1
|
Ali AS, Wu D, Bannach-Brown A, Dhamrait D, Berg J, Tolksdorf B, Lichtenstein D, Dressler C, Braeuning A, Kurreck J, Hülsemann M. 3D bioprinting of liver models: A systematic scoping review of methods, bioinks, and reporting quality. Mater Today Bio 2024; 26:100991. [PMID: 38558773 PMCID: PMC10978534 DOI: 10.1016/j.mtbio.2024.100991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/19/2024] [Accepted: 02/03/2024] [Indexed: 04/04/2024] Open
Abstract
Background Effective communication is crucial for broad acceptance and applicability of alternative methods in 3R biomedical research and preclinical testing. 3D bioprinting is used to construct intricate biological structures towards functional liver models, specifically engineered for deployment as alternative models in drug screening, toxicological investigations, and tissue engineering. Despite a growing number of reviews in this emerging field, a comprehensive study, systematically assessing practices and reporting quality for bioprinted liver models is missing. Methods In this systematic scoping review we systematically searched MEDLINE (Ovid), EMBASE (Ovid) and BioRxiv for studies published prior to June 2nd, 2022. We extracted data on methodological conduct, applied bioinks, the composition of the printed model, performed experiments and model applications. Records were screened for eligibility and data were extracted from included articles by two independent reviewers from a panel of seven domain experts specializing in bioprinting and liver biology. We used RAYYAN for the screening process and SyRF for data extraction. We used R for data analysis, and R and Graphpad PRISM for visualization. Results Through our systematic database search we identified 1042 records, from which 63 met the eligibility criteria for inclusion in this systematic scoping review. Our findings revealed that extrusion-based printing, in conjunction with bioinks composed of natural components, emerged as the predominant printing technique in the bioprinting of liver models. Notably, the HepG2 hepatoma cell line was the most frequently employed liver cell type, despite acknowledged limitations. Furthermore, 51% of the printed models featured co-cultures with non-parenchymal cells to enhance their complexity. The included studies offered a variety of techniques for characterizing these liver models, with their primary application predominantly focused on toxicity testing. Among the frequently analyzed liver markers, albumin and urea stood out. Additionally, Cytochrome P450 (CYP) isoforms, primarily CYP3A and CYP1A, were assessed, and select studies employed nuclear receptor agonists to induce CYP activity. Conclusion Our systematic scoping review offers an evidence-based overview and evaluation of the current state of research on bioprinted liver models, representing a promising and innovative technology for creating alternative organ models. We conducted a thorough examination of both the methodological and technical facets of model development and scrutinized the reporting quality within the realm of bioprinted liver models. This systematic scoping review can serve as a valuable template for systematically evaluating the progress of organ model development in various other domains. The transparently derived evidence presented here can provide essential support to the research community, facilitating the adaptation of technological advancements, the establishment of standards, and the enhancement of model robustness. This is particularly crucial as we work toward the long-term objective of establishing new approach methods as reliable alternatives to animal testing, with extensive and versatile applications.
Collapse
Affiliation(s)
- Ahmed S.M. Ali
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, Germany
| | - Dongwei Wu
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, Germany
| | - Alexandra Bannach-Brown
- Berlin Institute of Health (BIH) @Charité, QUEST Center for Responsible Research, Berlin, Germany
| | - Diyal Dhamrait
- Berlin Institute of Health (BIH) @Charité, QUEST Center for Responsible Research, Berlin, Germany
| | - Johanna Berg
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, Germany
| | - Beatrice Tolksdorf
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, Germany
| | - Dajana Lichtenstein
- German Federal Institute for Risk Assessment (BfR), Department Food Safety, Berlin, Germany
| | - Corinna Dressler
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Medical Library, Germany
| | - Albert Braeuning
- German Federal Institute for Risk Assessment (BfR), Department Food Safety, Berlin, Germany
| | - Jens Kurreck
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, Germany
| | - Maren Hülsemann
- Berlin Institute of Health (BIH) @Charité, QUEST Center for Responsible Research, Berlin, Germany
| |
Collapse
|
2
|
Gómez‐Álvarez M, Bueno‐Fernandez C, Rodríguez‐Eguren A, Francés‐Herrero E, Agustina‐Hernández M, Faus A, Gisbert Roca F, Martínez‐Ramos C, Galán A, Pellicer A, Ferrero H, Cervelló I. Hybrid Endometrial-Derived Hydrogels: Human Organoid Culture Models and In Vivo Perspectives. Adv Healthc Mater 2024; 13:e2303838. [PMID: 37983675 PMCID: PMC11468130 DOI: 10.1002/adhm.202303838] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Indexed: 11/22/2023]
Abstract
The endometrium plays a vital role in fertility, providing a receptive environment for embryo implantation and development. Understanding the endometrial physiology is essential for developing new strategies to improve reproductive healthcare. Human endometrial organoids (hEOs) are emerging as powerful models for translational research and personalized medicine. However, most hEOs are cultured in a 3D microenvironment that significantly differs from the human endometrium, limiting their applicability in bioengineering. This study presents a hybrid endometrial-derived hydrogel that combines the rigidity of PuraMatrix (PM) with the natural scaffold components and interactions of a porcine decellularized endometrial extracellular matrix (EndoECM) hydrogel. This hydrogel provides outstanding support for hEO culture, enhances hEO differentiation efficiency due to its biochemical similarity with the native tissue, exhibits superior in vivo stability, and demonstrates xenogeneic biocompatibility in mice over a 2-week period. Taken together, these attributes position this hybrid endometrial-derived hydrogel as a promising biomaterial for regenerative treatments in reproductive medicine.
Collapse
Affiliation(s)
- María Gómez‐Álvarez
- IVIRMA Global Research AllianceIVI FoundationInstituto de Investigación Sanitaria La Fe (IIS La Fe)Valencia46026Spain
| | - Clara Bueno‐Fernandez
- IVIRMA Global Research AllianceIVI FoundationInstituto de Investigación Sanitaria La Fe (IIS La Fe)Valencia46026Spain
- Universitat de ValènciaDepartment of PediatricsObstetrics and GynaecologyValencia46010Spain
| | - Adolfo Rodríguez‐Eguren
- IVIRMA Global Research AllianceIVI FoundationInstituto de Investigación Sanitaria La Fe (IIS La Fe)Valencia46026Spain
| | - Emilio Francés‐Herrero
- IVIRMA Global Research AllianceIVI FoundationInstituto de Investigación Sanitaria La Fe (IIS La Fe)Valencia46026Spain
- Universitat de ValènciaDepartment of PediatricsObstetrics and GynaecologyValencia46010Spain
| | - Marcos Agustina‐Hernández
- IVIRMA Global Research AllianceIVI FoundationInstituto de Investigación Sanitaria La Fe (IIS La Fe)Valencia46026Spain
| | - Amparo Faus
- IVIRMA Global Research AllianceIVI FoundationInstituto de Investigación Sanitaria La Fe (IIS La Fe)Valencia46026Spain
| | - Fernando Gisbert Roca
- Universitat Politècnica de ValènciaCentre for Biomaterials and Tissue EngineeringValencia46022Spain
| | - Cristina Martínez‐Ramos
- Universitat Politècnica de ValènciaCentre for Biomaterials and Tissue EngineeringValencia46022Spain
- Unitat Predepartamental de MedicinaUniversitat Jaume ICastellón de la Plana12071Spain
| | - Amparo Galán
- Laboratory of NeuroendocrinologyPrince Felipe Research Center (CIPF)Valencia46012Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)Instituto de Salud Carlos IIIMadrid28029Spain
| | | | - Hortensia Ferrero
- IVIRMA Global Research AllianceIVI FoundationInstituto de Investigación Sanitaria La Fe (IIS La Fe)Valencia46026Spain
| | - Irene Cervelló
- IVIRMA Global Research AllianceIVI FoundationInstituto de Investigación Sanitaria La Fe (IIS La Fe)Valencia46026Spain
| |
Collapse
|
3
|
Suominen S, Hyypijev T, Venäläinen M, Yrjänäinen A, Vuorenpää H, Lehti-Polojärvi M, Räsänen M, Seppänen A, Hyttinen J, Miettinen S, Aalto-Setälä K, Viiri LE. Improvements in Maturity and Stability of 3D iPSC-Derived Hepatocyte-like Cell Cultures. Cells 2023; 12:2368. [PMID: 37830581 PMCID: PMC10571736 DOI: 10.3390/cells12192368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/14/2023] Open
Abstract
Induced pluripotent stem cell (iPSC) technology enables differentiation of human hepatocytes or hepatocyte-like cells (iPSC-HLCs). Advances in 3D culturing platforms enable the development of more in vivo-like liver models that recapitulate the complex liver architecture and functionality better than traditional 2D monocultures. Moreover, within the liver, non-parenchymal cells (NPCs) are critically involved in the regulation and maintenance of hepatocyte metabolic function. Thus, models combining 3D culture and co-culturing of various cell types potentially create more functional in vitro liver models than 2D monocultures. Here, we report the establishment of 3D cultures of iPSC-HLCs alone and in co-culture with human umbilical vein endothelial cells (HUVECs) and adipose tissue-derived mesenchymal stem/stromal cells (hASCs). The 3D cultures were performed as spheroids or on microfluidic chips utilizing various biomaterials. Our results show that both 3D spheroid and on-chip culture enhance the expression of mature liver marker genes and proteins compared to 2D. Among the spheroid models, we saw the best functionality in iPSC-HLC monoculture spheroids. On the contrary, in the chip system, the multilineage model outperformed the monoculture chip model. Additionally, the optical projection tomography (OPT) and electrical impedance tomography (EIT) system revealed changes in spheroid size and electrical conductivity during spheroid culture, suggesting changes in cell-cell connections. Altogether, the present study demonstrates that iPSC-HLCs can successfully be cultured in 3D as spheroids and on microfluidic chips, and co-culturing iPSC-HLCs with NPCs enhances their functionality. These 3D in vitro liver systems are promising human-derived platforms usable in various liver-related studies, specifically when using patient-specific iPSCs.
Collapse
Affiliation(s)
- Siiri Suominen
- Heart Group, Finnish Cardiovascular Research Center and Science Mimicking Life Research Center, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland (L.E.V.)
| | - Tinja Hyypijev
- Heart Group, Finnish Cardiovascular Research Center and Science Mimicking Life Research Center, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland (L.E.V.)
| | - Mari Venäläinen
- Heart Group, Finnish Cardiovascular Research Center and Science Mimicking Life Research Center, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland (L.E.V.)
| | - Alma Yrjänäinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, 33520 Tampere, Finland
| | - Hanna Vuorenpää
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, 33520 Tampere, Finland
| | - Mari Lehti-Polojärvi
- Computational Biophysics and Imaging Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
| | - Mikko Räsänen
- Department of Technical Physics, University of Eastern Finland, 70210 Kuopio, Finland
| | - Aku Seppänen
- Department of Technical Physics, University of Eastern Finland, 70210 Kuopio, Finland
| | - Jari Hyttinen
- Computational Biophysics and Imaging Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
| | - Susanna Miettinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, 33520 Tampere, Finland
| | - Katriina Aalto-Setälä
- Heart Group, Finnish Cardiovascular Research Center and Science Mimicking Life Research Center, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland (L.E.V.)
- Heart Hospital, Tampere University Hospital, 33520 Tampere, Finland
| | - Leena E. Viiri
- Heart Group, Finnish Cardiovascular Research Center and Science Mimicking Life Research Center, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland (L.E.V.)
| |
Collapse
|
4
|
Yao X, Hu Y, Lin M, Peng K, Wang P, Gao Y, Gao X, Guo T, Zhang X, Zhou H. Self-assembling peptide RADA16: a promising scaffold for tissue engineering and regenerative medicine. Nanomedicine (Lond) 2023. [PMID: 37750388 DOI: 10.2217/nnm-2023-0161] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023] Open
Abstract
RADA16 is a peptide-based biomaterial whose acidic aqueous solution spontaneously forms an extracellular matrix-like 3D structure within seconds upon contact with physiological pH body fluids. Meanwhile, its good biocompatibility, low immunogenicity, nontoxic degradation products and ease of modification make it an ideal scaffold for tissue engineering. RADA16 is a good delivery vehicle for cells, drugs and factors. Its shear thinning and thixotropic properties allow it to fill tissue voids by injection and not to swell. However, the weaker mechanical properties and poor hydrophilicity are troubling limitations of RADA16. To compensate for this limitation, various functional groups and polymers have been designed to modify RADA16, thus contributing to its scope and progress in the field of tissue engineering.
Collapse
Affiliation(s)
- Xin Yao
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
- Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China
| | - Yicun Hu
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
- Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China
| | - Maoqiang Lin
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
- Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China
| | - Kaichen Peng
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
- Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China
| | - Peng Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
- Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China
| | - Yanbing Gao
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
- Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China
| | - Xidan Gao
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710000, Shaanxi, China
| | - Taowen Guo
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
- Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China
| | - Xiaobo Zhang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710000, Shaanxi, China
| | - Haiyu Zhou
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
- Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China
| |
Collapse
|
5
|
Sztankovics D, Moldvai D, Petővári G, Gelencsér R, Krencz I, Raffay R, Dankó T, Sebestyén A. 3D bioprinting and the revolution in experimental cancer model systems-A review of developing new models and experiences with in vitro 3D bioprinted breast cancer tissue-mimetic structures. Pathol Oncol Res 2023; 29:1610996. [PMID: 36843955 PMCID: PMC9946983 DOI: 10.3389/pore.2023.1610996] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/16/2023] [Indexed: 02/11/2023]
Abstract
Growing evidence propagates those alternative technologies (relevant human cell-based-e.g., organ-on-chips or biofabricated models-or artificial intelligence-combined technologies) that could help in vitro test and predict human response and toxicity in medical research more accurately. In vitro disease model developments have great efforts to create and serve the need of reducing and replacing animal experiments and establishing human cell-based in vitro test systems for research use, innovations, and drug tests. We need human cell-based test systems for disease models and experimental cancer research; therefore, in vitro three-dimensional (3D) models have a renaissance, and the rediscovery and development of these technologies are growing ever faster. This recent paper summarises the early history of cell biology/cellular pathology, cell-, tissue culturing, and cancer research models. In addition, we highlight the results of the increasing use of 3D model systems and the 3D bioprinted/biofabricated model developments. Moreover, we present our newly established 3D bioprinted luminal B type breast cancer model system, and the advantages of in vitro 3D models, especially the bioprinted ones. Based on our results and the reviewed developments of in vitro breast cancer models, the heterogeneity and the real in vivo situation of cancer tissues can be represented better by using 3D bioprinted, biofabricated models. However, standardising the 3D bioprinting methods is necessary for future applications in different high-throughput drug tests and patient-derived tumour models. Applying these standardised new models can lead to the point that cancer drug developments will be more successful, efficient, and consequently cost-effective in the near future.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Anna Sebestyén
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| |
Collapse
|
6
|
Shafique H, de Vries J, Strauss J, Khorrami Jahromi A, Siavash Moakhar R, Mahshid S. Advances in the Translation of Electrochemical Hydrogel-Based Sensors. Adv Healthc Mater 2023; 12:e2201501. [PMID: 36300601 DOI: 10.1002/adhm.202201501] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/26/2022] [Indexed: 02/03/2023]
Abstract
Novel biomaterials for bio- and chemical sensing applications have gained considerable traction in the diagnostic community with rising trends of using biocompatible and lowly cytotoxic material. Hydrogel-based electrochemical sensors have become a promising candidate for their swellable, nano-/microporous, and aqueous 3D structures capable of immobilizing catalytic enzymes, electroactive species, whole cells, and complex tissue models, while maintaining tunable mechanical properties in wearable and implantable applications. With advances in highly controllable fabrication and processability of these novel biomaterials, the possibility of bio-nanocomposite hydrogel-based electrochemical sensing presents a paradigm shift in the development of biocompatible, "smart," and sensitive health monitoring point-of-care devices. Here, recent advances in electrochemical hydrogels for the detection of biomarkers in vitro, in situ, and in vivo are briefly reviewed to demonstrate their applicability in ideal conditions, in complex cellular environments, and in live animal models, respectively, to provide a comprehensive assessment of whether these biomaterials are ready for point-of-care translation and biointegration. Sensors based on conductive and nonconductive polymers are presented, with highlights of nano-/microstructured electrodes that provide enhanced sensitivity and selectivity in biocompatible matrices. An outlook on current challenges that shall be addressed for the realization of truly continuous real-time sensing platforms is also presented.
Collapse
Affiliation(s)
- Houda Shafique
- Department of Bioengineering, McGill University, Montreal, QC, H3A 0E9, Canada
| | - Justin de Vries
- Department of Bioengineering, McGill University, Montreal, QC, H3A 0E9, Canada
| | - Julia Strauss
- Department of Bioengineering, McGill University, Montreal, QC, H3A 0E9, Canada
| | | | | | - Sara Mahshid
- Department of Bioengineering, McGill University, Montreal, QC, H3A 0E9, Canada
| |
Collapse
|
7
|
Merivaara A, Koivunotko E, Manninen K, Kaseva T, Monola J, Salli E, Koivuniemi R, Savolainen S, Valkonen S, Yliperttula M. Stiffness-Controlled Hydrogels for 3D Cell Culture Models. Polymers (Basel) 2022; 14:polym14245530. [PMID: 36559897 PMCID: PMC9786583 DOI: 10.3390/polym14245530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/08/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Nanofibrillated cellulose (NFC) hydrogel is a versatile biomaterial suitable, for example, for three-dimensional (3D) cell spheroid culturing, drug delivery, and wound treatment. By freeze-drying NFC hydrogel, highly porous NFC structures can be manufactured. We freeze-dried NFC hydrogel and subsequently reconstituted the samples into a variety of concentrations of NFC fibers, which resulted in different stiffness of the material, i.e., different mechanical cues. After the successful freeze-drying and reconstitution, we showed that freeze-dried NFC hydrogel can be used for one-step 3D cell spheroid culturing of primary mesenchymal stem/stromal cells, prostate cancer cells (PC3), and hepatocellular carcinoma cells (HepG2). No difference was observed in the viability or morphology between the 3D cell spheroids cultured in the freeze-dried and reconstituted NFC hydrogel and fresh NFC hydrogel. Furthermore, the 3D cultured spheroids showed stable metabolic activity and nearly 100% viability. Finally, we applied a convolutional neural network (CNN)-based automatic nuclei segmentation approach to automatically segment individual cells of 3D cultured PC3 and HepG2 spheroids. These results provide an application to culture 3D cell spheroids more readily with the NFC hydrogel and a step towards automatization of 3D cell culturing and analysis.
Collapse
Affiliation(s)
- Arto Merivaara
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
- Correspondence: (A.M.); (M.Y.); Tel.:+358-294-159-577 (A.M.); +358-294-159-141 (M.Y.)
| | - Elle Koivunotko
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Kalle Manninen
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Tuomas Kaseva
- HUS Medical Imaging Center, Radiology, University of Helsinki and Helsinki University Hospital, 00290 Helsinki, Finland
| | - Julia Monola
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Eero Salli
- HUS Medical Imaging Center, Radiology, University of Helsinki and Helsinki University Hospital, 00290 Helsinki, Finland
| | - Raili Koivuniemi
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Sauli Savolainen
- HUS Medical Imaging Center, Radiology, University of Helsinki and Helsinki University Hospital, 00290 Helsinki, Finland
- Department of Physics, University of Helsinki, 00014 Helsinki, Finland
| | - Sami Valkonen
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
- School of Pharmacy, University of Eastern Finland, 70210 Kuopio, Finland
| | - Marjo Yliperttula
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
- Correspondence: (A.M.); (M.Y.); Tel.:+358-294-159-577 (A.M.); +358-294-159-141 (M.Y.)
| |
Collapse
|
8
|
Yang Z, Liu X, Cribbin EM, Kim AM, Li JJ, Yong KT. Liver-on-a-chip: Considerations, advances, and beyond. BIOMICROFLUIDICS 2022; 16:061502. [PMID: 36389273 PMCID: PMC9646254 DOI: 10.1063/5.0106855] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/25/2022] [Indexed: 05/14/2023]
Abstract
The liver is the largest internal organ in the human body with largest mass of glandular tissue. Modeling the liver has been challenging due to its variety of major functions, including processing nutrients and vitamins, detoxification, and regulating body metabolism. The intrinsic shortfalls of conventional two-dimensional (2D) cell culture methods for studying pharmacokinetics in parenchymal cells (hepatocytes) have contributed to suboptimal outcomes in clinical trials and drug development. This prompts the development of highly automated, biomimetic liver-on-a-chip (LOC) devices to simulate native liver structure and function, with the aid of recent progress in microfluidics. LOC offers a cost-effective and accurate model for pharmacokinetics, pharmacodynamics, and toxicity studies. This review provides a critical update on recent developments in designing LOCs and fabrication strategies. We highlight biomimetic design approaches for LOCs, including mimicking liver structure and function, and their diverse applications in areas such as drug screening, toxicity assessment, and real-time biosensing. We capture the newest ideas in the field to advance the field of LOCs and address current challenges.
Collapse
Affiliation(s)
| | | | - Elise M. Cribbin
- School of Biomedical Engineering, University of Technology Sydney, New South Wales 2007, Australia
| | - Alice M. Kim
- School of Biomedical Engineering, University of Technology Sydney, New South Wales 2007, Australia
| | - Jiao Jiao Li
- Authors to whom correspondence should be addressed: and
| | - Ken-Tye Yong
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
9
|
McDuffie D, Barr D, Agarwal A, Thomas E. Physiologically relevant microsystems to study viral infection in the human liver. Front Microbiol 2022; 13:999366. [PMID: 36246284 PMCID: PMC9555087 DOI: 10.3389/fmicb.2022.999366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Viral hepatitis is a leading cause of liver disease and mortality. Infection can occur acutely or chronically, but the mechanisms that govern the clearance of virus or lack thereof are poorly understood and merit further investigation. Though cures for viral hepatitis have been developed, they are expensive, not readily accessible in vulnerable populations and some patients may remain at an increased risk of developing hepatocellular carcinoma (HCC) even after viral clearance. To sustain infection in vitro, hepatocytes must be fully mature and remain in a differentiated state. However, primary hepatocytes rapidly dedifferentiate in conventional 2D in vitro platforms. Physiologically relevant or physiomimetic microsystems, are increasingly popular alternatives to traditional two-dimensional (2D) monocultures for in vitro studies. Physiomimetic systems reconstruct and incorporate elements of the native cellular microenvironment to improve biologic functionality in vitro. Multiple elements contribute to these models including ancillary tissue architecture, cell co-cultures, matrix proteins, chemical gradients and mechanical forces that contribute to increased viability, longevity and physiologic function for the tissue of interest. These microsystems are used in a wide variety of applications to study biological phenomena. Here, we explore the use of physiomimetic microsystems as tools for studying viral hepatitis infection in the liver and how the design of these platforms is tailored for enhanced investigation of the viral lifecycle when compared to conventional 2D cell culture models. Although liver-based physiomimetic microsystems are typically applied in the context of drug studies, the platforms developed for drug discovery purposes offer a solid foundation to support studies on viral hepatitis. Physiomimetic platforms may help prolong hepatocyte functionality in order to sustain chronic viral hepatitis infection in vitro for studying virus-host interactions for prolonged periods.
Collapse
Affiliation(s)
- Dennis McDuffie
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
| | - David Barr
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Emmanuel Thomas
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
- Schiff Center for Liver Diseases, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
10
|
Germain N, Dhayer M, Dekiouk S, Marchetti P. Current Advances in 3D Bioprinting for Cancer Modeling and Personalized Medicine. Int J Mol Sci 2022; 23:3432. [PMID: 35408789 PMCID: PMC8998835 DOI: 10.3390/ijms23073432] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 02/01/2023] Open
Abstract
Tumor cells evolve in a complex and heterogeneous environment composed of different cell types and an extracellular matrix. Current 2D culture methods are very limited in their ability to mimic the cancer cell environment. In recent years, various 3D models of cancer cells have been developed, notably in the form of spheroids/organoids, using scaffold or cancer-on-chip devices. However, these models have the disadvantage of not being able to precisely control the organization of multiple cell types in complex architecture and are sometimes not very reproducible in their production, and this is especially true for spheroids. Three-dimensional bioprinting can produce complex, multi-cellular, and reproducible constructs in which the matrix composition and rigidity can be adapted locally or globally to the tumor model studied. For these reasons, 3D bioprinting seems to be the technique of choice to mimic the tumor microenvironment in vivo as closely as possible. In this review, we discuss different 3D-bioprinting technologies, including bioinks and crosslinkers that can be used for in vitro cancer models and the techniques used to study cells grown in hydrogels; finally, we provide some applications of bioprinted cancer models.
Collapse
Affiliation(s)
- Nicolas Germain
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
- Banque de Tissus, Centre de Biologie-Pathologie, CHU Lille, F-59000 Lille, France
| | - Melanie Dhayer
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
| | - Salim Dekiouk
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
| | - Philippe Marchetti
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
- Banque de Tissus, Centre de Biologie-Pathologie, CHU Lille, F-59000 Lille, France
| |
Collapse
|
11
|
Kang SY, Joshi P, Lee MY. High-Throughput Screening of Compound Neurotoxicity Using 3D-Cultured Neural Stem Cells on a 384-Pillar Plate. Curr Protoc 2021; 1:e107. [PMID: 33887124 DOI: 10.1002/cpz1.107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Assessing the neurotoxicity of test chemicals has typically been performed using two-dimensionally (2D)-cultured neuronal cell monolayers and animal models. The in vitro 2D cell models are simple and straightforward compared to animal models, which have the disadvantage of being relatively low throughput, expensive, and time consuming. Despite their extensive use in this area of neurotoxicology research, both models often do not accurately recapitulate human outcomes. To bridge this gap and attempt to better replicate what happens in vivo, three-dimensionally (3D) cultured neural stem cells (NSCs) encapsulated in hydrogels on a 384-pillar plate have been developed via miniature 3D bioprinting. This technology allows users to print NSCs on a pillar plate for rapid 3D cell culture as well as high-throughput compound screening. For this, the 384-pillar plate with bioprinted NSCs is sandwiched with a standard 384-well plate with growth medium for 3D culture, allowing researchers to expose the cells to test compounds and stain them with various fluorescent dyes for a suite of high-content imaging assays, including assays for DNA damage, mitochondrial impairment, cell membrane integrity, intracellular glutathione levels, and apoptosis. After acquiring cell images from an automated fluorescence microscope and extracting fluorescence intensities, researchers can obtain the IC50 value of each compound to evaluate critical parameters in neurotoxicity. Here, we provide a detailed description of protocols for cell printing on a 384-pillar plate, 3D NSC culture, compound testing, 3D cell staining, and image acquisition and analysis, which altogether will allow researchers to investigate mechanisms of compound neurotoxicity with 3D-cultured NSCs in a high-throughput manner. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Three-dimensional neural stem cell culture on a 384-pillar plate Basic Protocol 2: Compound treatment and cell staining Basic Protocol 3: Image acquisition, processing, and data analysis.
Collapse
Affiliation(s)
- Soo-Yeon Kang
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, Ohio
| | - Pranav Joshi
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, Ohio
| | - Moo-Yeal Lee
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, Ohio
| |
Collapse
|
12
|
Tissue Chips and Microphysiological Systems for Disease Modeling and Drug Testing. MICROMACHINES 2021; 12:mi12020139. [PMID: 33525451 PMCID: PMC7911320 DOI: 10.3390/mi12020139] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/23/2021] [Accepted: 01/26/2021] [Indexed: 12/15/2022]
Abstract
Tissue chips (TCs) and microphysiological systems (MPSs) that incorporate human cells are novel platforms to model disease and screen drugs and provide an alternative to traditional animal studies. This review highlights the basic definitions of TCs and MPSs, examines four major organs/tissues, identifies critical parameters for organization and function (tissue organization, blood flow, and physical stresses), reviews current microfluidic approaches to recreate tissues, and discusses current shortcomings and future directions for the development and application of these technologies. The organs emphasized are those involved in the metabolism or excretion of drugs (hepatic and renal systems) and organs sensitive to drug toxicity (cardiovascular system). This article examines the microfluidic/microfabrication approaches for each organ individually and identifies specific examples of TCs. This review will provide an excellent starting point for understanding, designing, and constructing novel TCs for possible integration within MPS.
Collapse
|
13
|
Kucinska M, Plewinski A, Szczolko W, Kaczmarek M, Goslinski T, Murias M. Modeling the photodynamic effect in 2D versus 3D cell culture under normoxic and hypoxic conditions. Free Radic Biol Med 2021; 162:309-326. [PMID: 33141030 DOI: 10.1016/j.freeradbiomed.2020.10.304] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/01/2020] [Accepted: 10/20/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND Photodynamic therapy (PDT), mainly as a combined therapy, can still be considered a promising technology for targeted cancer treatment. Besides the several and essential benefits of PDT, there are some concerns and limitations, such as complex dosimetry, tumor hypoxia, and other mechanisms of resistance. In this study, we present how the cell culture model and cell culture conditions may affect the response to PDT treatment. It was studied by applying two different 3D cell culture, non-scaffold, and hydrogel-based models under normoxic and hypoxic conditions. In parallel, a detailed mechanism of the action of zinc phthalocyanine M2TG3 was presented. METHODS Hydrogel-based and tumor spheroids consisting of LNCaP cells, were used as 3D cell culture models in experiments performed under normoxic and hypoxic (1% of oxygen) conditions. Several analyses were performed to compare the activity of M2TG3 under different conditions, such as cytotoxicity, the level of proapoptotic and stress-related proteins, caspase activity, and antioxidant gene expression status. Additionally, we tested bioluminescence and fluorescence assays as a useful approach for a hydrogel-based 3D cell culture. RESULTS We found that M2TG3 might lead to apoptotic cancer cell death and is strongly dependent on the model and oxygen availability. Moreover, the expression of the genes modulated in the antioxidative system in 2D and 3D cell culture models were presented. The tested bioluminescence assay revealed several advantages, such as repetitive measurements on the same sample and simultaneous analysis of different parameters due to the non-lysing nature of this assay. CONCLUSIONS It was shown that M2TG3 can effectively cause cancer cell death via a different mechanism, depending on cell culture conditions such as the model and oxygen availability.
Collapse
Affiliation(s)
- Malgorzata Kucinska
- Department of Toxicology, Poznan University of Medical Sciences, Dojazd 30 Street, 60-631, Poznan, Poland.
| | - Adam Plewinski
- Centre for Advanced Technologies, Adam Mickiewicz University, Uniwersytet Poznanski 10 Street, 61-614, Poznan, Poland
| | - Wojciech Szczolko
- Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Grunwaldzka 6 Street, 60-780, Poznan, Poland
| | - Mariusz Kaczmarek
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Garbary 15 Street, 61-866, Poznan, Poland; Department of Cancer Diagnostics and Immunology, Gene Therapy Unit, Greater Poland Cancer Centre, Garbary 15 Street, 61-866, Poznan, Poland
| | - Tomasz Goslinski
- Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Grunwaldzka 6 Street, 60-780, Poznan, Poland
| | - Marek Murias
- Department of Toxicology, Poznan University of Medical Sciences, Dojazd 30 Street, 60-631, Poznan, Poland; Centre for Advanced Technologies, Adam Mickiewicz University, Uniwersytet Poznanski 10 Street, 61-614, Poznan, Poland.
| |
Collapse
|
14
|
Wang Y, Zhang W, Gong C, Liu B, Li Y, Wang L, Su Z, Wei G. Recent advances in the fabrication, functionalization, and bioapplications of peptide hydrogels. SOFT MATTER 2020; 16:10029-10045. [PMID: 32696801 DOI: 10.1039/d0sm00966k] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Self-assembled peptide-based nanomaterials have exhibited wide application potential in the fields of materials science, nanodevices, biomedicine, tissue engineering, biosensors, energy storage, environmental science, and others. Due to their porous structure, strong mechanical stability, high biocompatibility, and easy functionalization, three-dimensional self-assembled peptide hydrogels revealed promising potential in bio-related applications. To present the advances in this interesting topic, we present a review on the synthesis and functionalization of peptide hydrogels, as well as their applications in drug delivery, antibacterial materials, cell culture, biomineralization, bone tissue engineering, and biosensors. Specifically, we focus on the fabrication methods of peptide hydrogels through physical, chemical, and biological stimulations. In addition, the functional design of peptide hydrogels by incorporation with polymers, DNA, protein, nanoparticles, and carbon materials is introduced and discussed in detail. It is expected that this work will be helpful not only for the design and synthesis of various peptide-based nanostructures and nanomaterials, but also for the structural and functional tailoring of peptide-based nanomaterials to meet specific demands.
Collapse
Affiliation(s)
- Yan Wang
- College of Chemistry and Chemical Engineering, Qingdao University, 266071 Qingdao, P. R. China.
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Bigdelou P, Chan KK, Tang J, Yu KN, Whited J, Wang D, Lee MY, Sun XL. High-throughput multiplex assays with mouse macrophages on pillar plate platforms. Exp Cell Res 2020; 396:112243. [PMID: 32835658 PMCID: PMC7572780 DOI: 10.1016/j.yexcr.2020.112243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/08/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022]
Abstract
It is challenging to rapidly identify immune responses that reflect the state and capability of immune cells due to complex heterogeneity of immune cells and their plasticity to pathogens and modulating molecules. Thus, high-throughput and easy-to-use cell culture and analysis platforms are highly desired for characterizing complex immune responses and elucidating their underlying mechanisms as well. In response to this need, we have developed a micropillar chip and a 384-pillar plate, printed mouse macrophage, RAW 264.7 cell line in alginate on the pillar plate platforms, and established multiplex cell-based assays to rapidly measure cell viability, expression of cell surface markers, and secretion of cytokines upon stimulation with model compound, lipopolysaccharide (LPS), as well as synthetic N-glycan polymers that mimic native glycoconjugates and could bind to lectin receptors on RAW 264.7 cells. Interestingly, changes in RAW 264.7 cell viability, expression levels of cell surface makers, and release of cytokines measured from the pillar plate platforms in the presence and absence of LPS were well correlated with those obtained from their counterpart, the 96-well plate with 2D-cultured macrophages. With this approach, we identified that α2,3-linked N-sialyllactose polymer has significant macrophage modulation activity among the N-glycan polymers tested. Therefore, we successfully demonstrated that our pillar plate platforms with 3D-cultured macrophages can streamline immune cell imaging and analysis in high throughput in response to compound stimulation. We envision that the pillar plate platforms could potentially be used for rapid characterization of immune cell responses and for screening immune cell-modulating molecules.
Collapse
Affiliation(s)
- Parnian Bigdelou
- Department of Chemical & Biomedical Engineering, Cleveland State University, Cleveland, OH, 44115, USA
| | - Ka Keung Chan
- Department of Chemistry and Center of Gene Regulation of Health and Disease (GRHD), Cleveland State University, Cleveland, OH, 44115, USA
| | - Jinshan Tang
- Department of Chemistry and Center of Gene Regulation of Health and Disease (GRHD), Cleveland State University, Cleveland, OH, 44115, USA; Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, Jinan University, West 601, Huangpu Avenue, Guangzhou, PR China
| | - Kyeong-Nam Yu
- Department of Chemical & Biomedical Engineering, Cleveland State University, Cleveland, OH, 44115, USA
| | - Joshua Whited
- Department of Chemistry and Center of Gene Regulation of Health and Disease (GRHD), Cleveland State University, Cleveland, OH, 44115, USA
| | - Dan Wang
- Department of Chemistry and Center of Gene Regulation of Health and Disease (GRHD), Cleveland State University, Cleveland, OH, 44115, USA
| | - Moo-Yeal Lee
- Department of Chemical & Biomedical Engineering, Cleveland State University, Cleveland, OH, 44115, USA.
| | - Xue-Long Sun
- Department of Chemical & Biomedical Engineering, Cleveland State University, Cleveland, OH, 44115, USA; Department of Chemistry and Center of Gene Regulation of Health and Disease (GRHD), Cleveland State University, Cleveland, OH, 44115, USA.
| |
Collapse
|
16
|
GhavamiNejad A, Ashammakhi N, Wu XY, Khademhosseini A. Crosslinking Strategies for 3D Bioprinting of Polymeric Hydrogels. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2002931. [PMID: 32734720 PMCID: PMC7754762 DOI: 10.1002/smll.202002931] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Indexed: 05/15/2023]
Abstract
Three-dimensional (3D) bioprinting has recently advanced as an important tool to produce viable constructs that can be used for regenerative purposes or as tissue models. To develop biomimetic and sustainable 3D constructs, several important processing aspects need to be considered, among which crosslinking is most important for achieving desirable biomechanical stability of printed structures, which is reflected in subsequent behavior and use of these constructs. In this work, crosslinking methods used in 3D bioprinting studies are reviewed, parameters that affect bioink chemistry are discussed, and the potential toward improving crosslinking outcomes and construct performance is highlighted. Furthermore, current challenges and future prospects are discussed. Due to the direct connection between crosslinking methods and properties of 3D bioprinted structures, this Review can provide a basis for developing necessary modifications to the design and manufacturing process of advanced tissue-like constructs in future.
Collapse
Affiliation(s)
- Amin GhavamiNejad
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada
| | - Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics, California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Radiological Sciences, University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Xiao Yu Wu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics, California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Radiological Sciences, University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, California, USA
| |
Collapse
|
17
|
Huang Y, Feng Q, Jiang H, Zhou W, Chen J, Gao J, Wang K, Wan X, Yu Y. Mimicking the Endometrial Cancer Tumor Microenvironment to Reprogram Tumor-Associated Macrophages in Disintegrable Supramolecular Gelatin Hydrogel. Int J Nanomedicine 2020; 15:4625-4637. [PMID: 32636622 PMCID: PMC7326693 DOI: 10.2147/ijn.s252074] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/09/2020] [Indexed: 12/31/2022] Open
Abstract
Purpose Besides the tumor cells themselves, solid tumors are comprised of numerous cell types including infiltrating immune cells such as tumor-associated macrophages (TAMs). TAMs are vital stromal components of host immune system and play a critical role in the development of cancer. TAMs can be divided into two subtypes: M1 tumor-suppressive macrophage and M2 tumor-supportive macrophage. To better address the observations of TAMs functional performance, we describe an in vitro system that mimics the populations of TAMs infiltrated into the tumor mass by using our disintegrable supramolecular gelatin (DSG) hydrogels, which are physically crosslinked by host-guest complexations. Materials and Methods The host–guest interaction was adopted between the aromatic groups of gelatin and the photocrosslinkable acrylated β-cyclodextrins (Ac-β-CDs) to form the DSG hydrogels. The convenient macrophage/endometrial cancer cells heterospheroid 3D model was set up by DSG hydrogels. RT-PCR and Western blot assays were developed to evaluate the efficiencies of inducers on the macrophages. The ELISA and oxygen saturation assays were performed to measure the secretion of VEGF and consumption of oxygen of tumor and/or macrophages, respectively. To determine the antitumor effects of M2 reprogrammed macrophages in vitro and in vivo, migration assay and tumor xenograft model were used, respectively. Results The host-guest complexations of DSG hydrogels were controllably broken efficiently by soaking into the solution of competitive guest monomers 1-adamantanamine hydrochloride. The DSG hydrogels help IFN-γ reprogram the M2 to M1 and then decrease the tumor/M2 reprogrammed macrophage cells heterospheroid secretion of VEGF and increase the relative oxygen saturation. Significantly, the co-cultural tumor/M2 reprogrammed group from the disintegrated DSG hydrogels reduced the migration of cancer cells in vitro and the tumor growth in vivo. Conclusion We obtain a TAMs/tumor microenvironment-responsive 3D model based on the novel DSG hydrogels, and will be of utility in cancer therapy and drug discovery.
Collapse
Affiliation(s)
- Yujia Huang
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China.,Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Qian Feng
- Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, College of Chemistry and Materials Science, Fujian Normal University, Fuzhou, People's Republic of China
| | - Huabo Jiang
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Wanding Zhou
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China.,Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Jinhong Chen
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Jie Gao
- Institute of Translational Medicine, Shanghai University, Shanghai, People's Republic of China
| | - Kai Wang
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xiaoping Wan
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Yongsheng Yu
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
18
|
|
19
|
Kermanizadeh A, Moritz W. Next generation in vitro primary hepatic cell test systems-their suitability as an alternative to in vivo testing? Hepatobiliary Surg Nutr 2020; 9:103-105. [PMID: 32140492 DOI: 10.21037/hbsn.2019.09.09] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Ali Kermanizadeh
- Heriot Watt University, School of Engineering and Physical Sciences, Edinburgh, UK
| | | |
Collapse
|
20
|
Liu H, Wang Y, Cui K, Guo Y, Zhang X, Qin J. Advances in Hydrogels in Organoids and Organs-on-a-Chip. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1902042. [PMID: 31282047 DOI: 10.1002/adma.201902042] [Citation(s) in RCA: 205] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/25/2019] [Indexed: 05/10/2023]
Abstract
Significant advances in materials, microscale technology, and stem cell biology have enabled the construction of 3D tissues and organs, which will ultimately lead to more effective diagnostics and therapy. Organoids and organs-on-a-chip (OOC), evolved from developmental biology and bioengineering principles, have emerged as major technological breakthrough and distinct model systems to revolutionize biomedical research and drug discovery by recapitulating the key structural and functional complexity of human organs in vitro. There is growing interest in the development of functional biomaterials, especially hydrogels, for utilization in these promising systems to build more physiologically relevant 3D tissues with defined properties. The remarkable properties of defined hydrogels as proper extracellular matrix that can instruct cellular behaviors are presented. The recent trend where functional hydrogels are integrated into organoids and OOC systems for the construction of 3D tissue models is highlighted. Future opportunities and perspectives in the development of advanced hydrogels toward accelerating organoids and OOC research in biomedical applications are also discussed.
Collapse
Affiliation(s)
- Haitao Liu
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yaqing Wang
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kangli Cui
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yaqiong Guo
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xu Zhang
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Jianhua Qin
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| |
Collapse
|
21
|
Deng J, Wei W, Chen Z, Lin B, Zhao W, Luo Y, Zhang X. Engineered Liver-on-a-Chip Platform to Mimic Liver Functions and Its Biomedical Applications: A Review. MICROMACHINES 2019; 10:E676. [PMID: 31591365 PMCID: PMC6843249 DOI: 10.3390/mi10100676] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/03/2019] [Accepted: 10/03/2019] [Indexed: 02/07/2023]
Abstract
Hepatology and drug development for liver diseases require in vitro liver models. Typical models include 2D planar primary hepatocytes, hepatocyte spheroids, hepatocyte organoids, and liver-on-a-chip. Liver-on-a-chip has emerged as the mainstream model for drug development because it recapitulates the liver microenvironment and has good assay robustness such as reproducibility. Liver-on-a-chip with human primary cells can potentially correlate clinical testing. Liver-on-a-chip can not only predict drug hepatotoxicity and drug metabolism, but also connect other artificial organs on the chip for a human-on-a-chip, which can reflect the overall effect of a drug. Engineering an effective liver-on-a-chip device requires knowledge of multiple disciplines including chemistry, fluidic mechanics, cell biology, electrics, and optics. This review first introduces the physiological microenvironments in the liver, especially the cell composition and its specialized roles, and then summarizes the strategies to build a liver-on-a-chip via microfluidic technologies and its biomedical applications. In addition, the latest advancements of liver-on-a-chip technologies are discussed, which serve as a basis for further liver-on-a-chip research.
Collapse
Affiliation(s)
- Jiu Deng
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (J.D.); (W.W.); (W.Z.); (Y.L.)
| | - Wenbo Wei
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (J.D.); (W.W.); (W.Z.); (Y.L.)
| | - Zongzheng Chen
- Integrated Chinese and Western Medicine Postdoctoral research station, Jinan University, Guangzhou 510632, China;
| | - Bingcheng Lin
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (J.D.); (W.W.); (W.Z.); (Y.L.)
| | - Weijie Zhao
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (J.D.); (W.W.); (W.Z.); (Y.L.)
| | - Yong Luo
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (J.D.); (W.W.); (W.Z.); (Y.L.)
| | - Xiuli Zhang
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| |
Collapse
|