1
|
Liu S, Chen Y, Wang Z, Liu M, Zhao Y, Tan Y, Qu Z, Du L, Wu C. The cutting-edge progress in bioprinting for biomedicine: principles, applications, and future perspectives. MedComm (Beijing) 2024; 5:e753. [PMID: 39314888 PMCID: PMC11417428 DOI: 10.1002/mco2.753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 09/25/2024] Open
Abstract
Bioprinting is a highly promising application area of additive manufacturing technology that has been widely used in various fields, including tissue engineering, drug screening, organ regeneration, and biosensing. Its primary goal is to produce biomedical products such as artificial implant scaffolds, tissues and organs, and medical assistive devices through software-layered discrete and numerical control molding. Despite its immense potential, bioprinting technology still faces several challenges. It requires concerted efforts from researchers, engineers, regulatory bodies, and industry stakeholders are principal to overcome these challenges and unlock the full potential of bioprinting. This review systematically discusses bioprinting principles, applications, and future perspectives while also providing a topical overview of research progress in bioprinting over the past two decades. The most recent advancements in bioprinting are comprehensively reviewed here. First, printing techniques and methods are summarized along with advancements related to bioinks and supporting structures. Second, interesting and representative cases regarding the applications of bioprinting in tissue engineering, drug screening, organ regeneration, and biosensing are introduced in detail. Finally, the remaining challenges and suggestions for future directions of bioprinting technology are proposed and discussed. Bioprinting is one of the most promising application areas of additive manufacturing technology that has been widely used in various fields. It aims to produce biomedical products such as artificial implant scaffolds, tissues and organs, and medical assistive devices. This review systematically discusses bioprinting principles, applications, and future perspectives, which provides a topical description of the research progress of bioprinting.
Collapse
Affiliation(s)
- Shuge Liu
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Yating Chen
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Zhiyao Wang
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Minggao Liu
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Yundi Zhao
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Yushuo Tan
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Zhan Qu
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Liping Du
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Chunsheng Wu
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| |
Collapse
|
2
|
He H, Hong M, Yang F, Wang G, Wang Y, Yang M, Huang D, Liu H, Wang Y. Preparation of Controlled Multicompartmental Gel Microcarriers Based on Aqueous Two-Phase Emulsions for 3D Partitioned Cell Coculture In Vitro. Biomacromolecules 2024; 25:4469-4481. [PMID: 38877974 DOI: 10.1021/acs.biomac.4c00516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
A facile method was proposed for preparing controllable multicompartment gel microcarriers using an aqueous two-phase emulsion system. By leveraging the density difference between the upper polyethylene glycol solution and the lower dextran-calcium chloride (CaCl2) solution in the collection solution and the high viscosity of the lower solution, controllable fusion of core-shell droplets made by coextrusion devices was achieved at the water/water (w/w) interface to fabricate microcarriers with separated core compartments. By adjusting the sodium alginate concentration, collected solution composition, and number of fused liquid droplets, the pore size, shape, and number of compartments could be controlled. Caco-2 and HepG2 cells were encapsulated in different compartments to establish gut-liver coculture models, exhibiting higher viability and proliferation compared to monoculture models. Notably, significant differences in cytokine expression and functional proteins were observed between the coculture and monoculture models. This method provides new possibilities for preparing complex and functional three-dimensional coculture materials.
Collapse
Affiliation(s)
- Huatao He
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, P. R. China
| | - Meiying Hong
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, P. R. China
| | - Feng Yang
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, P. R. China
| | - Guanxiong Wang
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, P. R. China
| | - Yilan Wang
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, P. R. China
| | - Menghan Yang
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, P. R. China
| | - Deqing Huang
- School of Electrical Engineering, Southwest Jiaotong University, Chengdu 610031, China P. R. China
| | - Hong Liu
- Department of General Surgery, Wuxi No.5 People's Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214061, P. R. China
| | - Yaolei Wang
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, P. R. China
| |
Collapse
|
3
|
Fritschen A, Lindner N, Scholpp S, Richthof P, Dietz J, Linke P, Guttenberg Z, Blaeser A. High-Scale 3D-Bioprinting Platform for the Automated Production of Vascularized Organs-on-a-Chip. Adv Healthc Mater 2024; 13:e2304028. [PMID: 38511587 PMCID: PMC11469029 DOI: 10.1002/adhm.202304028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/18/2024] [Indexed: 03/22/2024]
Abstract
3D bioprinting possesses the potential to revolutionize contemporary methodologies for fabricating tissue models employed in pharmaceutical research and experimental investigations. This is enhanced by combining bioprinting with advanced organs-on-a-chip (OOCs), which includes a complex arrangement of multiple cell types representing organ-specific cells, connective tissue, and vasculature. However, both OOCs and bioprinting so far demand a high degree of manual intervention, thereby impeding efficiency and inhibiting scalability to meet technological requirements. Through the combination of drop-on-demand bioprinting with robotic handling of microfluidic chips, a print procedure is achieved that is proficient in managing three distinct tissue models on a chip within only a minute, as well as capable of consecutively processing numerous OOCs without manual intervention. This process rests upon the development of a post-printing sealable microfluidic chip, that is compatible with different types of 3D-bioprinters and easily connected to a perfusion system. The capabilities of the automized bioprint process are showcased through the creation of a multicellular and vascularized liver carcinoma model on the chip. The process achieves full vascularization and stable microvascular network formation over 14 days of culture time, with pronounced spheroidal cell growth and albumin secretion of HepG2 serving as a representative cell model.
Collapse
Affiliation(s)
- Anna Fritschen
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Nils Lindner
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Sebastian Scholpp
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Philipp Richthof
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Jonas Dietz
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | | | | | - Andreas Blaeser
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
- Centre for Synthetic BiologyTechnical University of Darmstadt64289DarmstadtGermany
| |
Collapse
|
4
|
Bloise N, Giannaccari M, Guagliano G, Peluso E, Restivo E, Strada S, Volpini C, Petrini P, Visai L. Growing Role of 3D In Vitro Cell Cultures in the Study of Cellular and Molecular Mechanisms: Short Focus on Breast Cancer, Endometriosis, Liver and Infectious Diseases. Cells 2024; 13:1054. [PMID: 38920683 PMCID: PMC11201503 DOI: 10.3390/cells13121054] [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: 04/16/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Over the past decade, the development of three-dimensional (3D) models has increased exponentially, facilitating the unravelling of fundamental and essential cellular mechanisms by which cells communicate with each other, assemble into tissues and organs and respond to biochemical and biophysical stimuli under both physiological and pathological conditions. This section presents a concise overview of the most recent updates on the significant contribution of different types of 3D cell cultures including spheroids, organoids and organ-on-chip and bio-printed tissues in advancing our understanding of cellular and molecular mechanisms. The case studies presented include the 3D cultures of breast cancer (BC), endometriosis, the liver microenvironment and infections. In BC, the establishment of 3D culture models has permitted the visualization of the role of cancer-associated fibroblasts in the delivery of exosomes, as well as the significance of the physical properties of the extracellular matrix in promoting cell proliferation and invasion. This approach has also become a valuable tool in gaining insight into general and specific mechanisms of drug resistance. Given the considerable heterogeneity of endometriosis, 3D models offer a more accurate representation of the in vivo microenvironment, thereby facilitating the identification and translation of novel targeted therapeutic strategies. The advantages provided by 3D models of the hepatic environment, in conjunction with the high throughput characterizing various platforms, have enabled the elucidation of complex molecular mechanisms underlying various threatening hepatic diseases. A limited number of 3D models for gut and skin infections have been developed. However, a more profound comprehension of the spatial and temporal interactions between microbes, the host and their environment may facilitate the advancement of in vitro, ex vivo and in vivo disease models. Additionally, it may pave the way for the development of novel therapeutic approaches in diverse research fields. The interested reader will also find concluding remarks on the challenges and prospects of using 3D cell cultures for discovering cellular and molecular mechanisms in the research areas covered in this review.
Collapse
Affiliation(s)
- Nora Bloise
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
- UOR6 Nanotechnology Laboratory, Department of Prevention and Rehabilitation in Occupational Medicine and Specialty Medicine, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), Operative Unit (OU) of University of Pavia, 27100 Pavia, Italy
| | - Marialaura Giannaccari
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
| | - Giuseppe Guagliano
- Department of Chemistry, Materials, and Chemical Engineering “G. Natta”, Politecnico di Milano, P.zza L. Da Vinci 32, 20133 Milan, Italy; (G.G.); (P.P.)
| | - Emanuela Peluso
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
| | - Elisa Restivo
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
| | - Silvia Strada
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
- UOR6 Nanotechnology Laboratory, Department of Prevention and Rehabilitation in Occupational Medicine and Specialty Medicine, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
| | - Cristina Volpini
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
- UOR6 Nanotechnology Laboratory, Department of Prevention and Rehabilitation in Occupational Medicine and Specialty Medicine, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
| | - Paola Petrini
- Department of Chemistry, Materials, and Chemical Engineering “G. Natta”, Politecnico di Milano, P.zza L. Da Vinci 32, 20133 Milan, Italy; (G.G.); (P.P.)
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), Operative Unit (OU) of Politecnico di Milano, 20133 Milan, Italy
| | - Livia Visai
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
- UOR6 Nanotechnology Laboratory, Department of Prevention and Rehabilitation in Occupational Medicine and Specialty Medicine, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), Operative Unit (OU) of University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
5
|
Simińska-Stanny J, Nicolas L, Chafai A, Jafari H, Hajiabbas M, Dodi G, Gardikiotis I, Delporte C, Nie L, Podstawczyk D, Shavandi A. Advanced PEG-tyramine biomaterial ink for precision engineering of perfusable and flexible small-diameter vascular constructs via coaxial printing. Bioact Mater 2024; 36:168-184. [PMID: 38463551 PMCID: PMC10924180 DOI: 10.1016/j.bioactmat.2024.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 02/09/2024] [Accepted: 02/16/2024] [Indexed: 03/12/2024] Open
Abstract
Vascularization is crucial for providing nutrients and oxygen to cells while removing waste. Despite advances in 3D-bioprinting, the fabrication of structures with void spaces and channels remains challenging. This study presents a novel approach to create robust yet flexible and permeable small (600-1300 μm) artificial vessels in a single processing step using 3D coaxial extrusion printing of a biomaterial ink, based on tyramine-modified polyethylene glycol (PEG-Tyr). We combined the gelatin biocompatibility/activity, robustness of PEG-Tyr and alginate with the shear-thinning properties of methylcellulose (MC) in a new biomaterial ink for the fabrication of bioinspired vessels. Chemical characterization using NMR and FTIR spectroscopy confirmed the successful modification of PEG with Tyr and rheological characterization indicated that the addition of PEG-Tyr decreased the viscosity of the ink. Enzyme-mediated crosslinking of PEG-Tyr allowed the formation of covalent crosslinks within the hydrogel chains, ensuring its stability. PEG-Tyr units improved the mechanical properties of the material, resulting in stretchable and elastic constructs without compromising cell viability and adhesion. The printed vessel structures displayed uniform wall thickness, shape retention, improved elasticity, permeability, and colonization by endothelial-derived - EA.hy926 cells. The chorioallantoic membrane (CAM) and in vivo assays demonstrated the hydrogel's ability to support neoangiogenesis. The hydrogel material with PEG-Tyr modification holds promise for vascular tissue engineering applications, providing a flexible, biocompatible, and functional platform for the fabrication of vascular structures.
Collapse
Affiliation(s)
- Julia Simińska-Stanny
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| | - Lise Nicolas
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
- European School of Materials Science and Engineering, University of Lorraine, Nancy, France
| | - Adam Chafai
- Université Libre de Bruxelles (ULB), Micro-milli Platform, Avenue F.D. Roosevelt, 50 - CP 165/67, 1050, Brussels, Belgium
| | - Hafez Jafari
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| | - Maryam Hajiabbas
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
- Université Libre de Bruxelles (ULB), Faculté de Médecine, Campus Erasme - CP 611, Laboratory of Pathophysiological and Nutritional Biochemistry, Route de Lennik, 808, 1070, Bruxelles, Belgium
| | - Gianina Dodi
- Faculty of Medical Bioengineering, Grigore T. Popa, University of Medicine and Pharmacy of Iasi, Romania
| | - Ioannis Gardikiotis
- Advanced Research and Development Center for Experimental Medicine, Grigore T. Popa, University of Medicine and Pharmacy of Iasi, Romania
| | - Christine Delporte
- Université Libre de Bruxelles (ULB), Faculté de Médecine, Campus Erasme - CP 611, Laboratory of Pathophysiological and Nutritional Biochemistry, Route de Lennik, 808, 1070, Bruxelles, Belgium
| | - Lei Nie
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
- College of Life Science, Xinyang Normal University, Xinyang, China
| | - Daria Podstawczyk
- Department of Process Engineering and Technology of Polymer and Carbon Materials, Faculty of Chemistry, Wroclaw University of Science and Technology, Norwida 4/6, 50-373, Wroclaw, Poland
| | - Amin Shavandi
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| |
Collapse
|
6
|
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
|
7
|
Bhatt S S, Krishna Kumar J, Laya S, Thakur G, Nune M. Scaffold-mediated liver regeneration: A comprehensive exploration of current advances. J Tissue Eng 2024; 15:20417314241286092. [PMID: 39411269 PMCID: PMC11475092 DOI: 10.1177/20417314241286092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/08/2024] [Indexed: 10/19/2024] Open
Abstract
The liver coordinates over 500 biochemical processes crucial for maintaining homeostasis, detoxification, and metabolism. Its specialized cells, arranged in hexagonal lobules, enable it to function as a highly efficient metabolic engine. However, diseases such as cirrhosis, fatty liver disease, and hepatitis present significant global health challenges. Traditional drug development is expensive and often ineffective at predicting human responses, driving interest in advanced in vitro liver models utilizing 3D bioprinting and microfluidics. These models strive to mimic the liver's complex microenvironment, improving drug screening and disease research. Despite its resilience, the liver is vulnerable to chronic illnesses, injuries, and cancers, leading to millions of deaths annually. Organ shortages hinder liver transplantation, highlighting the need for alternative treatments. Tissue engineering, employing polymer-based scaffolds and 3D bioprinting, shows promise. This review examines these innovative strategies, including liver organoids and liver tissue-on-chip technologies, to address the challenges of liver diseases.
Collapse
Affiliation(s)
- Supriya Bhatt S
- Manipal Institute of Regenerative Medicine, Bengaluru, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Jayanthi Krishna Kumar
- Manipal Institute of Regenerative Medicine, Bengaluru, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Shurthi Laya
- Manipal Institute of Regenerative Medicine, Bengaluru, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
- Department of Biomedical Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Goutam Thakur
- Department of Biomedical Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Manasa Nune
- Manipal Institute of Regenerative Medicine, Bengaluru, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
8
|
Di Gravina GM, Bari E, Croce S, Scocozza F, Pisani S, Conti B, Avanzini MA, Auricchio F, Cobianchi L, Torre ML, Conti M. Design and development of a hepatic lyo-dECM powder as a biomimetic component for 3D-printable hybrid hydrogels. Biomed Mater 2023; 19:015005. [PMID: 37992318 DOI: 10.1088/1748-605x/ad0ee2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/22/2023] [Indexed: 11/24/2023]
Abstract
Bioprinting offers new opportunities to obtain reliable 3Din vitromodels of the liver for testing new drugs and studying pathophysiological mechanisms, thanks to its main feature in controlling the spatial deposition of cell-laden hydrogels. In this context, decellularized extracellular matrix (dECM)-based hydrogels have caught more and more attention over the last years because of their characteristic to closely mimic the tissue-specific microenvironment from a biological point of view. In this work, we describe a new concept of designing dECM-based hydrogels; in particular, we set up an alternative and more practical protocol to develop a hepatic lyophilized dECM (lyo-dECM) powder as an 'off-the-shelf' and free soluble product to be incorporated as a biomimetic component in the design of 3D-printable hybrid hydrogels. To this aim, the powder was first characterized in terms of cytocompatibility on human and porcine mesenchymal stem cells (MSCs), and the optimal powder concentration (i.e. 3.75 mg ml-1) to use in the hydrogel formulation was identified. Moreover, its non-immunogenicity and capacity to reactivate the elastase enzyme potency was proved. Afterward, as a proof-of-concept, the powder was added to a sodium alginate/gelatin blend, and the so-defined multi-component hydrogel was studied from a rheological point of view, demonstrating that adding the lyo-dECM powder at the selected concentration did not alter the viscoelastic properties of the original material. Then, a printing assessment was performed with the support of computational simulations, which were useful to definea priorithe hydrogel printing parameters as window of printability and its post-printing mechanical collapse. Finally, the proposed multi-component hydrogel was bioprinted with cells inside, and its post-printing cell viability for up to 7 d was successfully demonstrated.
Collapse
Affiliation(s)
- Giulia M Di Gravina
- Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy
- Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy
| | - Elia Bari
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Stefania Croce
- Department of General Surgery, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Franca Scocozza
- Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy
| | - Silvia Pisani
- Department of Drug Science, University of Pavia, Pavia, Italy
| | - Bice Conti
- Department of Drug Science, University of Pavia, Pavia, Italy
| | - Maria A Avanzini
- Pediatric Hematology Oncology Unit and Cell Factory, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Ferdinando Auricchio
- Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy
| | - Lorenzo Cobianchi
- Department of General Surgery, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic & Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Maria Luisa Torre
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
- PharmaExceed s.r.l., Pavia, Italy
| | - Michele Conti
- Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy
| |
Collapse
|
9
|
Utagawa Y, Ino K, Takinoue M, Shiku H. Fabrication and Cell Culture Applications of Core-Shell Hydrogel Fibers Composed of Chitosan/DNA Interfacial Polyelectrolyte Complexation and Calcium Alginate: Straight and Beaded Core Variations. Adv Healthc Mater 2023; 12:e2302011. [PMID: 37478383 PMCID: PMC11468996 DOI: 10.1002/adhm.202302011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Indexed: 07/23/2023]
Abstract
Core-shell hydrogel fibers are widely used in cell culture applications. A simple and rapid method is presented for fabricating core-shell hydrogel fibers, consisting of straight or beaded core fibers, for cell culture applications. The core fibers are prepared using interfacial polyelectrolyte complexation (IPC) with chitosan and DNA. Briefly, two droplets of chitosan and DNA are brought in contact to form an IPC film, which is dragged to prepare an IPC fiber. The incubation time and DNA concentration are adjusted to prepare straight and beaded IPC fibers. The fibers with Ca2+ are immersed in an alginate solution to form calcium alginate shell hydrogels around the core IPC fibers. To the best of the knowledge, this is the first report of core-shell hydrogel fibers with IPC fiber cores. To demonstrate cell culture, straight hydrogel fibers are applied to fabricate hepatic models consisting of HepG2 and 3T3 fibroblasts, and vascular models consisting of human umbilical vein endothelial cells and 3T3 fibroblasts. To evaluate the effect of co-culture, albumin secretion, and angiogenesis are evaluated. Beaded hydrogel fibers are used to fabricate many size-controlled spheroids for fiber and cloning applications. This method can be widely applied in tissue engineering and cell analysis.
Collapse
Affiliation(s)
| | - Kosuke Ino
- Graduate School of EngineeringTohoku UniversitySendai980–8579Japan
| | - Masahiro Takinoue
- Department of Computer ScienceTokyo Institute of TechnologyYokohama226–8502Japan
| | - Hitoshi Shiku
- Graduate School of EngineeringTohoku UniversitySendai980–8579Japan
- Graduate School of Environmental StudiesTohoku UniversitySendai980–8579Japan
| |
Collapse
|
10
|
Joshi A, Singh N. Generation of Patterned Cocultures in 2D and 3D: State of the Art. ACS OMEGA 2023; 8:34249-34261. [PMID: 37780002 PMCID: PMC10536108 DOI: 10.1021/acsomega.3c02713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/30/2023] [Indexed: 10/03/2023]
Abstract
Cells inside the body are embedded into a highly structured microenvironment that consists of cells that lie in direct or close contact with other cell types that regulate the overall tissue function. Therefore, coculture models are versatile tools that can generate tissue engineering constructs with improved mimicking of in vivo conditions. While there are many reviews that have focused on pattering a single cell type, very few reviews have been focused on techniques for coculturing multiple cell types on a single substrate with precise control. In this regard, this Review covers various technologies that have been utilized for the development of these patterned coculture models while mentioning the limitations associated with each of them. Further, the application of these models to various tissue engineering applications has been discussed.
Collapse
Affiliation(s)
- Akshay Joshi
- Centre
for Biomedical Engineering, Indian Institute
of Technology Delhi, Hauz Khas, New Delhi, Delhi 110016, India
| | - Neetu Singh
- Centre
for Biomedical Engineering, Indian Institute
of Technology Delhi, Hauz Khas, New Delhi, Delhi 110016, India
- Biomedical
Engineering Unit, All India Institute of
Medical Sciences, Ansari
Nagar, New Delhi, Delhi 110029, India
| |
Collapse
|
11
|
Windisch J, Reinhardt O, Duin S, Schütz K, Rodriguez NJN, Liu S, Lode A, Gelinsky M. Bioinks for Space Missions: The Influence of Long-Term Storage of Alginate-Methylcellulose-Based Bioinks on Printability as well as Cell Viability and Function. Adv Healthc Mater 2023; 12:e2300436. [PMID: 37125819 PMCID: PMC11468998 DOI: 10.1002/adhm.202300436] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/14/2023] [Indexed: 05/02/2023]
Abstract
Bioprinting is considered a key technology for future space missions and is currently being established on the International Space Station (ISS). With the aim to perform bioink production as a critical and resource-consuming preparatory step already on Earth and transport a bioink cartridge "ready to use" to the ISS, the storability of bioinks is investigated. Hydrogel blends based on alginate and methylcellulose are laden with either green microalgae of the species Chlorella vulgaris or with different human cell lines including immortilized human mesenchymal stem cells, SaOS-2 and HepG2, as well as with primary human dental pulp stem cells. The bioinks are filled into printing cartridges and stored at 4°C for up to four weeks. Printability of the bioinks is maintained after storage. Viability and function of the cells embedded in constructs bioprinted from the stored bioinks are investigated during subsequent cultivation: The microalgae survive the storage period very well and show no loss of growth and functionality, however a significant decrease is visible for human cells, varying between the different cell types. The study demonstrates that storage of bioinks is in principle possible and is a promising starting point for future research, making complex printing processes more effective and reproducible.
Collapse
Affiliation(s)
- Johannes Windisch
- Centre for Translational Bone, Joint and Soft Tissue ResearchFaculty of MedicineTU DresdenFetscherstrasse 7401307DresdenGermany
| | - Olena Reinhardt
- Centre for Translational Bone, Joint and Soft Tissue ResearchFaculty of MedicineTU DresdenFetscherstrasse 7401307DresdenGermany
| | - Sarah Duin
- Centre for Translational Bone, Joint and Soft Tissue ResearchFaculty of MedicineTU DresdenFetscherstrasse 7401307DresdenGermany
| | - Kathleen Schütz
- Centre for Translational Bone, Joint and Soft Tissue ResearchFaculty of MedicineTU DresdenFetscherstrasse 7401307DresdenGermany
| | - Nuria Juliana Novoa Rodriguez
- Centre for Translational Bone, Joint and Soft Tissue ResearchFaculty of MedicineTU DresdenFetscherstrasse 7401307DresdenGermany
| | - Suihong Liu
- Centre for Translational Bone, Joint and Soft Tissue ResearchFaculty of MedicineTU DresdenFetscherstrasse 7401307DresdenGermany
| | - Anja Lode
- Centre for Translational Bone, Joint and Soft Tissue ResearchFaculty of MedicineTU DresdenFetscherstrasse 7401307DresdenGermany
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue ResearchFaculty of MedicineTU DresdenFetscherstrasse 7401307DresdenGermany
| |
Collapse
|
12
|
Li W, Liu Z, Tang F, Jiang H, Zhou Z, Hao X, Zhang JM. Application of 3D Bioprinting in Liver Diseases. MICROMACHINES 2023; 14:1648. [PMID: 37630184 PMCID: PMC10457767 DOI: 10.3390/mi14081648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/03/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023]
Abstract
Liver diseases are the primary reason for morbidity and mortality in the world. Owing to a shortage of organ donors and postoperative immune rejection, patients routinely suffer from liver failure. Unlike 2D cell models, animal models, and organoids, 3D bioprinting can be successfully employed to print living tissues and organs that contain blood vessels, bone, and kidney, heart, and liver tissues and so on. 3D bioprinting is mainly classified into four types: inkjet 3D bioprinting, extrusion-based 3D bioprinting, laser-assisted bioprinting (LAB), and vat photopolymerization. Bioinks for 3D bioprinting are composed of hydrogels and cells. For liver 3D bioprinting, hepatic parenchymal cells (hepatocytes) and liver nonparenchymal cells (hepatic stellate cells, hepatic sinusoidal endothelial cells, and Kupffer cells) are commonly used. Compared to conventional scaffold-based approaches, marked by limited functionality and complexity, 3D bioprinting can achieve accurate cell settlement, a high resolution, and more efficient usage of biomaterials, better mimicking the complex microstructures of native tissues. This method will make contributions to disease modeling, drug discovery, and even regenerative medicine. However, the limitations and challenges of this method cannot be ignored. Limitation include the requirement of diverse fabrication technologies, observation of drug dynamic response under perfusion culture, the resolution to reproduce complex hepatic microenvironment, and so on. Despite this, 3D bioprinting is still a promising and innovative biofabrication strategy for the creation of artificial multi-cellular tissues/organs.
Collapse
Affiliation(s)
- Wenhui Li
- Department of Radiology, Yancheng Third People’s Hospital, Affiliated Hospital 6 of Nantong University, Yancheng 224000, China
| | - Zhaoyue Liu
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics; Nanjing 210016, China
| | - Fengwei Tang
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics; Nanjing 210016, China
| | - Hao Jiang
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics; Nanjing 210016, China
| | - Zhengyuan Zhou
- Nanjing Hangdian Intelligent Manufacturing Technology Co., Ltd., Nanjing 210014, China
| | - Xiuqing Hao
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics; Nanjing 210016, China
| | - Jia Ming Zhang
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics; Nanjing 210016, China
- Nanjing Hangdian Intelligent Manufacturing Technology Co., Ltd., Nanjing 210014, China
- Yangtze River Delta Intelligent Manufacturing Innovation Center, Nanjing 210014, China
| |
Collapse
|
13
|
Maji S, Lee M, Lee J, Lee J, Lee H. Development of lumen-based perfusable 3D liver in vitro model using single-step bioprinting with composite bioinks. Mater Today Bio 2023; 21:100723. [PMID: 37502830 PMCID: PMC10368928 DOI: 10.1016/j.mtbio.2023.100723] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/07/2023] [Accepted: 07/06/2023] [Indexed: 07/29/2023] Open
Abstract
Hepatic sinusoids are uniquely organized structures that help maintain a spectrum of hepatic functions. Although several in vitro liver models have been developed to replicate liver sinusoids, most of these platforms require complex, multi-step fabrication methods making it difficult to achieve truly three-dimensional (3D) channel geometries. In this study, a single-step bioprinting technique was demonstrated to simultaneously print a chip platform and develop a perfusable vascularized liver sinusoid in vitro model. The integrated system uses a co-axial-based bioprinting approach to develop a liver sinusoid-like model that consists of a sacrificial core compartment containing a perfusable pre-vascular structure and an alginate-collagen-based shell compartment containing hepatocytes. The lumen-based perfusable 3D liver sinusoid-on-a-chip (LSOC-P) demonstrated significantly better hepatocyte viability, proliferation, and liver-specific gene and protein expression compared to a 3D hepatocyte-based core/shell model with static media and the standard hepatocyte-based 2D sandwich culture system. A drug toxicity evaluation of hepatotoxins highlighted the comparatively higher sensitivity of the LSOC system with a close estimation of the therapeutic range of safe drug concentrations for humans. In conclusion, the current findings indicate that the combinatorial single-step co-axial bioprinting technique is a promising fabrication approach for the development of a perfusable LSOC platform for drug screening applications.
Collapse
Affiliation(s)
- Somnath Maji
- Department of Mechanical and Biomedical Engineering, Kangwon National University, Chuncheon, Republic of Korea
| | - Minkyoung Lee
- Department of Animal Industry Convergence, Kangwon National University, Chuncheon, Republic of Korea
- Department of Smart Health Science and Technology, Kangwon National University, Chuncheon, Republic of Korea
| | - Jooyoung Lee
- Department of Smart Health Science and Technology, Kangwon National University, Chuncheon, Republic of Korea
| | - Jaehee Lee
- Department of Smart Health Science and Technology, Kangwon National University, Chuncheon, Republic of Korea
| | - Hyungseok Lee
- Department of Mechanical and Biomedical Engineering, Kangwon National University, Chuncheon, Republic of Korea
- Department of Smart Health Science and Technology, Kangwon National University, Chuncheon, Republic of Korea
| |
Collapse
|
14
|
Xu KL, Mauck RL, Burdick JA. Modeling development using hydrogels. Development 2023; 150:dev201527. [PMID: 37387575 PMCID: PMC10323241 DOI: 10.1242/dev.201527] [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] [Indexed: 07/01/2023]
Abstract
The development of multicellular complex organisms relies on coordinated signaling from the microenvironment, including both biochemical and mechanical interactions. To better understand developmental biology, increasingly sophisticated in vitro systems are needed to mimic these complex extracellular features. In this Primer, we explore how engineered hydrogels can serve as in vitro culture platforms to present such signals in a controlled manner and include examples of how they have been used to advance our understanding of developmental biology.
Collapse
Affiliation(s)
- Karen L. Xu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert L. Mauck
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104, USA
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
| |
Collapse
|
15
|
Juraski AC, Sharma S, Sparanese S, da Silva VA, Wong J, Laksman Z, Flannigan R, Rohani L, Willerth SM. 3D bioprinting for organ and organoid models and disease modeling. Expert Opin Drug Discov 2023; 18:1043-1059. [PMID: 37431937 DOI: 10.1080/17460441.2023.2234280] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 07/05/2023] [Indexed: 07/12/2023]
Abstract
INTRODUCTION 3D printing, a versatile additive manufacturing technique, has diverse applications ranging from transportation, rapid prototyping, clean energy, and medical devices. AREAS COVERED The authors focus on how 3D printing technology can enhance the drug discovery process through automating tissue production that enables high-throughput screening of potential drug candidates. They also discuss how the 3D bioprinting process works and what considerations to address when using this technology to generate cell laden constructs for drug screening as well as the outputs from such assays necessary for determining the efficacy of potential drug candidates. They focus on how bioprinting how has been used to generate cardiac, neural, and testis tissue models, focusing on bio-printed 3D organoids. EXPERT OPINION The next generation of 3D bioprinted organ model holds great promises for the field of medicine. In terms of drug discovery, the incorporation of smart cell culture systems and biosensors into 3D bioprinted models could provide highly detailed and functional organ models for drug screening. By addressing current challenges of vascularization, electrophysiological control, and scalability, researchers can obtain more reliable and accurate data for drug development, reducing the risk of drug failures during clinical trials.
Collapse
Affiliation(s)
- Amanda C Juraski
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria BC, Canada
- Department of Chemical Engineering, Polytechnic School, University of Sao Paulo, Sao Paulo, Brazil
| | - Sonali Sharma
- Faculty of Medicine, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Sydney Sparanese
- Faculty of Medicine, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver BC, Canada
| | - Victor A da Silva
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria BC, Canada
| | - Julie Wong
- Department of Urologic Sciences, University of British Columbia, Vancouver BC, Canada
| | - Zachary Laksman
- Faculty of Medicine, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Ryan Flannigan
- Department of Urologic Sciences, University of British Columbia, Vancouver BC, Canada
| | - Leili Rohani
- Faculty of Medicine, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Stephanie M Willerth
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria BC, Canada
- Faculty of Medicine, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
| |
Collapse
|
16
|
Saemundsson SA, Ganguly S, Curry SD, Goodwin AP, Cha JN. Controlling Cell Organization in 3D Coculture Spheroids Using DNA Interactions. ACS Biomater Sci Eng 2023. [PMID: 37155244 DOI: 10.1021/acsbiomaterials.3c00546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
The role of stromal and immune cells in transforming the tumor microenvironment is a key consideration in understanding tumor cell behavior and anticancer drug development. To better model these systems in vitro, 3D coculture tumor spheroids have been engineered using a variety of techniques including centrifugation to microwells, hanging drop, low adhesion cultures, and culture of cells in a microfluidic platform. Aside from using bioprinting, however, it has remained more challenging to direct the spatial organization of heterotypic cells in standalone 3D spheroids. To address this, we present an in vitro 3D coculture tumor model where we modulated the interactions between cancer cells and fibroblasts through DNA hybridization. When native heterotypic cells are simply mixed, the cell aggregates typically show cell sorting behavior to form phase separated structures composed of single cell types. In this work, we demonstrate that when MDA-MB-468 breast cancer and NIH/3T3 fibroblasts are directed to associate via complementary DNA, a uniform distribution of the two cell types within a single spheroid was observed. In contrast, in the absence of specific DNA interactions between the cancer cells and fibroblasts, individual clusters of the NIH/3T3 cells formed in each spheroid due to cell sorting. To better understand the effect of heterotypic cell organization on either cell-cell contacts or matrix protein production, the spheroids were further stained with anti-E-cadherin and antifibronectin antibodies. While the amounts of E-cadherin appeared to be similar between the spheroids, a significantly higher amount of fibronectin secretion was observed in the coculture spheroids with uniform mixing of two cell types. This result showed that different heterotypic cell distributions within 3D architecture can influence the ECM protein production that can again alter the properties of the tumor or tumor microenvironment. The present study thus describes the use of DNA templating to direct the organization of cells in coculture spheroids, which can provide mechanistic biological insight into how heterotypic distribution in tumor spheroids can influence tumor progression, metastasis, and drug resistance.
Collapse
Affiliation(s)
- Sven A Saemundsson
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, 596 UCB, Boulder, Colorado 80303, United States
| | - Saheli Ganguly
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, 596 UCB, Boulder, Colorado 80303, United States
| | - Shane D Curry
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, 596 UCB, Boulder, Colorado 80303, United States
| | - Andrew P Goodwin
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, 596 UCB, Boulder, Colorado 80303, United States
- Materials Science and Engineering Program, University of Colorado, Boulder, 596 UCB, Boulder, Colorado 80303, United States
| | - Jennifer N Cha
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, 596 UCB, Boulder, Colorado 80303, United States
- Materials Science and Engineering Program, University of Colorado, Boulder, 596 UCB, Boulder, Colorado 80303, United States
- Biomedical Engineering Program, University of Colorado, Boulder, 596 UCB, Boulder, Colorado 80303, United States
| |
Collapse
|
17
|
Ahmadian M, Hosseini S, Alipour A, Jahanfar M, Farrokhi N, Homaeigohar S, Shahsavarani H. In vitro modeling of hepatocellular carcinoma niche on decellularized tomato thorny leaves: a novel natural three-dimensional (3D) scaffold for liver cancer therapeutics. Front Bioeng Biotechnol 2023; 11:1189726. [PMID: 37251569 PMCID: PMC10212619 DOI: 10.3389/fbioe.2023.1189726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Liver cancer is now one of the main causes leading to death worldwide. To achieve reliable therapeutic effects, it is crucial to develop efficient approaches to test novel anticancer drugs. Considering the significant contribution of tumor microenvironment to cell's response to medications, in vitro 3D bioinspiration of cancer cell niches can be regarded as an advanced strategy to improve the accuracy and reliability of the drug-based treatment. In this regard, decellularized plant tissues can perform as suitable 3D scaffolds for mammalian cell culture to create a near-to-real condition to test drug efficacy. Here, we developed a novel 3D natural scaffold made from decellularized tomato hairy leaves (hereafter called as DTL) to mimic the microenvironment of human hepatocellular carcinoma (HCC) for pharmaceutical purposes. The surface hydrophilicity, mechanical properties, and topography measurement and molecular analyses revealed that the 3D DTL scaffold is an ideal candidate for liver cancer modeling. The cells exhibited a higher growth and proliferation rate within the DTL scaffold, as verified by quantifying the expression of related genes, DAPI staining, and SEM imaging of the cells. Moreover, prilocaine, an anticancer drug, showed a higher effectiveness against the cancer cells cultured on the 3D DTL scaffold, compared to a 2D platform. Taken together, this new cellulosic 3D scaffold can be confidently proposed for chemotherapeutic testing of drugs on hepatocellular carcinoma.
Collapse
Affiliation(s)
- Mariye Ahmadian
- Department of Cell and Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
- Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, National Cell Bank, Tehran, Iran
| | - Saadi Hosseini
- Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, National Cell Bank, Tehran, Iran
| | - Atefeh Alipour
- Department of Nanobiotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Mehdi Jahanfar
- Department of Cell and Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Naser Farrokhi
- Department of Cell and Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Shahin Homaeigohar
- School of Science and Engineering, University of Dundee, Dundee, United Kingdom
| | - Hosein Shahsavarani
- Department of Cell and Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
- Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, National Cell Bank, Tehran, Iran
| |
Collapse
|
18
|
Modification, 3D printing process and application of sodium alginate based hydrogels in soft tissue engineering: A review. Int J Biol Macromol 2023; 232:123450. [PMID: 36709808 DOI: 10.1016/j.ijbiomac.2023.123450] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/26/2022] [Accepted: 01/24/2023] [Indexed: 01/27/2023]
Abstract
Sodium alginate (SA) is an inexpensive and biocompatible biomaterial with fast and gentle crosslinking that has been widely used in biological soft tissue repair/regeneration. Especially with the advent of 3D bioprinting technology, SA hydrogels have been applied more deeply in tissue engineering due to their excellent printability. Currently, the research on material modification, molding process and application of SA-based composite hydrogels has become a hot topic in tissue engineering, and a lot of fruitful results have been achieved. To better help readers have a comprehensive understanding of the development status of SA based hydrogels and their molding process in tissue engineering, in this review, we summarized SA modification methods, and provided a comparative analysis of the characteristics of various SA based hydrogels. Secondly, various molding methods of SA based hydrogels were introduced, the processing characteristics and the applications of different molding methods were analyzed and compared. Finally, the applications of SA based hydrogels in tissue engineering were reviewed, the challenges in their applications were also analyzed, and the future research directions were prospected. We believe this review is of great helpful for the researchers working in biomedical and tissue engineering.
Collapse
|
19
|
Ma Y, Hu L, Tang J, Guo W, Feng Y, Liu Y, Tang F. Three-Dimensional Cell Co-Culture Liver Models and Their Applications in Pharmaceutical Research. Int J Mol Sci 2023; 24:ijms24076248. [PMID: 37047220 PMCID: PMC10094553 DOI: 10.3390/ijms24076248] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/14/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
As the primary site for the biotransformation of drugs, the liver is the most focused on organ type in pharmaceutical research. However, despite being widely used in pharmaceutical research, animal models have inherent species differences, while two-dimensional (2D) liver cell monocultures or co-cultures and three-dimensional (3D) liver cell monoculture in vitro liver models do not sufficiently represent the complexity of the human liver’s structure and function, making the evaluation results from these tools less reliable. Therefore, there is a pressing need to develop more representative in vitro liver models for pharmaceutical research. Fortunately, an exciting new development in recent years has been the emergence of 3D liver cell co-culture models. These models hold great promise as in vitro pharmaceutical research tools, because they can reproduce liver structure and function more practically. This review begins by explaining the structure and main cell composition of the liver, before introducing the potential advantages of 3D cell co-culture liver models for pharmaceutical research. We also discuss the main sources of hepatocytes and the 3D cell co-culture methods used in constructing these models. In addition, we explore the applications of 3D cell co-culture liver models with different functional states and suggest prospects for their further development.
Collapse
|
20
|
Prashantha K, Krishnappa A, Muthappa M. 3D bioprinting of gastrointestinal cancer models: A comprehensive review on processing, properties, and therapeutic implications. Biointerphases 2023; 18:020801. [PMID: 36963961 DOI: 10.1116/6.0002372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2023] Open
Abstract
Gastrointestinal tract (GIT) malignancies are an important public health problem considering the increased incidence in recent years and the high morbidity and mortality associated with it. GIT malignancies constitute 26% of the global cancer incidence burden and 35% of all cancer-related deaths. Gastrointestinal cancers are complex and heterogenous diseases caused by the interplay of genetic and environmental factors. The tumor microenvironment (TME) of gastrointestinal tract carcinomas is dynamic and complex; it cannot be recapitulated in the basic two-dimensional cell culture systems. In contrast, three-dimensional (3D) in vitro models can mimic the TME more closely, enabling an improved understanding of the microenvironmental cues involved in the various stages of cancer initiation, progression, and metastasis. However, the heterogeneity of the TME is incompletely reproduced in these 3D culture models, as they fail to regulate the orientation and interaction of various cell types in a complex architecture. To emulate the TME, 3D bioprinting has emerged as a useful technique to engineer cancer tissue models. Bioprinted cancer tissue models can potentially recapitulate cancer pathology and increase drug resistance in an organ-mimicking 3D environment. In this review, we describe the 3D bioprinting methods, bioinks, characterization of 3D bioprinted constructs, and their application in developing gastrointestinal tumor models that integrate their microenvironment with different cell types and substrates, as well as bioprinting modalities and their application in therapy and drug screening. We review prominent studies on the 3D bioprinted esophageal, hepatobiliary, and colorectal cancer models. In addition, this review provides a comprehensive understanding of the cancer microenvironment in printed tumor models, highlights current challenges with respect to their clinical translation, and summarizes future perspectives.
Collapse
Affiliation(s)
- Kalappa Prashantha
- Centre for Research and Innovation, Adichunchanagiri School of Natural Sciences, Adichunchanagiri University, BGSIT, B.G. Nagara, Mandya District 571448, Karnataka, India
| | - Amita Krishnappa
- Department of Pathology, Adichunchanagiri Institute of Medicinal Sciences Adichunchanagiri University, B.G. Nagara, Mandya District 571448, Karnataka, India
| | - Malini Muthappa
- Department of Physiology, Adichunchanagiri Institute of Medicinal Sciences Adichunchanagiri University, B.G. Nagara, Mandya District 571448, Karnataka, India
| |
Collapse
|
21
|
Patrício D, Santiago J, Mano JF, Fardilha M. Organoids of the male reproductive system: Challenges, opportunities, and their potential use in fertility research. WIREs Mech Dis 2023; 15:e1590. [PMID: 36442887 DOI: 10.1002/wsbm.1590] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/17/2022] [Accepted: 11/12/2022] [Indexed: 11/30/2022]
Abstract
Organoids are units of function of a given organ able to reproduce, in culture, a biological structure similar in architecture and function to its counterpart in vivo. Today, it is possible to develop an organoid from a fragment of tissue, a stem cell located in an adult organ, an embryonic stem cell, or an induced pluripotent stem cell. In the past decade, many organoids have been developed which mimic stomach, pancreas, liver and brain tissues, optic cups, among many others. Additionally, different male reproductive system organs have already been developed as organoids, including the prostate and testis. These 3D cultures may be of great importance for urological cancer research and have the potential to be used in fertility research for the study of spermatozoa production and maturation, germ cells-somatic cells interactions, and mechanisms of disease. They also provide an accurate preclinical pipeline for drug testing and discovery, as well as for the study of drug resistance. In this work, we revise the current knowledge on organoid technology and its use in healthcare and research, describe the male reproductive system organoids and other biomaterials already developed, and discuss their current application. Finally, we highlight the research gaps, challenges, and opportunities in the field and propose strategies to improve the use of organoids for the study of male infertility situations. This article is categorized under: Reproductive System Diseases > Stem Cells and Development Reproductive System Diseases > Biomedical Engineering.
Collapse
Affiliation(s)
- Daniela Patrício
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal.,Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Joana Santiago
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Margarida Fardilha
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
22
|
Liu S, Kilian D, Ahlfeld T, Hu Q, Gelinsky M. Egg white improves the biological properties of an alginate-methylcellulose bioink for 3D bioprinting of volumetric bone constructs. Biofabrication 2023; 15. [PMID: 36735961 DOI: 10.1088/1758-5090/acb8dc] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 02/03/2023] [Indexed: 02/05/2023]
Abstract
Three-dimensional microextrusion bioprinting has attracted great interest for fabrication of hierarchically structured, functional tissue substitutes with spatially defined cell distribution. Despite considerable progress, several significant limitations remain such as a lack of suitable bioinks which combine favorable cell response with high shape fidelity. Therefore, in this work a novel bioink of alginate-methylcellulose (AlgMC) blend functionalized with egg white (EW) was developed with the aim of solving this limitation. In this regard, a stepwise strategy was proposed to improve and examine the cell response in low-viscosity alginate inks (3%, w/v) with different EW concentrations, and in high-viscosity inks after gradual methylcellulose addition for enhancing printability. The rheological properties and printability of these cell-responsive bioinks were characterized to obtain an optimized formulation eliciting balanced physicochemical and biological properties for fabrication of volumetric scaffolds. The bioprinted AlgMC + EW constructs exhibited excellent shape fidelity while encapsulated human mesenchymal stem cells showed high post-printing viability as well as adhesion and spreading within the matrix. In a proof-of-concept experiment, the impact of these EW-mediated effects on osteogenesis of bioprinted primary human pre-osteoblasts (hOB) was evaluated. Results confirmed a high viability of hOB (93.7 ± 0.15%) post-fabrication in an EW-supported AlgMC bioink allowing cell adhesion, proliferation and migration. EW even promoted the expression of osteogenic genes, coding for bone sialoprotein (integrin binding sialoprotein/bone sialoprotein precursor (IBSP)) and osteocalcin (BGLAP) on mRNA level. To demonstrate the suitability of the novel ink for future fabrication of multi-zonal bone substitutes, AlgMC + EW was successfully co-printed together with a pasty calcium phosphate bone cement biomaterial ink to achieve a partly mineralized 3D volumetric environment with good cell viability and spreading. Along with the EW-mediated positive effects within bioprinted AlgMC-based scaffolds, this highlighted the promising potential of this novel ink for biofabrication of bone tissue substitutes in clinically relevant dimensions.
Collapse
Affiliation(s)
- Suihong Liu
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.,Rapid Manufacturing Engineering Center, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, People's Republic of China.,Shanghai Key Laboratory of Intelligent Manufacturing and Robotics, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, People's Republic of China
| | - David Kilian
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Tilman Ahlfeld
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Qingxi Hu
- Rapid Manufacturing Engineering Center, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, People's Republic of China.,Shanghai Key Laboratory of Intelligent Manufacturing and Robotics, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, People's Republic of China.,National Demonstration Center for Experimental Engineering Training Education, Shanghai University, Shanghai 200444, People's Republic of China
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| |
Collapse
|
23
|
Kaur I, Vasudevan A, Rawal P, Tripathi DM, Ramakrishna S, Kaur S, Sarin SK. Primary Hepatocyte Isolation and Cultures: Technical Aspects, Challenges and Advancements. Bioengineering (Basel) 2023; 10:131. [PMID: 36829625 PMCID: PMC9952008 DOI: 10.3390/bioengineering10020131] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/20/2023] Open
Abstract
Hepatocytes are differentiated cells that account for 80% of the hepatic volume and perform all major functions of the liver. In vivo, after an acute insult, adult hepatocytes retain their ability to proliferate and participate in liver regeneration. However, in vitro, prolonged culture and proliferation of viable and functional primary hepatocytes have remained the major and the most challenging goal of hepatocyte-based cell therapies and liver tissue engineering. The first functional cultures of rat primary hepatocytes between two layers of collagen gel, also termed as the "sandwich cultures", were reported in 1989. Since this study, several technical developments including choice of hydrogels, type of microenvironment, growth factors and culture conditions, mono or co-cultures of hepatocytes along with other supporting cell types have evolved for both rat and human primary hepatocytes in recent years. All these improvements have led to a substantial improvement in the number, life-span and hepatic functions of these cells in vitro for several downstream applications. In the current review, we highlight the details, limitations and prospects of different technical strategies being used in primary hepatocyte cultures. We discuss the use of newer biomaterials as scaffolds for efficient culture of primary hepatocytes. We also describe the derivation of mature hepatocytes from other cellular sources such as induced pluripotent stem cells, bone marrow stem cells and 3D liver organoids. Finally, we also explain the use of perfusion-based bioreactor systems and bioengineering strategies to support the long-term function of hepatocytes in 3D conditions.
Collapse
Affiliation(s)
- Impreet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Ashwini Vasudevan
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Preety Rawal
- School of Biotechnology, Gautam Buddha University, Greater Noida 201312, India
| | - Dinesh M. Tripathi
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Seeram Ramakrishna
- Department of Mechanical Engineering, National University of Singapore, Singapore 117581, Singapore
| | - Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Shiv K. Sarin
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| |
Collapse
|
24
|
Díaz‐Payno PJ, Kalogeropoulou M, Muntz I, Kingma E, Kops N, D'Este M, Koenderink GH, Fratila‐Apachitei LE, van Osch GJVM, Zadpoor AA. Swelling-Dependent Shape-Based Transformation of a Human Mesenchymal Stromal Cells-Laden 4D Bioprinted Construct for Cartilage Tissue Engineering. Adv Healthc Mater 2023; 12:e2201891. [PMID: 36308047 PMCID: PMC11468569 DOI: 10.1002/adhm.202201891] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/10/2022] [Indexed: 01/18/2023]
Abstract
3D bioprinting is usually implemented on flat surfaces, posing serious limitations in the fabrication of multilayered curved constructs. 4D bioprinting, combining 3D bioprinting with time-dependent stimuli-induced transformation, enables the fabrication of shape-changing constructs. Here, a 4D biofabrication method is reported for cartilage engineering based on the differential swelling of a smart multi-material system made from two hydrogel-based materials: hyaluronan and alginate. Two ink formulations are used: tyramine-functionalized hyaluronan (HAT, high-swelling) and alginate with HAT (AHAT, low-swelling). Both inks have similar elastic, shear-thinning, and printability behavior. The inks are 3D printed into a bilayered scaffold before triggering the shape-change by using liquid immersion as stimulus. In time (4D), the differential swelling between the two zones leads to the scaffold's self-bending. Different designs are made to tune the radius of curvature and shape. A bioprinted formulation of AHAT and human bone marrow cells demonstrates high cell viability. After 28 days in chondrogenic medium, the curvature is clearly present while cartilage-like matrix production is visible on histology. A proof-of-concept of the recently emerged technology of 4D bioprinting with a specific application for the design of curved structures potentially mimicking the curvature and multilayer cellular nature of native cartilage is demonstrated.
Collapse
Affiliation(s)
- Pedro J. Díaz‐Payno
- Department of Biomechanical EngineeringFaculty of MechanicalMaritime and Materials EngineeringDelft University of TechnologyDelft2628CDNetherlands
- Department of Orthopedics and Sports MedicineErasmus MC University Medical CenterRotterdam3015GDNetherlands
| | - Maria Kalogeropoulou
- Department of Biomechanical EngineeringFaculty of MechanicalMaritime and Materials EngineeringDelft University of TechnologyDelft2628CDNetherlands
| | - Iain Muntz
- Department of BionanoscienceKavli Institute of Nanoscience DelftDelft University of TechnologyDelft2628CDNetherlands
| | - Esther Kingma
- Department of Biomechanical EngineeringFaculty of MechanicalMaritime and Materials EngineeringDelft University of TechnologyDelft2628CDNetherlands
| | - Nicole Kops
- Department of Orthopedics and Sports MedicineErasmus MC University Medical CenterRotterdam3015GDNetherlands
| | | | - Gijsje H. Koenderink
- Department of BionanoscienceKavli Institute of Nanoscience DelftDelft University of TechnologyDelft2628CDNetherlands
| | - Lidy E. Fratila‐Apachitei
- Department of Biomechanical EngineeringFaculty of MechanicalMaritime and Materials EngineeringDelft University of TechnologyDelft2628CDNetherlands
| | - Gerjo J. V. M. van Osch
- Department of Biomechanical EngineeringFaculty of MechanicalMaritime and Materials EngineeringDelft University of TechnologyDelft2628CDNetherlands
- Department of Orthopedics and Sports MedicineErasmus MC University Medical CenterRotterdam3015GDNetherlands
- Department of OtorhinolaryngologyErasmus MC University Medical CenterRotterdam3015GDNetherlands
| | - Amir A. Zadpoor
- Department of Biomechanical EngineeringFaculty of MechanicalMaritime and Materials EngineeringDelft University of TechnologyDelft2628CDNetherlands
| |
Collapse
|
25
|
Tharakan S, Khondkar S, Lee S, Ahn S, Mathew C, Gresita A, Hadjiargyrou M, Ilyas A. 3D Printed Osteoblast-Alginate/Collagen Hydrogels Promote Survival, Proliferation and Mineralization at Low Doses of Strontium Calcium Polyphosphate. Pharmaceutics 2022; 15:pharmaceutics15010011. [PMID: 36678641 PMCID: PMC9865428 DOI: 10.3390/pharmaceutics15010011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/11/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
The generation of biomaterials via 3D printing is an emerging biotechnology with novel methods that seeks to enhance bone regeneration. Alginate and collagen are two commonly used biomaterials for bone tissue engineering and have demonstrated biocompatibility. Strontium (Sr) and Calcium phosphate (CaP) are vital elements of bone and their incorporation in composite materials has shown promising results for skeletal repair. In this study, we investigated strontium calcium polyphosphate (SCPP) doped 3D printed alginate/collagen hydrogels loaded with MC3T3-E1 osteoblasts. These cell-laden scaffolds were crosslinked with different concentrations of 1% SCPP to evaluate the effect of strontium ions on cell behavior and the biomaterial properties of the scaffolds. Through scanning electron microscopy and Raman spectroscopy, we showed that the scaffolds had a granular surface topography with the banding pattern of alginate around 1100 cm-1 and of collagen around 1430 cm-1. Our results revealed that 2 mg/mL of SCPP induced the greatest scaffold degradation after 7 days and least amount of swelling after 24 h. Exposure of osteoblasts to SCPP induced severe cytotoxic effects after 1 mg/mL. pH analysis demonstrated acidity in the presence of SCPP at a pH between 2 and 4 at 0.1, 0.3, 0.5, and 1 mg/mL, which can be buffered with cell culture medium. However, when the SCPP was added to the scaffolds, the overall pH increased indicating intrinsic activity of the scaffold to buffer the SCPP. Moreover, cell viability was observed for up to 21 days in scaffolds with early mineralization at 0.3, 0.5, and 1 mg/mL of SCPP. Overall, low doses of SCPP proved to be a potential additive in biomaterial approaches for bone tissue engineering; however, the cytotoxic effects due to its pH must be monitored closely.
Collapse
Affiliation(s)
- Shebin Tharakan
- Bio-Nanotechnology and Biomaterials (BNB) Lab, New York Institute of Technology, Old Westbury, NY 11568, USA
- College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY 11568, USA
| | - Shams Khondkar
- Bio-Nanotechnology and Biomaterials (BNB) Lab, New York Institute of Technology, Old Westbury, NY 11568, USA
- Department of Bioengineering, New York Institute of Technology, Old Westbury, NY 11568, USA
| | - Sally Lee
- Bio-Nanotechnology and Biomaterials (BNB) Lab, New York Institute of Technology, Old Westbury, NY 11568, USA
- Department of Biological and Chemical Sciences, New York Institute of Technology, Old Westbury, NY 11568, USA
| | - Serin Ahn
- Bio-Nanotechnology and Biomaterials (BNB) Lab, New York Institute of Technology, Old Westbury, NY 11568, USA
- Department of Biological and Chemical Sciences, New York Institute of Technology, Old Westbury, NY 11568, USA
| | - Chris Mathew
- Bio-Nanotechnology and Biomaterials (BNB) Lab, New York Institute of Technology, Old Westbury, NY 11568, USA
- Department of Biological and Chemical Sciences, New York Institute of Technology, Old Westbury, NY 11568, USA
| | - Andrei Gresita
- College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY 11568, USA
| | - Michael Hadjiargyrou
- Department of Biological and Chemical Sciences, New York Institute of Technology, Old Westbury, NY 11568, USA
- Correspondence: (M.H.); (A.I.)
| | - Azhar Ilyas
- Bio-Nanotechnology and Biomaterials (BNB) Lab, New York Institute of Technology, Old Westbury, NY 11568, USA
- Department Electrical and Computer Engineering, New York Institute of Technology, Old Westbury, NY 11568, USA
- Correspondence: (M.H.); (A.I.)
| |
Collapse
|
26
|
Guagliano G, Volpini C, Briatico-Vangosa F, Cornaglia AI, Visai L, Petrini P. Toward 3D-Bioprinted Models of the Liver to Boost Drug Development. Macromol Biosci 2022; 22:e2200264. [PMID: 36106413 DOI: 10.1002/mabi.202200264] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/06/2022] [Indexed: 01/15/2023]
Abstract
The main problems in drug development are connected to enormous costs related to the paltry success rate. The current situation empowered the development of high-throughput and reliable instruments, in addition to the current golden standards, able to predict the failures in the early preclinical phase. Being hepatotoxicity responsible for the failure of 30% of clinical trials, and the 21% of withdrawal of marketed drugs, the development of complex in vitro models (CIVMs) of liver is currently one of the hottest topics in the field. Among the different fabrication techniques, 3D-bioprinting is emerging as a powerful ally for their production, allowing the manufacture of three-dimensional constructs characterized by computer-controlled and customized geometry, and inter-batches reproducibility. Thanks to these, it is possible to rapidly produce tailored cell-laden constructs, to be cultured within static and dynamic systems, thus reaching a further degree of personalization when designing in vitro models. This review highlights and prioritizes the most recent advances related to the development of CIVMs of the hepatic environment to be specifically applied to pharmaceutical research, with a special focus on 3D-bioprinting, since the liver is primarily involved in the metabolism of drugs.
Collapse
Affiliation(s)
- Giuseppe Guagliano
- Department of Chemistry, Materials, and Chemical Engineering "G. Natta", Politecnico di Milano, Piazza Leonardo da Vinci 32, Milano, MI, 20133, Italy
| | - Cristina Volpini
- Molecular Medicine Department (DMM), Center for Health Technologies (CHT), UdR INSTM, University of Pavia, Via Forlanini 14, Pavia, PV, 27100, Italy.,Medicina Clinica-Specialistica, UOR5 Laboratorio Di Nanotecnologie, ICS Maugeri IRCCS, Via S. Boezio 28, Pavia, PV, 27100, Italy
| | - Francesco Briatico-Vangosa
- Department of Chemistry, Materials, and Chemical Engineering "G. Natta", Politecnico di Milano, Piazza Leonardo da Vinci 32, Milano, MI, 20133, Italy
| | - Antonia Icaro Cornaglia
- University of Pavia - Department of Public Health, Experimental and Forensic Medicine, Histology and Embryology Unit, Via Forlanini 2, Pavia, PV, 27100, Italy
| | - Livia Visai
- Molecular Medicine Department (DMM), Center for Health Technologies (CHT), UdR INSTM, University of Pavia, Via Forlanini 14, Pavia, PV, 27100, Italy.,Medicina Clinica-Specialistica, UOR5 Laboratorio Di Nanotecnologie, ICS Maugeri IRCCS, Via S. Boezio 28, Pavia, PV, 27100, Italy.,Interuniversity Center for the promotion of the 3Rs principles in teaching and research (Centro 3R), Università di Pavia Unit, Pavia, PV, 27100, Italy
| | - Paola Petrini
- Department of Chemistry, Materials, and Chemical Engineering "G. Natta", Politecnico di Milano, Piazza Leonardo da Vinci 32, Milano, MI, 20133, Italy.,Interuniversity Center for the promotion of the 3Rs principles in teaching and research (Centro 3R), Politecnico di Milano Unit, Milano, MI, 20133, Italy
| |
Collapse
|
27
|
Jin B, Liu Y, Du S, Sang X, Yang H, Mao Y. Current trends and research topics regarding liver 3D bioprinting: A bibliometric analysis research. Front Cell Dev Biol 2022; 10:1047524. [PMID: 36518541 PMCID: PMC9742412 DOI: 10.3389/fcell.2022.1047524] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/16/2022] [Indexed: 09/28/2023] Open
Abstract
Introduction: Over recent years, 3D bioprinting has changed dramatically. The articles related to liver 3D bioprinting have not been quantitatively analyzed. In this article, we screen all articles related to liver 3D bioprinting until January 2022 and analyzed them using bibliometric citation analysis to characterize the current trends in liver 3D bioprinting. Methods: The articles were identified and analyzed from the Clarivate Analytics Web of Science Core Collection database. Results: Until 1 January 2022, 71 articles focusing on liver 3D bioprinting were identified. There was an increase in the number of articles in 2015. Most articles came from the USA (n = 27), followed by South Korea (n = 22), China (n = 16), and Japan (n = 5). The printing technology of liver 3D printing was the most studied topic (n = 29). Biofabrication published the highest number of papers (n = 16) with 1,524 total citations. Conclusion: Based on bibliometric analysis of the articles until January 2022, a comprehensive analysis of the liver 3D bioprinting articles highlighted the current trends and research topics of this field. The data should provide clinicians and researchers insight into future directions relative to the liver 3D bioprinting.
Collapse
Affiliation(s)
- Bao Jin
- Department of Liver Surgery, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Yitong Liu
- Department of Liver Surgery, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
- Peking Union Medical College (PUMC), PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Shunda Du
- Department of Liver Surgery, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Xinting Sang
- Department of Liver Surgery, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Huayu Yang
- Department of Liver Surgery, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| |
Collapse
|
28
|
Lv W, Zhou H, Aazmi A, Yu M, Xu X, Yang H, Huang YYS, Ma L. Constructing biomimetic liver models through biomaterials and vasculature engineering. Regen Biomater 2022; 9:rbac079. [PMID: 36338176 PMCID: PMC9629974 DOI: 10.1093/rb/rbac079] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/19/2022] [Accepted: 10/08/2022] [Indexed: 04/04/2024] Open
Abstract
The occurrence of various liver diseases can lead to organ failure of the liver, which is one of the leading causes of mortality worldwide. Liver tissue engineering see the potential for replacing liver transplantation and drug toxicity studies facing donor shortages. The basic elements in liver tissue engineering are cells and biomaterials. Both mature hepatocytes and differentiated stem cells can be used as the main source of cells to construct spheroids and organoids, achieving improved cell function. To mimic the extracellular matrix (ECM) environment, biomaterials need to be biocompatible and bioactive, which also help support cell proliferation and differentiation and allow ECM deposition and vascularized structures formation. In addition, advanced manufacturing approaches are required to construct the extracellular microenvironment, and it has been proved that the structured three-dimensional culture system can help to improve the activity of hepatocytes and the characterization of specific proteins. In summary, we review biomaterials for liver tissue engineering, including natural hydrogels and synthetic polymers, and advanced processing techniques for building vascularized microenvironments, including bioassembly, bioprinting and microfluidic methods. We then summarize the application fields including transplant and regeneration, disease models and drug cytotoxicity analysis. In the end, we put the challenges and prospects of vascularized liver tissue engineering.
Collapse
Affiliation(s)
- Weikang Lv
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China
| | - Hongzhao Zhou
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China
| | - Abdellah Aazmi
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China
| | - Mengfei Yu
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Xiaobin Xu
- School of Materials Science and Engineering, Tongji University, Shanghai 201804, China
| | - Huayong Yang
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China
| | | | - Liang Ma
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
29
|
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] [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
- *Correspondence: Ashutosh Agarwal,
| | - 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
- Emmanuel Thomas,
| |
Collapse
|
30
|
Taymour R, Chicaiza-Cabezas NA, Gelinsky M, Lode A. Core-shell bioprinting of vascularized in vitro liver sinusoid models. Biofabrication 2022; 14. [PMID: 36070706 DOI: 10.1088/1758-5090/ac9019] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 09/07/2022] [Indexed: 11/12/2022]
Abstract
In vitro liver models allow the investigation of the cell behavior in disease conditions or in response to changes in the microenvironment. A major challenge in liver tissue engineering is to mimic the tissue-level complexity: Besides the selection of suitable biomaterial(s) replacing the extracellular matrix (ECM) and cell sources, the three-dimensional (3D) microarchitecture defined by the fabrication method is a critical factor to achieve functional constructs. In this study, coaxial extrusion-based 3D bioprinting has been applied to develop a liver sinusoid-like model that consists of a core compartment containing pre-vascular structures and a shell compartment containing hepatocytes. The shell ink was composed of alginate and methylcellulose (algMC), dissolved in human fresh frozen plasma. The algMC blend conferred high printing fidelity and stability to the core-shell constructs and the plasma as biologically active component enhanced viability and supported cluster formation and biomarker expression of HepG2 embedded in the shell. For the core, a natural ECM-like ink based on angiogenesis-supporting collagen-fibrin (CF) matrices was developed; the addition of gelatin (G) enabled 3D printing in combination with the plasma-algMC shell ink. Human endothelial cells (HUVEC), laden in the CFG core ink together with human fibroblasts as supportive cells, formed a pre-vascular network in the core in the absence and presence of HepG2 in the shell. The cellular interactions occurring in the triple culture model enhanced the albumin secretion. In conclusion, core-shell bioprinting was shown to be a valuable tool to study cell-cell-interactions and to develop complex tissue-like models.
Collapse
Affiliation(s)
- Rania Taymour
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine, Dresden University of Technology, Fetscherstrasse 74, Dresden, Sachsen, 01307, GERMANY
| | - Nathaly Alejandra Chicaiza-Cabezas
- Centre for Translational Bone, Joint and Soft Tissue Research, Technische Universitaet Dresden, Fetscherstrasse 74, Dresden, Sachsen, 01307, GERMANY
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, Technische Universitat Dresden, Fetscherstr. 74, Dresden, 01062, GERMANY
| | - Anja Lode
- Centre for Translational Bone, Joint and Soft Tissue Research, Technische Universitaet Dresden, Fetscherstrasse 74, Dresden, 01307, GERMANY
| |
Collapse
|
31
|
Zhang P, Li X, Chen JY, Abate A. Controlled fabrication of functional liver spheroids with microfluidic flow cytometric printing. Biofabrication 2022; 14. [PMID: 35917810 DOI: 10.1088/1758-5090/ac8622] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/02/2022] [Indexed: 11/11/2022]
Abstract
Multicellular liver spheroids are 3D culture models useful in the development of therapies for liver fibrosis. While these models can recapitulate fibrotic disease, current methods for generating them via random aggregation are uncontrolled, yielding spheroids of variable size, function, and utility. Here, we report fabrication of precision liver spheroids with microfluidic flow cytometric printing. Our approach fabricates spheroids cell-by-cell, yielding structures with exact numbers of different cell types. Because spheroid function depends on composition, our precision spheroids have superior functional uniformity, allowing more accurate and statistically significant screens compared to randomly generated spheroids. The approach produces thousands of spheroids per hour, and thus affords a scalable platform by which to manufacture single-cell precision spheroids for disease modeling and high throughput drug testing.
Collapse
Affiliation(s)
- Pengfei Zhang
- University of California San Francisco, 1700 4th St, San Francisco, California, 94143, UNITED STATES
| | - Xiangpeng Li
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, 1700 4th St, San Francisco, 94143, UNITED STATES
| | - Jennifer Y Chen
- Department of Medicine, University of California San Francisco, 555 Mission Bay Blvd South, San Francisco, 94143, UNITED STATES
| | - Adam Abate
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, 1700 4th St, San Francisco, San Francisco, California, 94158, UNITED STATES
| |
Collapse
|
32
|
Panda S, Hajra S, Mistewicz K, Nowacki B, In-Na P, Krushynska A, Mishra YK, Kim HJ. A focused review on three-dimensional bioprinting technology for artificial organ fabrication. Biomater Sci 2022; 10:5054-5080. [PMID: 35876134 DOI: 10.1039/d2bm00797e] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Three-dimensional (3D) bioprinting technology has attracted a great deal of interest because it can be easily adapted to many industries and research sectors, such as biomedical, manufacturing, education, and engineering. Specifically, 3D bioprinting has provided significant advances in the medical industry, since such technology has led to significant breakthroughs in the synthesis of biomaterials, cells, and accompanying elements to produce composite living tissues. 3D bioprinting technology could lead to the immense capability of replacing damaged or injured tissues or organs with newly dispensed cell biomaterials and functional tissues. Several types of bioprinting technology and different bio-inks can be used to replicate cells and generate supporting units as complex 3D living tissues. Bioprinting techniques have undergone great advancements in the field of regenerative medicine to provide 3D printed models for numerous artificial organs and transplantable tissues. This review paper aims to provide an overview of 3D-bioprinting technologies by elucidating the current advancements, recent progress, opportunities, and applications in this field. It highlights the most recent advancements in 3D-bioprinting technology, particularly in the area of artificial organ development and cancer research. Additionally, the paper speculates on the future progress in 3D-bioprinting as a versatile foundation for several biomedical applications.
Collapse
Affiliation(s)
- Swati Panda
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology, Daegu-42988, South Korea.
| | - Sugato Hajra
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology, Daegu-42988, South Korea.
| | - Krystian Mistewicz
- Institute of Physics - Center for Science and Education, Silesian University of Technology, Krasińskiego 8, Katowice, Poland
| | - Bartłomiej Nowacki
- Faculty of Materials Engineering, Silesian University of Technology, Krasińskiego 8, Katowice, Poland
| | - Pichaya In-Na
- Department of Chemical Technology, Faculty of Science, Chulalongkorn University, 254 Phyathai Road, Wangmai, Pathumwan, Bangkok-10330, Thailand
| | - Anastasiia Krushynska
- Engineering and Technology Institute Groningen (ENTEG), Faculty of Science and Engineering, University of Groningen, Nijenborgh 4, Groningen, 9747 AG, Netherlands
| | - Yogendra Kumar Mishra
- Mads Clausen Institute, NanoSYD, University of Southern Denmark, Alsion 2, 6400 Sønderborg, Denmark
| | - Hoe Joon Kim
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology, Daegu-42988, South Korea. .,Robotics and Mechatronics Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu-42988, South Korea
| |
Collapse
|
33
|
Tissue engineering approaches for the in vitro production of spermatids to treat male infertility: A review. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
34
|
Viability and Functionality of Neonatal Porcine Islet-like Cell Clusters Bioprinted in Alginate-Based Bioinks. Biomedicines 2022; 10:biomedicines10061420. [PMID: 35740440 PMCID: PMC9220255 DOI: 10.3390/biomedicines10061420] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 11/17/2022] Open
Abstract
The transplantation of pancreatic islets can prevent severe long-term complications in diabetes mellitus type 1 patients. With respect to a shortage of donor organs, the transplantation of xenogeneic islets is highly attractive. To avoid rejection, islets can be encapsulated in immuno-protective hydrogel-macrocapsules, whereby 3D bioprinted structures with macropores allow for a high surface-to-volume ratio and reduced diffusion distances. In the present study, we applied 3D bioprinting to encapsulate the potentially clinically applicable neonatal porcine islet-like cell clusters (NICC) in alginate-methylcellulose. The material was additionally supplemented with bovine serum albumin or the human blood plasma derivatives platelet lysate and fresh frozen plasma. NICC were analysed for viability, proliferation, the presence of hormones, and the release of insulin in reaction to glucose stimulation. Bioprinted NICC are homogeneously distributed, remain morphologically intact, and show a comparable viability and proliferation to control NICC. The number of insulin-positive cells is comparable between the groups and over time. The amount of insulin release increases over time and is released in response to glucose stimulation over 4 weeks. In summary, we show the successful bioprinting of NICC and could demonstrate functionality over the long-term in vitro. Supplementation resulted in a trend for higher viability, but no additional benefit on functionality was observed.
Collapse
|
35
|
Turner PR, McConnell M, Young SL, Cabral JD. 3D living dressing improves healing and modulates immune response in a thermal injury model. Tissue Eng Part C Methods 2022; 28:431-439. [PMID: 35658609 DOI: 10.1089/ten.tec.2022.0088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Thermal injury trauma can induce a state of immunosuppression causing wounds to become chronic in nature. Stem-cell based therapies represent a promising new approach to treat such wounds due to their capacity to self-renew and their multi-lineage potential. Mesenchymal stem cells (MSCs) are known to secrete endogenous factors that stimulate wound healing by promoting angiogenesis, extracellular matrix (ECM) remodeling, skin re-generation, and by dampening down inflammation. MSC delivery in a biomaterial construct can augment their wound healing capacity by concentrating cells at the burn site and upregulating trophic factor secretion. The work presented is the first to evaluate repair in an in vitro raft thermal injury model using a regenerative, dual cell delivery 3D core/shell (c/s) "living dressing" construct. This previously characterized 3D c/s bioprinted construct, that delivers both MSCs and endothelial cells, was used to treat an in vitro 3D raft skin thermal injury wound model. The mesenchymal stromal cell line (T0523) was encapsulated within a gelatin-based shell bioink, and human um-bilical vein endothelial cells (HUVECs) within a chitosan-based core bioink to biofabricate a living dressing for enhanced thermal injury repair and regeneration. We hypothesized that the cell-laden c/s tissue engineered con-struct (TEC) would strengthen the wound's proangiogenic, anti-inflammatory, and skin regeneration potential. An in vitro thermal injury in a 3D raft skin model showed a slight delay in wound closure in the presence of the c/s TEC but was augmented by corresponding increases in the release of wound healing factors, EGF, MMP-9, TGF-α, PDGF; a decrease in pro-inflammatory factor IL-6, and evidence of neovascularization. Impact statement C/s 3D bioprinted living dressings were used to heal an in vitro thermal injury wound in a 3D raft skin model. The MSC/HUVEC-laden 3D TEC when compared to the untreated control resulted in a slight de-lay in wound closure along with corresponding increases in the secretion of wound healing factors EGF, MMP-9, TGF-α, PDGF; a decrease in pro-inflammatory factor IL-6, and promotion of neovascularization. The present work is the first to evaluate repair and corresponding cytokine response in an in vitro 3D raft thermal injury model using a "living dressing" construct.
Collapse
Affiliation(s)
- Paul R Turner
- University of Otago, 2495, Surgical Sciences, Dunedin, New Zealand;
| | - Michelle McConnell
- University of Otago, 2495, Microbiology & Immunology, Dunedin, New Zealand;
| | - Sarah L Young
- The University of Sydney, 4334, School of Medical Sciences, Sydney, New South Wales, Australia;
| | - Jaydee Dones Cabral
- University of Otago, 2495, Microbiology & Immunology, Dunedin, Otago, New Zealand;
| |
Collapse
|
36
|
Böttcher B, Pflieger A, Schumacher J, Jungnickel B, Feller KH. 3D Bioprinting of Prevascularized Full-Thickness Gelatin-Alginate Structures with Embedded Co-Cultures. Bioengineering (Basel) 2022; 9:bioengineering9060242. [PMID: 35735485 PMCID: PMC9219913 DOI: 10.3390/bioengineering9060242] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 12/24/2022] Open
Abstract
The use of bioprinting allows the creation of complex three-dimensional cell laden grafts with spatial placements of different cell lines. However, a major challenge is insufficient nutrient transfer, especially with the increased size of the graft causing necrosis and reduced proliferation. A possibility to improve nutrient support is the integration of tubular structures for reducing diffusion paths. In this study the influence of prevascularization in full-thickness grafts on cell growth with a variation of cultivation style and cellular composition was investigated. To perform this, the rheological properties of the used gelatin-alginate hydrogel as well as possibilities to improve growth conditions in the hydrogel were assessed. Prevascularized grafts were manufactured using a pneumatic extrusion-based bioprinter with a coaxial extrusion tool. The prevascularized grafts were statically and dynamically cultured with a monoculture of HepG2 cells. Additionally, a co-culture of HepG2 cells, fibroblasts and HUVEC-TERT2 was created while HUVEC-TERT2s were concentrically placed around the hollow channels. A static culture of prevascularized grafts showed short-term improvements in cell proliferation compared to avascular grafts, while a perfusion-based culture showed improvements in mid-term cultivation times. The cultivation of the co-culture indicated the formation of vascular structures from the hollow channels toward avascular areas. According to these results, the integration of prevascular structures show beneficial effects for the in vitro cultivation of bioprinted grafts for which its impact can be increased in larger grafts.
Collapse
Affiliation(s)
- Bastian Böttcher
- Institute for Microsystem and Precision Engineering, Ernst-Abbe University of Applied Science Jena, 07745 Jena, Germany; (B.B.); (A.P.); (J.S.)
| | - Astrid Pflieger
- Institute for Microsystem and Precision Engineering, Ernst-Abbe University of Applied Science Jena, 07745 Jena, Germany; (B.B.); (A.P.); (J.S.)
| | - Jan Schumacher
- Institute for Microsystem and Precision Engineering, Ernst-Abbe University of Applied Science Jena, 07745 Jena, Germany; (B.B.); (A.P.); (J.S.)
| | - Berit Jungnickel
- Department of Cell Biology, Institute of Biochemistry and Biophysics, Faculty of Biological Sciences, Friedrich Schiller University Jena, 07745 Jena, Germany;
| | - Karl-Heinz Feller
- Institute for Microsystem and Precision Engineering, Ernst-Abbe University of Applied Science Jena, 07745 Jena, Germany; (B.B.); (A.P.); (J.S.)
- Correspondence: ; Tel.: +49-3641-205-621
| |
Collapse
|
37
|
Mai P, Hampl J, Baca M, Brauer D, Singh S, Weise F, Borowiec J, Schmidt A, Küstner JM, Klett M, Gebinoga M, Schroeder IS, Markert UR, Glahn F, Schumann B, Eckstein D, Schober A. MatriGrid® Based Biological Morphologies: Tools for 3D Cell Culturing. Bioengineering (Basel) 2022; 9:bioengineering9050220. [PMID: 35621498 PMCID: PMC9138054 DOI: 10.3390/bioengineering9050220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 02/06/2023] Open
Abstract
Recent trends in 3D cell culturing has placed organotypic tissue models at another level. Now, not only is the microenvironment at the cynosure of this research, but rather, microscopic geometrical parameters are also decisive for mimicking a tissue model. Over the years, technologies such as micromachining, 3D printing, and hydrogels are making the foundation of this field. However, mimicking the topography of a particular tissue-relevant substrate can be achieved relatively simply with so-called template or morphology transfer techniques. Over the last 15 years, in one such research venture, we have been investigating a micro thermoforming technique as a facile tool for generating bioinspired topographies. We call them MatriGrid®s. In this research account, we summarize our learning outcome from this technique in terms of the influence of 3D micro morphologies on different cell cultures that we have tested in our laboratory. An integral part of this research is the evolution of unavoidable aspects such as possible label-free sensing and fluidic automatization. The development in the research field is also documented in this account.
Collapse
Affiliation(s)
- Patrick Mai
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Jörg Hampl
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
- Correspondence: (J.H.); (A.S.); Tel.: +49-3677-6933387 (A.S.)
| | - Martin Baca
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Dana Brauer
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Sukhdeep Singh
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Frank Weise
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Justyna Borowiec
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - André Schmidt
- Placenta Lab, Department of Obstetrics, Jena University Hospital, 07747 Jena, Germany; (A.S.); (U.R.M.)
| | - Johanna Merle Küstner
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Maren Klett
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Michael Gebinoga
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Insa S. Schroeder
- Biophysics Division, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany;
| | - Udo R. Markert
- Placenta Lab, Department of Obstetrics, Jena University Hospital, 07747 Jena, Germany; (A.S.); (U.R.M.)
| | - Felix Glahn
- Institute of Environmental Toxicology, Martin-Luther-University Halle-Wittenberg, 06097 Halle, Germany; (F.G.); (B.S.); (D.E.)
| | - Berit Schumann
- Institute of Environmental Toxicology, Martin-Luther-University Halle-Wittenberg, 06097 Halle, Germany; (F.G.); (B.S.); (D.E.)
| | - Diana Eckstein
- Institute of Environmental Toxicology, Martin-Luther-University Halle-Wittenberg, 06097 Halle, Germany; (F.G.); (B.S.); (D.E.)
| | - Andreas Schober
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
- Correspondence: (J.H.); (A.S.); Tel.: +49-3677-6933387 (A.S.)
| |
Collapse
|
38
|
Bernal PN, Bouwmeester M, Madrid-Wolff J, Falandt M, Florczak S, Rodriguez NG, Li Y, Größbacher G, Samsom RA, van Wolferen M, van der Laan LJW, Delrot P, Loterie D, Malda J, Moser C, Spee B, Levato R. Volumetric Bioprinting of Organoids and Optically Tuned Hydrogels to Build Liver-Like Metabolic Biofactories. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2110054. [PMID: 35166410 DOI: 10.1002/adma.202110054] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/28/2022] [Indexed: 06/14/2023]
Abstract
Organ- and tissue-level biological functions are intimately linked to microscale cell-cell interactions and to the overarching tissue architecture. Together, biofabrication and organoid technologies offer the unique potential to engineer multi-scale living constructs, with cellular microenvironments formed by stem cell self-assembled structures embedded in customizable bioprinted geometries. This study introduces the volumetric bioprinting of complex organoid-laden constructs, which capture key functions of the human liver. Volumetric bioprinting via optical tomography shapes organoid-laden gelatin hydrogels into complex centimeter-scale 3D structures in under 20 s. Optically tuned bioresins enable refractive index matching of specific intracellular structures, countering the disruptive impact of cell-mediated light scattering on printing resolution. This layerless, nozzle-free technique poses no harmful mechanical stresses on organoids, resulting in superior viability and morphology preservation post-printing. Bioprinted organoids undergo hepatocytic differentiation showing albumin synthesis, liver-specific enzyme activity, and remarkably acquired native-like polarization. Organoids embedded within low stiffness gelatins (<2 kPa) are bioprinted into mathematically defined lattices with varying degrees of pore network tortuosity, and cultured under perfusion. These structures act as metabolic biofactories in which liver-specific ammonia detoxification can be enhanced by the architectural profile of the constructs. This technology opens up new possibilities for regenerative medicine and personalized drug testing.
Collapse
Affiliation(s)
- Paulina Nuñez Bernal
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
| | - Manon Bouwmeester
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CT, The Netherlands
| | - Jorge Madrid-Wolff
- Laboratory of Applied Photonics Devices, École Polytechnique Fédéral Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Marc Falandt
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CT, The Netherlands
| | - Sammy Florczak
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
| | - Nuria Ginés Rodriguez
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
| | - Yang Li
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
| | - Gabriel Größbacher
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
| | - Roos-Anne Samsom
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CT, The Netherlands
| | - Monique van Wolferen
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CT, The Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, 3015GD, The Netherlands
| | - Paul Delrot
- Readily3D SA, EPFL Innovation Park, Building A, Lausanne, CH-1015, Switzerland
| | - Damien Loterie
- Readily3D SA, EPFL Innovation Park, Building A, Lausanne, CH-1015, Switzerland
| | - Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CT, The Netherlands
| | - Christophe Moser
- Laboratory of Applied Photonics Devices, École Polytechnique Fédéral Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Bart Spee
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CT, The Netherlands
| | - Riccardo Levato
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CT, The Netherlands
| |
Collapse
|
39
|
Mohan TS, Datta P, Nesaei S, Ozbolat V, Ozbolat IT. 3D Coaxial Bioprinting: Process Mechanisms, Bioinks and Applications. PROGRESS IN BIOMEDICAL ENGINEERING (BRISTOL, ENGLAND) 2022; 4:022003. [PMID: 35573639 PMCID: PMC9103990 DOI: 10.1088/2516-1091/ac631c] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In the last decade, bioprinting has emerged as a facile technique for fabricating tissues constructs mimicking the architectural complexity and compositional heterogeneity of native tissues. Amongst different bioprinting modalities, extrusion-based bioprinting (EBB) is the most widely used technique. Coaxial bioprinting, a type of EBB, enables fabrication of concentric cell-material layers and enlarges the scope of EBB to mimic several key aspects of native tissues. Over the period of development of bioprinting, tissue constructs integrated with vascular networks, have been one of the major achievements made possible largely by coaxial bioprinting. In this review, current advancements in biofabrication of constructs with coaxial bioprinting are discussed with a focus on different bioinks that are particularly suitable for this modality. This review also expounds the properties of different bioinks suitable for coaxial bioprinting and then analyses the key achievements made by the application of coaxial bioprinting in tissue engineering, drug delivery and in-vitro disease modelling. The major limitations and future perspectives on the critical factors that will determine the ultimate clinical translation of the versatile technique are also presented to the reader.
Collapse
Affiliation(s)
- Tarun Shyam Mohan
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, West Bengal, India
| | - Pallab Datta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, West Bengal, India
| | - Sepehr Nesaei
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, United States of America
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, United States of America
| | - Veli Ozbolat
- Department of Mechanical Engineering, Ceyhan Engineering Faculty, Cukurova University, Adana, Turkey
| | - Ibrahim T Ozbolat
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, United States of America
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, United States of America
- Biomedical Engineering Department, Penn State University, University Park, PA, United States of America
- Materials Research Institute, Penn State University, University Park, PA, United States of America
- Department of Neurosurgery, Penn State University, Hershey, PA, United States of America
| |
Collapse
|
40
|
Kim D, Kim M, Lee J, Jang J. Review on Multicomponent Hydrogel Bioinks Based on Natural Biomaterials for Bioprinting 3D Liver Tissues. Front Bioeng Biotechnol 2022; 10:764682. [PMID: 35237569 PMCID: PMC8884173 DOI: 10.3389/fbioe.2022.764682] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/11/2022] [Indexed: 12/12/2022] Open
Abstract
Three-dimensional (3D)-printed in vitro tissue models have been used in various biomedical fields owing to numerous advantages such as enhancements in cell response and functionality. In liver tissue engineering, several studies have been reported using 3D-printed liver tissue models with improved cellular responses and functions in drug screening, liver disease, and liver regenerative medicine. However, the application of conventional single-component bioinks for the printing of 3D in vitro liver constructs remains problematic because of the complex structural and physiological characteristics of the liver. The use of multicomponent bioinks has become an attractive strategy for bioprinting 3D functional in vitro liver tissue models because of the various advantages of multicomponent bioinks, such as improved mechanical properties of the printed tissue construct and cell functionality. Therefore, it is essential to review various 3D bioprinting techniques and multicomponent hydrogel bioinks proposed for liver tissue engineering to suggest future directions for liver tissue engineering. Accordingly, we herein review multicomponent bioinks for 3D-bioprinted liver tissues. We first describe the fabrication methods capable of printing multicomponent bioinks and introduce considerations for bioprinting. We subsequently categorize and evaluate the materials typically utilized for multicomponent bioinks based on their characteristics. In addition, we also review recent studies for the application of multicomponent bioinks to fabricate in vitro liver tissue models. Finally, we discuss the limitations of current studies and emphasize aspects that must be resolved to enhance the future applicability of such bioinks.
Collapse
Affiliation(s)
- Daekeun Kim
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Minseok Kim
- Department of Mechanical System Engineering, Kumoh National Institute of Technology, Gumi, South Korea
- Department of Aeronautics, Mechanical and Electronic Convergence Engineering, Kumoh National Institute of Technology, Gumi, South Korea
| | - Jongwan Lee
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Jinah Jang
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
- Institute of Convergence Science, Yonsei University, Seoul, South Korea
| |
Collapse
|
41
|
Shahabipour F, Tavafoghi M, Aninwene GE, Bonakdar S, Oskuee RK, Shokrgozar MA, Potyondy T, Alambeigi F, Ahadian S. Coaxial 3D bioprinting of tri-polymer scaffolds to improve the osteogenic and vasculogenic potential of cells in co-culture models. J Biomed Mater Res A 2022; 110:1077-1089. [PMID: 35025130 DOI: 10.1002/jbm.a.37354] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 12/20/2021] [Accepted: 12/27/2021] [Indexed: 12/20/2022]
Abstract
The crosstalk between osteoblasts and endothelial cells is critical for bone vascularization and regeneration. Here, we used a coaxial 3D bioprinting method to directly print an osteon-like structure by depositing angiogenic and osteogenic bioinks from the core and shell regions of the coaxial nozzle, respectively. The bioinks were made up of gelatin, gelatin methacryloyl (GelMA), alginate, and hydroxyapatite (HAp) nanoparticles and were loaded with human umbilical vascular endothelial cells (HUVECs) and osteoblasts (MC3T3) in the core and shell regions, respectively. Conventional monoaxial 3D bioprinting was used as a control method, where the hydrogels, HAp nanoparticles, MC3T3 cells, and HUVECs were all mixed in one bioink and printed from the core nozzle. As a result, the bioprinted scaffolds were composed of cell-laden fibers with either a core-shell or homogenous structure, providing a non-contact (indirect) or contact (direct) co-culture of MC3T3 cells and HUVECs, respectively. Both structures supported the 3D culture of HUVECs and osteoblasts over a long period. The scaffolds also supported the expression of osteogenic and angiogenic factors. However, the gene expression was significantly higher for the core-shell structure than the homogeneous structure due to the well-defined distribution of osteoblasts and endothelial cells and the formation of vessel-like structures in the co-culture system. Our results indicated that the coaxial bioprinting technique, with the ability to create a non-contact co-culture of cells, can provide a more efficient bioprinting strategy for printing highly vascularized and bioactive bone structures.
Collapse
Affiliation(s)
- Fahimeh Shahabipour
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran.,Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Maryam Tavafoghi
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, California, USA.,Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, California, USA
| | - George E Aninwene
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, California, USA.,Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, California, USA.,California NanoSystems Institute (CNSI), University of California-Los Angeles, Los Angeles, California, USA
| | - Shahin Bonakdar
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Kazemi Oskuee
- Biomedical Applied Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Tyler Potyondy
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, California, USA.,Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, California, USA
| | - Farshid Alambeigi
- Walker Department of Mechanical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Samad Ahadian
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, USA
| |
Collapse
|
42
|
Kilian D, Cometta S, Bernhardt A, Taymour R, Golde J, Ahlfeld T, Emmermacher J, Gelinsky M, Lode A. Core-shell bioprinting as a strategy to apply differentiation factors in a spatially defined manner inside osteochondral tissue substitutes. Biofabrication 2021; 14. [PMID: 34933296 DOI: 10.1088/1758-5090/ac457b] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/21/2021] [Indexed: 11/12/2022]
Abstract
One of the key challenges in osteochondral tissue engineering is to define specified zones with varying material properties, cell types and biochemical factors supporting locally adjusted differentiation into the osteogenic and chondrogenic lineage, respectively. Herein, extrusion-based core-shell bioprinting is introduced as a potent tool allowing a spatially defined delivery of cell types and differentiation factors TGF-β3 and BMP-2 in separated compartments of hydrogel strands, and, therefore, a local supply of matching factors for chondrocytes and osteoblasts. Ink development was based on blends of alginate and methylcellulose, in combination with varying concentrations of the nanoclay Laponite whose high affinity binding capacity for various molecules was exploited. Release kinetics of model molecules was successfully tuned by Laponite addition. Core-shell bioprinting was proven to generate well-oriented compartments within one strand as monitored by optical coherence tomography in a non-invasive manner. Chondrocytes and osteoblasts were applied each in the shell while the respective differentiation factors (TGF-β3, BMP-2) were provided by a Laponite-supported core serving as central factor depot within the strand, allowing directed differentiation of cells in close contact to the core. Experiments with bi-zonal constructs, comprising an osteogenic and a chondrogenic zone, revealed that the local delivery of the factors from the core reduces effects of these factors on the cells in the other scaffold zone. These observations prove the general suitability of the suggested system for co-differentiation of different cell types within a zonal construct.
Collapse
Affiliation(s)
- David Kilian
- Centre for Translational Bone, Joint and Soft Tissue Research, Technische Universitaet Dresden, Fetscherstrasse 74, Dresden, 01307, GERMANY
| | - Silvia Cometta
- Dresden University of Technology, Fetscherstrasse 74, Dresden, Sachsen, 01307, GERMANY
| | - Anne Bernhardt
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine, Dresden University of Technology, Fetscherstrasse 74, Dresden, Sachsen, 01307, GERMANY
| | - Rania Taymour
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine, Dresden University of Technology, Fetscherstrasse 74, Dresden, Sachsen, 01307, GERMANY
| | - Jonas Golde
- Clinical Sensoring and Monitoring, Department of Anesthesiology and Intensive Care Medicine, Dresden University of Technology, Fetscherstrasse 74, Dresden, Sachsen, 01307, GERMANY
| | - Tilman Ahlfeld
- Centre for Translational Bone, Joint and Soft Tissue Research, Technische Universitaet Dresden, Fetscherstrasse 74, Dresden, 01307, GERMANY
| | - Julia Emmermacher
- Centre for translational bone, joint and soft tissue research, Technische Universitat Dresden, Fetscherstrasse 74, Dresden, 01307, GERMANY
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, Technische Universitaet Dresden, Fetscherstr. 74, Dresden, 01307, GERMANY
| | - Anja Lode
- Centre for Translational Bone, Joint and Soft Tissue Research, Technische Universitaet Dresden, Fetscherstrasse 74, Dresden, 01307, GERMANY
| |
Collapse
|
43
|
de Hoyos-Vega JM, Hong HJ, Stybayeva G, Revzin A. Hepatocyte cultures: From collagen gel sandwiches to microfluidic devices with integrated biosensors. APL Bioeng 2021; 5:041504. [PMID: 34703968 PMCID: PMC8519630 DOI: 10.1063/5.0058798] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocytes are parenchymal cells of the liver responsible for drug detoxification, urea and bile production, serum protein synthesis, and glucose homeostasis. Hepatocytes are widely used for drug toxicity studies in bioartificial liver devices and for cell-based liver therapies. Because hepatocytes are highly differentiated cells residing in a complex microenvironment in vivo, they tend to lose hepatic phenotype and function in vitro. This paper first reviews traditional culture approaches used to rescue hepatic function in vitro and then discusses the benefits of emerging microfluidic-based culture approaches. We conclude by reviewing integration of hepatocyte cultures with bioanalytical or sensing approaches.
Collapse
Affiliation(s)
- Jose M. de Hoyos-Vega
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Hye Jin Hong
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| |
Collapse
|
44
|
Lin C, Wang Y, Huang Z, Wu T, Xu W, Wu W, Xu Z. Advances in Filament Structure of 3D Bioprinted Biodegradable Bone Repair Scaffolds. Int J Bioprint 2021; 7:426. [PMID: 34805599 PMCID: PMC8600304 DOI: 10.18063/ijb.v7i4.426] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/03/2021] [Indexed: 12/18/2022] Open
Abstract
Conventional bone repair scaffolds can no longer meet the high standards and requirements of clinical applications in terms of preparation process and service performance. Studies have shown that the diversity of filament structures of implantable scaffolds is closely related to their overall properties (mechanical properties, degradation properties, and biological properties). To better elucidate the characteristics and advantages of different filament structures, this paper retrieves and summarizes the state of the art in the filament structure of the three-dimensional (3D) bioprinted biodegradable bone repair scaffolds, mainly including single-layer structure, double-layer structure, hollow structure, core-shell structure and bionic structures. The eximious performance of the novel scaffolds was discussed from different aspects (material composition, ink configuration, printing parameters, etc.). Besides, the additional functions of the current bone repair scaffold, such as chondrogenesis, angiogenesis, anti-bacteria, and anti-tumor, were also concluded. Finally, the paper prospects the future material selection, structural design, functional development, and performance optimization of bone repair scaffolds.
Collapse
Affiliation(s)
- Chengxiong Lin
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Polymer Products, Guangdong Medical Device Research Institute, Guangzhou 510500, China
| | - Yaocheng Wang
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Polymer Products, Guangdong Medical Device Research Institute, Guangzhou 510500, China.,School of Railway Tracks and Transportation, Wuyi University, Jiangmen 529020, China
| | - Zhengyu Huang
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Polymer Products, Guangdong Medical Device Research Institute, Guangzhou 510500, China.,School of Railway Tracks and Transportation, Wuyi University, Jiangmen 529020, China
| | - Tingting Wu
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Polymer Products, Guangdong Medical Device Research Institute, Guangzhou 510500, China
| | - Weikang Xu
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Polymer Products, Guangdong Medical Device Research Institute, Guangzhou 510500, China
| | - Wenming Wu
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Polymer Products, Guangdong Medical Device Research Institute, Guangzhou 510500, China
| | - Zhibiao Xu
- School of Railway Tracks and Transportation, Wuyi University, Jiangmen 529020, China
| |
Collapse
|
45
|
Homogeneous and Reproducible Mixing of Highly Viscous Biomaterial Inks and Cell Suspensions to Create Bioinks. Gels 2021; 7:gels7040227. [PMID: 34842704 PMCID: PMC8628813 DOI: 10.3390/gels7040227] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/28/2022] Open
Abstract
Highly viscous bioinks offer great advantages for the three-dimensional fabrication of cell-laden constructs by microextrusion printing. However, no standardised method of mixing a high viscosity biomaterial ink and a cell suspension has been established so far, leading to non-reproducible printing results. A novel method for the homogeneous and reproducible mixing of the two components using a mixing unit connecting two syringes is developed and investigated. Several static mixing units, based on established mixing designs, were adapted and their functionality was determined by analysing specific features of the resulting bioink. As a model system, we selected a highly viscous ink consisting of fresh frozen human blood plasma, alginate, and methylcellulose, and a cell suspension containing immortalized human mesenchymal stem cells. This bioink is crosslinked after fabrication. A pre-crosslinked gellan gum-based bioink providing a different extrusion behaviour was introduced to validate the conclusions drawn from the model system. For characterisation, bioink from different zones within the mixing device was analysed by measurement of its viscosity, shape fidelity after printing and visual homogeneity. When taking all three parameters into account, a comprehensive and reliable comparison of the mixing quality was possible. In comparison to the established method of manual mixing inside a beaker using a spatula, a significantly higher proportion of viable cells was detected directly after mixing and plotting for both bioinks when the mixing unit was used. A screw-like mixing unit, termed “HighVisc”, was found to result in a homogenous bioink after a low number of mixing cycles while achieving high cell viability rates.
Collapse
|
46
|
Three-Dimensional Liver Culture Systems to Maintain Primary Hepatic Properties for Toxicological Analysis In Vitro. Int J Mol Sci 2021; 22:ijms221910214. [PMID: 34638555 PMCID: PMC8508724 DOI: 10.3390/ijms221910214] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/15/2021] [Accepted: 09/19/2021] [Indexed: 12/13/2022] Open
Abstract
Drug-induced liver injury (DILI) is the major reason for failures in drug development and withdrawal of approved drugs from the market. Two-dimensional cultures of hepatocytes often fail to reliably predict DILI: hepatoma cell lines such as HepG2 do not reflect important primary-like hepatic properties and primary human hepatocytes (pHHs) dedifferentiate quickly in vitro and are, therefore, not suitable for long-term toxicity studies. More predictive liver in vitro models are urgently required in drug development and compound safety evaluation. This review discusses available human hepatic cell types for in vitro toxicology analysis and their usage in established and emerging three-dimensional (3D) culture systems. Generally, 3D cultures maintain or improve primary hepatic functions (including expression of drug-metabolizing enzymes) of different liver cells for several weeks of culture, thus allowing long-term and repeated-dose toxicity studies. Spheroid cultures of pHHs have been comprehensively tested, but also other cell types such as HepaRG benefit from 3D culture systems. Emerging 3D culture techniques include usage of induced pluripotent stem-cell-derived hepatocytes and primary-like upcyte cells, as well as advanced culture techniques such as microfluidic liver-on-a-chip models. In-depth characterization of existing and emerging 3D hepatocyte technologies is indispensable for successful implementation of such systems in toxicological analysis.
Collapse
|
47
|
Del Amo C, Perez-Valle A, Perez-Garrastachu M, Jauregui I, Andollo N, Arluzea J, Guerrero P, de la Caba K, Andia I. Plasma-Based Bioinks for Extrusion Bioprinting of Advanced Dressings. Biomedicines 2021; 9:1023. [PMID: 34440227 PMCID: PMC8392180 DOI: 10.3390/biomedicines9081023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 12/04/2022] Open
Abstract
Extrusion bioprinting based on the development of novel bioinks offers the possibility of manufacturing clinically useful tools for wound management. In this study, we show the rheological properties and printability outcomes of two advanced dressings based on platelet-rich plasma (PRP) and platelet-poor plasma (PPP) blended with alginate and loaded with dermal fibroblasts. Measurements taken at 1 h, 4 days, and 18 days showed that both the PRP- and PPP-based dressings retain plasma and platelet proteins, which led to the upregulation of angiogenic and immunomodulatory proteins by embedded fibroblasts (e.g., an up to 69-fold increase in vascular endothelial growth factor (VEGF), an up to 188-fold increase in monocyte chemotactic protein 1 (MCP-1), and an up to 456-fold increase in hepatocyte growth factor (HGF) 18 days after printing). Conditioned media harvested from both PRP and PPP constructs stimulated the proliferation of human umbilical vein endothelial cells (HUVECs), whereas only those from PRP dressings stimulated HUVEC migration, which correlated with the VEGF/MCP-1 and VEGF/HGF ratios. Similarly, the advanced dressings increased the level of interleukin-8 and led to a four-fold change in the level of extracellular matrix protein 1. These findings suggest that careful selection of plasma formulations to fabricate wound dressings can enable regulation of the molecular composition of the microenvironment, as well as paracrine interactions, thereby improving the clinical potential of dressings and providing the possibility to tailor each composition to specific wound types and healing stages.
Collapse
Affiliation(s)
- Cristina Del Amo
- Regenerative Therapies, Bioprinting Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, 48903 Barakaldo, Spain; (C.D.A.); (A.P.-V.); (I.J.)
| | - Arantza Perez-Valle
- Regenerative Therapies, Bioprinting Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, 48903 Barakaldo, Spain; (C.D.A.); (A.P.-V.); (I.J.)
| | - Miguel Perez-Garrastachu
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain; (M.P.-G.); (N.A.); (J.A.)
| | - Ines Jauregui
- Regenerative Therapies, Bioprinting Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, 48903 Barakaldo, Spain; (C.D.A.); (A.P.-V.); (I.J.)
| | - Noelia Andollo
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain; (M.P.-G.); (N.A.); (J.A.)
- BEGIKER, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Jon Arluzea
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain; (M.P.-G.); (N.A.); (J.A.)
| | - Pedro Guerrero
- BIOMAT Research Group, Escuela de Ingeniería de Gipuzkoa Donostia-San Sebastián, University of the Basque Country (UPV/EHU), 20018 Donostia-San Sebastian, Spain; (P.G.); (K.d.l.C.)
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain
| | - Koro de la Caba
- BIOMAT Research Group, Escuela de Ingeniería de Gipuzkoa Donostia-San Sebastián, University of the Basque Country (UPV/EHU), 20018 Donostia-San Sebastian, Spain; (P.G.); (K.d.l.C.)
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain
| | - Isabel Andia
- Regenerative Therapies, Bioprinting Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, 48903 Barakaldo, Spain; (C.D.A.); (A.P.-V.); (I.J.)
| |
Collapse
|
48
|
Cordero-Espinoza L, Dowbaj AM, Kohler TN, Strauss B, Sarlidou O, Belenguer G, Pacini C, Martins NP, Dobie R, Wilson-Kanamori JR, Butler R, Prior N, Serup P, Jug F, Henderson NC, Hollfelder F, Huch M. Dynamic cell contacts between periportal mesenchyme and ductal epithelium act as a rheostat for liver cell proliferation. Cell Stem Cell 2021; 28:1907-1921.e8. [PMID: 34343491 PMCID: PMC8577825 DOI: 10.1016/j.stem.2021.07.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 05/19/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023]
Abstract
In the liver, ductal cells rarely proliferate during homeostasis but do so transiently after tissue injury. These cells can be expanded as organoids that recapitulate several of the cell-autonomous mechanisms of regeneration but lack the stromal interactions of the native tissue. Here, using organoid co-cultures that recapitulate the ductal-to-mesenchymal cell architecture of the portal tract, we demonstrate that a subpopulation of mouse periportal mesenchymal cells exerts dual control on proliferation of the epithelium. Ductal cell proliferation is either induced and sustained or, conversely, completely abolished, depending on the number of direct mesenchymal cell contacts, through a mechanism mediated, at least in part, by Notch signaling. Our findings expand the concept of the cellular niche in epithelial tissues, whereby not only soluble factors but also cell-cell contacts are the key regulatory cues involved in the control of cellular behaviors, suggesting a critical role for cell-cell contacts during regeneration.
Collapse
Affiliation(s)
- Lucía Cordero-Espinoza
- Wellcome Trust/Cancer Research UK Gurdon Institute, Cambridge CB2 1QN, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, Cambridge CB2 1QR, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Anna M Dowbaj
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden 01307, Germany
| | - Timo N Kohler
- Wellcome Trust-Medical Research Council Stem Cell Institute, Cambridge CB2 1QR, UK; Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK
| | - Bernhard Strauss
- Wellcome Trust/Cancer Research UK Gurdon Institute, Cambridge CB2 1QN, UK
| | - Olga Sarlidou
- Wellcome Trust/Cancer Research UK Gurdon Institute, Cambridge CB2 1QN, UK
| | - German Belenguer
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden 01307, Germany
| | - Clare Pacini
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Nuno P Martins
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden 01307, Germany
| | - Ross Dobie
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - John R Wilson-Kanamori
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Richard Butler
- Wellcome Trust/Cancer Research UK Gurdon Institute, Cambridge CB2 1QN, UK
| | - Nicole Prior
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden 01307, Germany
| | - Palle Serup
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen 2200, Denmark
| | - Florian Jug
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden 01307, Germany
| | - Neil C Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK; MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Florian Hollfelder
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK
| | - Meritxell Huch
- Wellcome Trust/Cancer Research UK Gurdon Institute, Cambridge CB2 1QN, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, Cambridge CB2 1QR, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK; Max Planck Institute of Molecular Cell Biology and Genetics, Dresden 01307, Germany.
| |
Collapse
|
49
|
Blidisel A, Marcovici I, Coricovac D, Hut F, Dehelean CA, Cretu OM. Experimental Models of Hepatocellular Carcinoma-A Preclinical Perspective. Cancers (Basel) 2021; 13:3651. [PMID: 34359553 PMCID: PMC8344976 DOI: 10.3390/cancers13153651] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC), the most frequent form of primary liver carcinoma, is a heterogenous and complex tumor type with increased incidence, poor prognosis, and high mortality. The actual therapeutic arsenal is narrow and poorly effective, rendering this disease a global health concern. Although considerable progress has been made in terms of understanding the pathogenesis, molecular mechanisms, genetics, and therapeutical approaches, several facets of human HCC remain undiscovered. A valuable and prompt approach to acquire further knowledge about the unrevealed aspects of HCC and novel therapeutic candidates is represented by the application of experimental models. Experimental models (in vivo and in vitro 2D and 3D models) are considered reliable tools to gather data for clinical usability. This review offers an overview of the currently available preclinical models frequently applied for the study of hepatocellular carcinoma in terms of initiation, development, and progression, as well as for the discovery of efficient treatments, highlighting the advantages and the limitations of each model. Furthermore, we also focus on the role played by computational studies (in silico models and artificial intelligence-based prediction models) as promising novel tools in liver cancer research.
Collapse
Affiliation(s)
- Alexandru Blidisel
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (A.B.); (F.H.); (O.M.C.)
| | - Iasmina Marcovici
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania;
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania
| | - Dorina Coricovac
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania;
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania
| | - Florin Hut
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (A.B.); (F.H.); (O.M.C.)
| | - Cristina Adriana Dehelean
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania;
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania
| | - Octavian Marius Cretu
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, RO-300041 Timișoara, Romania; (A.B.); (F.H.); (O.M.C.)
| |
Collapse
|
50
|
Tan B, Gan S, Wang X, Liu W, Li X. Applications of 3D bioprinting in tissue engineering: advantages, deficiencies, improvements, and future perspectives. J Mater Chem B 2021; 9:5385-5413. [PMID: 34124724 DOI: 10.1039/d1tb00172h] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Over the past decade, 3D bioprinting technology has progressed tremendously in the field of tissue engineering in its ability to fabricate individualized biological constructs with precise geometric designability, which offers us the capability to bridge the divergence between engineered tissue constructs and natural tissues. In this work, we first review the current widely used 3D bioprinting approaches, cells, and materials. Next, the updated applications of this technique in tissue engineering, including bone tissue, cartilage tissue, vascular grafts, skin, neural tissue, heart tissue, liver tissue and lung tissue, are briefly introduced. Then, the prominent advantages of 3D bioprinting in tissue engineering are summarized in detail: rapidly prototyping the customized structure, delivering cell-laden materials with high precision in space, and engineering with a highly controllable microenvironment. The current technical deficiencies of 3D bioprinted constructs in terms of mechanical properties and cell behaviors are afterward illustrated, as well as corresponding improvements. Finally, we conclude with future perspectives about 3D bioprinting in tissue engineering.
Collapse
Affiliation(s)
- Baosen Tan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| | - Shaolei Gan
- Jiangxi Borayer Biotech Co., Ltd, Nanchang 330052, China
| | - Xiumei Wang
- Key Laboratory of Advanced Materials of Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Wenyong Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| | - Xiaoming Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| |
Collapse
|