cell-based therapies for critical clinical applications. Copyright © 2014 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Aubrey R Tiernan
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Athanassios Sambanis
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
46
|
Workman VL, Tezera LB, Elkington PT, Jayasinghe SN. Controlled Generation of Microspheres Incorporating Extracellular Matrix Fibrils for Three-Dimensional Cell Culture. ADVANCED FUNCTIONAL MATERIALS 2014; 24:2648-2657. [PMID: 25411575 PMCID: PMC4233144 DOI: 10.1002/adfm.201303891] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
A growing body of evidence suggests that studying cell biology in classical two-dimensional formats, such as cell culture plasticware, results in misleading, non-physiological findings. For example, some aspects of cancer biology cannot be observed in 2D, but require 3D culture methods to recapitulate observations in vivo. Therefore, we developed a microsphere-based model to permit 3D cell culture incorporating physiological extracellular matrix components. Bio-electrospraying was chosen as it is the most advanced method to produce microspheres, with THP-1 cells as a model cell line. Bio-electrospraying parameters, such as nozzle size, polymer flow rate, and voltage, were systematically optimized to allow stable production of size controlled microspheres containing extracellular matrix material and human cells. We investigated the effect of bio-electrospraying parameters, alginate type and cell concentration on cell viability using trypan blue and propidium iodide staining. Bio-electrospraying had no effect on cell viability nor the ability of cells to proliferate. Cell viability was similarly minimally affected by encapsulation in all types of alginate tested (MVM, MVG, chemical- and food-grade). Cell density of 5 × 106 cells ml-1 within microspheres was the optimum for cell survival and proliferation. The stable generation of microspheres incorporating cells and extracellular matrix for use in a 3D cell culture will benefit study of many diverse diseases and permit investigation of cellular biology within a 3D matrix.
Collapse
Affiliation(s)
- Victoria L. Workman
- BioPhysics Group, UCL Institute of Biomedical Engineering, UCL Centre for Stem Cells and Regenerative Medicine and Department of Mechanical Engineering, University College London, Torrington Place, London, WC1E 7JE, United Kingdom
| | - Liku B. Tezera
- Clinical and Experimental Sciences, Institute for Life Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, United Kingdom
| | - Paul T. Elkington
- Clinical and Experimental Sciences, Institute for Life Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, United Kingdom
| | - Suwan N. Jayasinghe
- BioPhysics Group, UCL Institute of Biomedical Engineering, UCL Centre for Stem Cells and Regenerative Medicine and Department of Mechanical Engineering, University College London, Torrington Place, London, WC1E 7JE, United Kingdom
| |
Collapse
|
47
|
Wilson JL, Najia MA, Saeed R, McDevitt TC. Alginate encapsulation parameters influence the differentiation of microencapsulated embryonic stem cell aggregates. Biotechnol Bioeng 2014; 111:618-31. [PMID: 24166004 PMCID: PMC4163549 DOI: 10.1002/bit.25121] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 08/26/2013] [Accepted: 09/17/2013] [Indexed: 02/06/2023]
Abstract
Pluripotent embryonic stem cells (ESCs) have tremendous potential as tools for regenerative medicine and drug discovery, yet the lack of processes to manufacture viable and homogenous cell populations of sufficient numbers limits the clinical translation of current and future cell therapies. Microencapsulation of ESCs within microbeads can shield cells from hydrodynamic shear forces found in bioreactor environments while allowing for sufficient diffusion of nutrients and oxygen through the encapsulation material. Despite initial studies examining alginate microbeads as a platform for stem cell expansion and directed differentiation, the impact of alginate encapsulation parameters on stem cell phenotype has not been thoroughly investigated. Therefore, the objective of this study was to systematically examine the effects of varying alginate compositions on microencapsulated ESC expansion and phenotype. Pre-formed aggregates of murine ESCs were encapsulated in alginate microbeads composed of a high or low ratio of guluronic to mannuronic acid residues (High G and High M, respectively), with and without a poly-L-lysine (PLL) coating, thereby providing four distinct alginate bead compositions for analysis. Encapsulation in all alginate compositions was found to delay differentiation, with encapsulation within High G alginate yielding the least differentiated cell population. The addition of a PLL coating to the High G alginate prevented cell escape from beads for up to 14 days. Furthermore, encapsulation within High M alginate promoted differentiation toward a primitive endoderm phenotype. Taken together, the findings of this study suggest that distinct ESC expansion capacities and differentiation trajectories emerge depending on the alginate composition employed, indicating that encapsulation material physical properties can be used to control stem cell fate.
Collapse
Affiliation(s)
- Jenna L Wilson
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | | | | | | |
Collapse
|
48
|
Spasojevic M, Bhujbal S, Paredes G, de Haan BJ, Schouten AJ, de Vos P. Considerations in binding diblock copolymers on hydrophilic alginate beads for providing an immunoprotective membrane. J Biomed Mater Res A 2013; 102:1887-96. [PMID: 23853069 PMCID: PMC4232034 DOI: 10.1002/jbm.a.34863] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 06/13/2013] [Accepted: 06/28/2013] [Indexed: 01/14/2023]
Abstract
Alginate-based microcapsules are being proposed for treatment of many types of diseases. A major obstacle however in the successes is that these capsules are having large lab-to-lab variations. To make the process more reproducible, we propose to cover the surface of alginate capsules with diblock polymers that can form polymer brushes. In the present study, we describe the stepwise considerations for successful application of diblock copolymer of polyethylene glycol (PEG) and poly-l-lysine (PLL) on the surface of alginate beads. Special procedures had to be designed as alginate beads are hydrophilic and most protocols are designed for hydrophobic biomaterials. The successful attachment of diblock copolymer and the presence of PEG blocks on the surface of the capsules were studied by fluorescence microscopy. Longer time periods, that is, 30–60 min, are required to achieve saturation of the surface. The block lengths influenced the strength of the capsules. Shorter PLL blocks resulted in less stable capsules. Adequate permeability of the capsules was achieved with poly(ethylene glycol)-block-poly(l-lysine hydrochloride) (PEG454-b-PLL100) diblock copolymers. The capsules were a barrier for immunoglobulin G. The PEG454-b-PLL100 capsules have similar mechanical properties as PLL capsules. Minor immune activation of nuclear factor κB in THP-1 monocytes was observed with both PLL and PEG454-b-PLL100 capsules prepared from purified alginate. Our results show that we can successfully apply block copolymers on the surface of hydrophilic alginate beads without interfering with the physicochemical properties.
Collapse
Affiliation(s)
- Milica Spasojevic
- Department of Polymer Chemistry, Zernike Institute for Advanced Materials, University of Groningen, 9747, AG Groningen, the Netherlands; Departments of Pathology and Laboratory Medicine, Section of Medical Biology, Division of Immunoendocrinology, University of Groningen, Hanzeplein 1, 9700, RB Groningen, The Netherlands
| | | | | | | | | | | |
Collapse
|