1
|
Ioannidis K, Dimopoulos A, Decoene I, Guilliams M, Svitina H, Storozhuk L, de Oliveira‐Silva R, Basov S, Thanh NTK, Mourdikoudis S, Van Bael MJ, Smeets B, Sakellariou D, Papantoniou I. 4D Biofabrication of Magnetically Augmented Callus Assembloid Implants Enables Rapid Endochondral Ossification via Activation of Mechanosensitive Pathways. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413680. [PMID: 39998420 PMCID: PMC12005758 DOI: 10.1002/advs.202413680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/31/2025] [Indexed: 02/26/2025]
Abstract
The use of magnetic-driven strategies for non-contact manipulation of engineered living modules opens up new possibilities for tissue engineering. The integration of magnetic nanoparticles (MNPs) with cartilaginous microtissues enables model-driven 4D bottom-up biofabrication of remotely actuated assembloids, providing unique properties to mechanoresponsive tissues, particularly skeletal constructs. However, for clinical use, the long-term effects of magnetic stimulation on phenotype and in vivo functionality need further exploration. Magnetic-driven biofabrication includes both rapid processes, such as guided microtissue assembly, and slower biological processes, like extracellular matrix secretion. This work explores the interplay between magnetic fields and MNP-loaded cartilaginous microtissues through mathematical modeling and experimental approaches, investigating long-term stimulation effects on ECM maturation and chondrogenic hypertrophy. Transcriptomic analysis reveal that magnetic stimulation activated mechanosensitive pathways and catabolic processes, driving accelerated cartilage-to-bone transitions via endochondral ossification, outcomes not observed in non-stimulated controls. This study paves the way for pre-programmed, remotely actuated skeletal assembloids with superior bone-forming capacity for regenerating challenging bone fractures.
Collapse
Affiliation(s)
- Konstantinos Ioannidis
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
| | - Andreas Dimopoulos
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
| | - Isaak Decoene
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
| | - Maya Guilliams
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
- MeBioS division, Biosystems DepartmentKU LeuvenKasteelpark, Arenberg 30Leuven3001Belgium
| | - Hanna Svitina
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
| | - Liudmyla Storozhuk
- Healthcare Biomagnetics and Nanomaterials Laboratories, Department of Medical Physics and Biomedical EngineeringUniversity College London21 Albemarle StreetLondonW1S 4BSUK
- London Centre for NanotechnologyUniversity College London17‐19 Gordon StreetLondonWC1H 0AHUK
| | - Rodrigo de Oliveira‐Silva
- Membrane Separations, Adsorption, Catalysis, and Spectroscopy for Sustainable Solutions (cMACS), Department of Microbial and Molecular SystemsKU LeuvenCelestijnenlaan 200F, PB 2454Leuven3001Belgium
| | - Sergey Basov
- Quantum Solid State Physics, Department of Physics and AstronomyKU LeuvenCelestijnenlaan 200DLeuven3001Belgium
| | - Nguyen Thi Kim Thanh
- Healthcare Biomagnetics and Nanomaterials Laboratories, Department of Medical Physics and Biomedical EngineeringUniversity College London21 Albemarle StreetLondonW1S 4BSUK
- Biophysics Group, Department of Physics and AstronomyUniversity College LondonGower StreetLondonWC1E 6BTUK
| | - Stefanos Mourdikoudis
- CINBIO, Department of Physical Chemistry, Campus Universitario, Lagoas MarcosendeUniversidade de VigoVigo36310Spain
| | - Margriet J. Van Bael
- Quantum Solid State Physics, Department of Physics and AstronomyKU LeuvenCelestijnenlaan 200DLeuven3001Belgium
| | - Bart Smeets
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
- MeBioS division, Biosystems DepartmentKU LeuvenKasteelpark, Arenberg 30Leuven3001Belgium
| | - Dimitrios Sakellariou
- Membrane Separations, Adsorption, Catalysis, and Spectroscopy for Sustainable Solutions (cMACS), Department of Microbial and Molecular SystemsKU LeuvenCelestijnenlaan 200F, PB 2454Leuven3001Belgium
| | - Ioannis Papantoniou
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
| |
Collapse
|
2
|
Mallya D, Gadre MA, Varadharajan S, Vasanthan KS. 3D bioprinting for the construction of drug testing models-development strategies and regulatory concerns. Front Bioeng Biotechnol 2025; 13:1457872. [PMID: 40028291 PMCID: PMC11868281 DOI: 10.3389/fbioe.2025.1457872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 01/14/2025] [Indexed: 03/05/2025] Open
Abstract
A drug to be successfully launched in the market requires a significant amount of capital, resources and time, where the unsuccessful results in the last stages lead to catastrophic failure for discovering drugs. This is the very reason which calls for the invention of innovative models that can closely mimic the human in vivo model for producing reliable results. Throughout the innovation line, there has been improvement in the rationale in silico designing but yet there is requirement for in vitro-in vivo correlations. During the evolving of the drug testing models, the 3D models produced by different methods have been proven to produce better results than the traditional 2D models. However, the in vitro fabrications of live tissues are still bottleneck in realizing their complete potential. There is an urgent need for the development of single, standard and simplified in vitro 3D tissue models that can be reliable for investigating the biological and pathological aspects of drug discovery, which is yet to be achieved. The existing pre-clinical models have considerable drawbacks despite being the gold standard in pre-clinical research. The major drawback being the interspecies differences and low reliability on the generated results. This gap could be overcome by the fabrication of bioengineered human disease models for drug screening. The advancement in the fabrication of 3D models will provide a valuable tool in screening drugs at different stages as they are one step closer to bio-mimic human tissues. In this review, we have discussed on the evolution of preclinical studies, and different models, including mini tissues, spheroids, organoids, bioengineered three dimensional models and organs on chips. Furthermore, we provide details of different disease models fabricated across various organs and their applications. In addition to this, the review also focuses on the limitations and the current prospects of the role of three dimensionally bioprinted models in drug screening and development.
Collapse
Affiliation(s)
- Divya Mallya
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Mrunmayi Ashish Gadre
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - S. Varadharajan
- Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Kirthanashri S. Vasanthan
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
3
|
Kim MH, Singh YP, Celik N, Yeo M, Rizk E, Hayes DJ, Ozbolat IT. High-throughput bioprinting of spheroids for scalable tissue fabrication. Nat Commun 2024; 15:10083. [PMID: 39572584 PMCID: PMC11582690 DOI: 10.1038/s41467-024-54504-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024] Open
Abstract
Tissue biofabrication mimicking organ-specific architecture and function requires physiologically-relevant cell densities. Bioprinting using spheroids can achieve this, but is limited due to the lack of practical, scalable techniques. This study presents HITS-Bio (High-throughput Integrated Tissue Fabrication System for Bioprinting), a multiarray bioprinting technique for rapidly positioning multiple spheroids simultaneously using a digitally-controlled nozzle array (DCNA). HITS-Bio achieves an unprecedented speed, ten times faster compared to existing techniques while maintaining high cell viability ( > 90%). The utility of HITS-Bio was exemplified in multiple applications, including intraoperative bioprinting with microRNA transfected human adipose-derived stem cell spheroids for calvarial bone regeneration ( ~ 30 mm3) in a rat model achieving a near-complete defect closure (bone coverage area of ~ 91% in 3 weeks and ~96% in 6 weeks). Additionally, the successful fabrication of scalable cartilage constructs (1 cm3) containing ~600 chondrogenic spheroids highlights its high-throughput efficiency (under 40 min per construct) and potential for repairing volumetric defects.
Collapse
Affiliation(s)
- Myoung Hwan Kim
- Department of Biomedical Engineering, Penn State University, University Park, PA, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA
| | - Yogendra Pratap Singh
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, USA
| | - Nazmiye Celik
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, USA
| | - Miji Yeo
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, USA
| | - Elias Rizk
- Department of Neurosurgery, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Daniel J Hayes
- Department of Biomedical Engineering, Penn State University, University Park, PA, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA
- Materials Research Institute, Penn State University, University Park, PA, USA
| | - Ibrahim T Ozbolat
- Department of Biomedical Engineering, Penn State University, University Park, PA, USA.
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA.
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, USA.
- Department of Neurosurgery, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA.
- Materials Research Institute, Penn State University, University Park, PA, USA.
- Department of Medical Oncology, Cukurova University, Adana, Turkey.
| |
Collapse
|
4
|
Golcienė B, Vaickelionienė R, Endriulaitytė U, Mickevičius V, Petrikaitė V. Synthesis and effect of 4-acetylphenylamine-based imidazole derivatives on migration and growth of 3D cultures of breast, prostate and brain cancer cells. Sci Rep 2024; 14:28065. [PMID: 39543257 PMCID: PMC11564686 DOI: 10.1038/s41598-024-76533-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/15/2024] [Indexed: 11/17/2024] Open
Abstract
In this study, we have synthesized novel 4-acetophenone moiety-bearing functionalized imidazole derivatives containing S-, and N-ethyl substituents and evaluated their anticancer activity. Their anticancer activity was studied against human breast carcinoma (MDA-MB-231), human prostate carcinoma (PPC-1), and human glioblastoma (U-87). Compounds 4, 9, 14, and 22 were identified as the most promising anticancer agents from a series of imidazole derivatives. They showed the highest cytotoxicity by MTT assay against MDA-MB-231, PPC-1 and U-87 cell lines. Compounds 14 and 22 were most selective against PPC-1 and U-87 cell lines, and their EC50 values against these cell lines ranged from 3.1 to 47.2 µM. Most tested compounds showed lower activity against the triple-negative breast cancer MDA-MB-231 cell line. None of the imidazole derivatives possessed an inhibiting effect on the migration of PPC-1 and U-87 cells by 'wound' healing assay. In spheroid assay, the most promising were compounds 14 and 22, especially in PPC-1 3D cultures. They efficiently reduced both the size and the viability of PPC-1 spheroid cells.
Collapse
Affiliation(s)
- Božena Golcienė
- Kaunas University of Technology, Radvilėnų Rd. 19, Kaunas, LT-50254, Lithuania
| | - Rita Vaickelionienė
- Kaunas University of Technology, Radvilėnų Rd. 19, Kaunas, LT-50254, Lithuania
| | - Ugnė Endriulaitytė
- Lithuanian University of Health Sciences, A. Mickevičiaus St. 9, Kaunas, LT-44307, Lithuania
| | | | - Vilma Petrikaitė
- Lithuanian University of Health Sciences, A. Mickevičiaus St. 9, Kaunas, LT-44307, Lithuania.
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio Ave. 7, Vilnius, LT-10257, Lithuania.
| |
Collapse
|
5
|
de Villiers M, Kotzé AF, du Plessis LH. Pneumatic extrusion bioprinting-based high throughput fabrication of a melanoma 3D cell culture model for anti-cancer drug screening. Biomed Mater 2024; 19:055034. [PMID: 39025118 DOI: 10.1088/1748-605x/ad651f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 07/18/2024] [Indexed: 07/20/2024]
Abstract
The high incidence of malignant melanoma highlights the need forin vitromodels that accurately represent the tumour microenvironment, enabling developments in melanoma therapy and drug screening. Despite several advancements in 3D cell culture models, appropriate melanoma models for evaluating drug efficacy are still in high demand. The 3D pneumatic extrusion-based bioprinting technology offers numerous benefits, including the ability to achieve high-throughput capabilities. However, there is a lack of research that combines pneumatic extrusion-based bioprinting with analytical assays to enable efficient drug screening in 3D melanoma models. To address this gap, this study developed a simple and highly reproducible approach to fabricate a 3D A375 melanoma cell culture model using the pneumatic extrusion-based bioprinting technology. To optimise this method, the bioprinting parameters for producing 3D cell cultures in a 96-well plate were adjusted to improve reproducibility while maintaining the desired droplet size and a cell viability of 92.13 ± 6.02%. The cross-linking method was optimised by evaluating cell viability and proliferation of the 3D bioprinted cells in three different concentrations of calcium chloride. The lower concentration of 50 mM resulted in higher cell viability and increased cell proliferation after 9 d of incubation. The A375 cells exhibited a steadier proliferation rate in the 3D bioprinted cell cultures, and tended to aggregate into spheroids, whereas the 2D cell cultures generally formed monolayered cell sheets. In addition, we evaluated the drug responses of four different anti-cancer drugs on the A375 cells in both the 2D and 3D cell cultures. The 3D cell cultures exhibited higher levels of drug resistance in all four tested anti-cancer drugs. This method presents a simple and cost-effective method of producing and analysing 3D cell culture models that do not add additional complexity to current assays and shows considerable potential for advancing 3D cell culture models' drug efficacy evaluations.
Collapse
Affiliation(s)
- Maryke de Villiers
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences North-West University, Private Bag X6001, Potchefstroom 2520, South Africa
| | - Awie F Kotzé
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences North-West University, Private Bag X6001, Potchefstroom 2520, South Africa
| | - Lissinda H du Plessis
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences North-West University, Private Bag X6001, Potchefstroom 2520, South Africa
| |
Collapse
|
6
|
Budharaju H, Sundaramurthi D, Sethuraman S. Embedded 3D bioprinting - An emerging strategy to fabricate biomimetic & large vascularized tissue constructs. Bioact Mater 2024; 32:356-384. [PMID: 37920828 PMCID: PMC10618244 DOI: 10.1016/j.bioactmat.2023.10.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/16/2023] [Accepted: 10/10/2023] [Indexed: 11/04/2023] Open
Abstract
Three-dimensional bioprinting is an advanced tissue fabrication technique that allows printing complex structures with precise positioning of multiple cell types layer-by-layer. Compared to other bioprinting methods, extrusion bioprinting has several advantages to print large-sized tissue constructs and complex organ models due to large build volume. Extrusion bioprinting using sacrificial, support and embedded strategies have been successfully employed to facilitate printing of complex and hollow structures. Embedded bioprinting is a gel-in-gel approach developed to overcome the gravitational and overhanging limits of bioprinting to print large-sized constructs with a micron-scale resolution. In embedded bioprinting, deposition of bioinks into the microgel or granular support bath will be facilitated by the sol-gel transition of the support bath through needle movement inside the granular medium. This review outlines various embedded bioprinting strategies and the polymers used in the embedded systems with advantages, limitations, and efficacy in the fabrication of complex vascularized tissues or organ models with micron-scale resolution. Further, the essential requirements of support bath systems like viscoelasticity, stability, transparency and easy extraction to print human scale organs are discussed. Additionally, the organs or complex geometries like vascular constructs, heart, bone, octopus and jellyfish models printed using support bath assisted printing methods with their anatomical features are elaborated. Finally, the challenges in clinical translation and the future scope of these embedded bioprinting models to replace the native organs are envisaged.
Collapse
Affiliation(s)
- Harshavardhan Budharaju
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Center for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Center, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Dhakshinamoorthy Sundaramurthi
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Center for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Center, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Center for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Center, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| |
Collapse
|
7
|
Kasturi M, Mathur V, Gadre M, Srinivasan V, Vasanthan KS. Three Dimensional Bioprinting for Hepatic Tissue Engineering: From In Vitro Models to Clinical Applications. Tissue Eng Regen Med 2024; 21:21-52. [PMID: 37882981 PMCID: PMC10764711 DOI: 10.1007/s13770-023-00576-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 10/27/2023] Open
Abstract
Fabrication of functional organs is the holy grail of tissue engineering and the possibilities of repairing a partial or complete liver to treat chronic liver disorders are discussed in this review. Liver is the largest gland in the human body and plays a responsible role in majority of metabolic function and processes. Chronic liver disease is one of the leading causes of death globally and the current treatment strategy of organ transplantation holds its own demerits. Hence there is a need to develop an in vitro liver model that mimics the native microenvironment. The developed model should be a reliable to understand the pathogenesis, screen drugs and assist to repair and replace the damaged liver. The three-dimensional bioprinting is a promising technology that recreates in vivo alike in vitro model for transplantation, which is the goal of tissue engineers. The technology has great potential due to its precise control and its ability to homogeneously distribute cells on all layers in a complex structure. This review gives an overview of liver tissue engineering with a special focus on 3D bioprinting and bioinks for liver disease modelling and drug screening.
Collapse
Affiliation(s)
- Meghana Kasturi
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Vidhi Mathur
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Mrunmayi Gadre
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Varadharajan Srinivasan
- Department of Civil Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Kirthanashri S Vasanthan
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
8
|
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
|
9
|
Sanchez‐Rubio A, Jayawarna V, Maxwell E, Dalby MJ, Salmeron‐Sanchez M. Keeping It Organized: Multicompartment Constructs to Mimic Tissue Heterogeneity. Adv Healthc Mater 2023; 12:e2202110. [PMID: 36938891 PMCID: PMC11469230 DOI: 10.1002/adhm.202202110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 02/17/2023] [Indexed: 03/21/2023]
Abstract
Tissue engineering aims at replicating tissues and organs to develop applications in vivo and in vitro. In vivo, by engineering artificial constructs using functional materials and cells to provide both physiological form and function. In vitro, by engineering three-dimensional (3D) models to support drug discovery and enable understanding of fundamental biology. 3D culture constructs mimic cell-cell and cell-matrix interactions and use biomaterials seeking to increase the resemblance of engineered tissues with its in vivo homologues. Native tissues, however, include complex architectures, with compartmentalized regions of different properties containing different types of cells that can be captured by multicompartment constructs. Recent advances in fabrication technologies, such as micropatterning, microfluidics or 3D bioprinting, have enabled compartmentalized structures with defined compositions and properties that are essential in creating 3D cell-laden multiphasic complex architectures. This review focuses on advances in engineered multicompartment constructs that mimic tissue heterogeneity. It includes multiphasic 3D implantable scaffolds and in vitro models, including systems that incorporate different regions emulating in vivo tissues, highlighting the emergence and relevance of 3D bioprinting in the future of biological research and medicine.
Collapse
Affiliation(s)
| | - Vineetha Jayawarna
- Centre for the Cellular MicroenvironmentUniversity of GlasgowGlasgowG11 6EWUK
| | - Emily Maxwell
- Centre for the Cellular MicroenvironmentUniversity of GlasgowGlasgowG11 6EWUK
| | - Matthew J. Dalby
- Centre for the Cellular MicroenvironmentUniversity of GlasgowGlasgowG11 6EWUK
| | | |
Collapse
|
10
|
Ostrovidov S, Ramalingam M, Bae H, Orive G, Fujie T, Shi X, Kaji H. Latest developments in engineered skeletal muscle tissues for drug discovery and development. Expert Opin Drug Discov 2023; 18:47-63. [PMID: 36535280 DOI: 10.1080/17460441.2023.2160438] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION With the advances in skeletal muscle tissue engineering, new platforms have arisen with important applications in biology studies, disease modeling, and drug testing. Current developments highlight the quest for engineering skeletal muscle tissues with higher complexity . These new human skeletal muscle tissue models will be powerful tools for drug discovery and development and disease modeling. AREAS COVERED The authors review the latest advances in in vitro models of engineered skeletal muscle tissues used for testing drugs with a focus on the use of four main cell culture techniques: Cell cultures in well plates, in microfluidics, in organoids, and in bioprinted constructs. Additional information is provided on the satellite cell niche. EXPERT OPINION In recent years, more sophisticated in vitro models of skeletal muscle tissues have been fabricated. Important developments have been made in stem cell research and in the engineering of human skeletal muscle tissue. Some platforms have already started to be used for drug testing, notably those based on the parameters of hypertrophy/atrophy and the contractibility of myotubes. More developments are expected through the use of multicellular types and multi-materials as matrices . The validation and use of these models in drug testing should now increase.
Collapse
Affiliation(s)
- Serge Ostrovidov
- Department of Biomechanics, Institute of Biomaterials and Bioengineering (IBB), Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Murugan Ramalingam
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science, BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan, Republic of Korea.,School of Basic Medical Science, Chengdu University, Chengdu, Sichuan, China.,Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Republic of Korea.,Department of Metallurgical and Materials Engineering, Atilim University, Ankara, Turkey
| | - Hojae Bae
- KU Convergence Science and Technology Institute, Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Hwayang-dong, Kwangjin-gu, Seoul, Republic of Korea
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain.,Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.,Biomaterials and Nanomedicine (CIBER-BBN), Biomedical Research Networking Centre in Bioengineering, Vitoria-Gasteiz, Spain
| | - Toshinori Fujie
- Department of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Xuetao Shi
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, China
| | - Hirokazu Kaji
- Department of Biomechanics, Institute of Biomaterials and Bioengineering (IBB), Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
11
|
Anthon SG, Valente KP. Vascularization Strategies in 3D Cell Culture Models: From Scaffold-Free Models to 3D Bioprinting. Int J Mol Sci 2022; 23:14582. [PMID: 36498908 PMCID: PMC9737506 DOI: 10.3390/ijms232314582] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/21/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
The discrepancies between the findings in preclinical studies, and in vivo testing and clinical trials have resulted in the gradual decline in drug approval rates over the past decades. Conventional in vitro drug screening platforms employ two-dimensional (2D) cell culture models, which demonstrate inaccurate drug responses by failing to capture the three-dimensional (3D) tissue microenvironment in vivo. Recent advancements in the field of tissue engineering have made possible the creation of 3D cell culture systems that can accurately recapitulate the cell-cell and cell-extracellular matrix interactions, as well as replicate the intricate microarchitectures observed in native tissues. However, the lack of a perfusion system in 3D cell cultures hinders the establishment of the models as potential drug screening platforms. Over the years, multiple techniques have successfully demonstrated vascularization in 3D cell cultures, simulating in vivo-like drug interactions, proposing the use of 3D systems as drug screening platforms to eliminate the deviations between preclinical and in vivo testing. In this review, the basic principles of 3D cell culture systems are briefly introduced, and current research demonstrating the development of vascularization in 3D cell cultures is discussed, with a particular focus on the potential of these models as the future of drug screening platforms.
Collapse
Affiliation(s)
- Shamapto Guha Anthon
- Department of Biomedical Engineering, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | | |
Collapse
|
12
|
Prabhakaran P, Palaniyandi T, Kanagavalli B, Ram Kumar V, Hari R, Sandhiya V, Baskar G, Rajendran BK, Sivaji A. Prospect and retrospect of 3D bio-printing. Acta Histochem 2022; 124:151932. [PMID: 36027838 DOI: 10.1016/j.acthis.2022.151932] [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: 06/14/2022] [Revised: 07/23/2022] [Accepted: 07/23/2022] [Indexed: 11/01/2022]
Abstract
3D bioprinting has become a popular medical technique in recent years. The most compelling rationale for the development of 3D bioprinting is the paucity of biological structures required for the rehabilitation of missing organs and tissues. They're useful in a multitude of domains, including disease modelling, regenerative medicine, tissue engineering, drug discovery with testing, personalised medicine, organ development, toxicity studies, and implants. Bioprinting requires a range of bioprinting technologies and bioinks to finish their procedure, that Inkjet-based bioprinting, extrusion-based bioprinting, laser-assisted bioprinting, stereolithography-based bioprinting, and in situ bioprinting are some of the technologies listed here. Bioink is a 3D printing material that is used to construct engineered artificial living tissue. It can be constructed solely for cells, but it usually includes a carrier substance that envelops the cells, then there's Agarose-based bioinks, alginate-based bioinks, collagen-based bioinks, and hyaluronic acid-based bioinks, to name a few. Here we presented about the different bioprinting methods with the use of bioinks in it and then Prospected over various applications in different fields.
Collapse
Affiliation(s)
- Pranav Prabhakaran
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to University, Chennai, India
| | - Thirunavukkarsu Palaniyandi
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to University, Chennai, India; Department of Anatomy, Biomedical Reseach Unit and Laboratory Animal Centre, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| | - B Kanagavalli
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to University, Chennai, India
| | - V Ram Kumar
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to University, Chennai, India
| | - Rajeswari Hari
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to University, Chennai, India
| | - V Sandhiya
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to University, Chennai, India
| | - Gomathy Baskar
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to University, Chennai, India
| | | | - Asha Sivaji
- Department of Biochemistry, DKM College for Women, Vellore, India
| |
Collapse
|
13
|
Shukla P, Yeleswarapu S, Heinrich M, Prakash J, Pati F. Mimicking Tumor Microenvironment by 3D Bioprinting: 3D Cancer Modeling. Biofabrication 2022; 14. [PMID: 35512666 DOI: 10.1088/1758-5090/ac6d11] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 05/05/2022] [Indexed: 11/12/2022]
Abstract
The tumor microenvironment typically comprises cancer cells, tumor vasculature, stromal components like fibroblasts, and host immune cells that assemble to support tumorigenesis. However, preexisting classic cancer models like 2D cell culture methods, 3D cancer spheroids, and tumor organoids seem to lack essential tumor microenvironment components. 3D bioprinting offers enormous advantages for developing in vitro tumor models by allowing user-controlled deposition of multiple biomaterials, cells, and biomolecules in a predefined architecture. This review highlights the recent developments in 3D cancer modeling using different bioprinting techniques to recreate the TME. 3D bioprinters enable fabrication of high-resolution microstructures to reproduce TME intricacies. Furthermore, 3D bioprinted models can be applied as a preclinical model for versatile research applications in the tumor biology and pharmaceutical industries. These models provide an opportunity to develop high-throughput drug screening platforms and can further be developed to suit individual patient requirements hence giving a boost to the field of personalized anti-cancer therapeutics. We underlined the various ways the existing studies have tried to mimic the TME, mimic the hallmark events of cancer growth and metastasis within the 3D bioprinted models and showcase the 3D drug-tumor interaction and further utilization of such models to develop personalized medicine.
Collapse
Affiliation(s)
- Priyanshu Shukla
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Hyderabad, Telangana, 502285, INDIA
| | - Sriya Yeleswarapu
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Hyderabad, Telangana, 502285, INDIA
| | - Marcel Heinrich
- Department of Biomaterials, Science and Technology, University of Twente Faculty of Science and Technology, Department of Biomaterials, Science and Technology, Faculty of Science and Technology, University of Twente, Drienerlolaan 5, 7500AE, Enschede, The Netherlands, Enschede, Overijssel, 7500 AE, NETHERLANDS
| | - Jai Prakash
- Department of Biomaterials, Science and Technology, University of Twente Faculty of Science and Technology, Department of Biomaterials, Science and Technology, Faculty of Science and Technology, University of Twente, Drienerlolaan 5, 7500AE, Enschede, The Netherlands, Enschede, Overijssel, 7500 AE, NETHERLANDS
| | - Falguni Pati
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Hyderabad, Telangana, 502285, INDIA
| |
Collapse
|
14
|
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: 20] [Impact Index Per Article: 6.7] [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
|
15
|
Ramadan Q, Zourob M. 3D Bioprinting at the Frontier of Regenerative Medicine, Pharmaceutical, and Food Industries. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 2:607648. [PMID: 35047890 PMCID: PMC8757855 DOI: 10.3389/fmedt.2020.607648] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/08/2020] [Indexed: 12/22/2022] Open
Abstract
3D printing technology has emerged as a key driver behind an ongoing paradigm shift in the production process of various industrial domains. The integration of 3D printing into tissue engineering, by utilizing life cells which are encapsulated in specific natural or synthetic biomaterials (e.g., hydrogels) as bioinks, is paving the way toward devising many innovating solutions for key biomedical and healthcare challenges and heralds' new frontiers in medicine, pharmaceutical, and food industries. Here, we present a synthesis of the available 3D bioprinting technology from what is found and what has been achieved in various applications and discussed the capabilities and limitations encountered in this technology.
Collapse
Affiliation(s)
- Qasem Ramadan
- College of Science and General Studies, Alfaisal University, Riyadh, Saudi Arabia
| | - Mohammed Zourob
- College of Science and General Studies, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
16
|
Dermal Delivery of Lipid Nanoparticles: Effects on Skin and Assessment of Absorption and Safety. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1357:83-114. [DOI: 10.1007/978-3-030-88071-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
17
|
Abstract
AbstractThe multidisciplinary research field of bioprinting combines additive manufacturing, biology and material sciences to create bioconstructs with three-dimensional architectures mimicking natural living tissues. The high interest in the possibility of reproducing biological tissues and organs is further boosted by the ever-increasing need for personalized medicine, thus allowing bioprinting to establish itself in the field of biomedical research, and attracting extensive research efforts from companies, universities, and research institutes alike. In this context, this paper proposes a scientometric analysis and critical review of the current literature and the industrial landscape of bioprinting to provide a clear overview of its fast-changing and complex position. The scientific literature and patenting results for 2000–2020 are reviewed and critically analyzed by retrieving 9314 scientific papers and 309 international patents in order to draw a picture of the scientific and industrial landscape in terms of top research countries, institutions, journals, authors and topics, and identifying the technology hubs worldwide. This review paper thus offers a guide to researchers interested in this field or to those who simply want to understand the emerging trends in additive manufacturing and 3D bioprinting.
Graphic abstract
Collapse
|
18
|
Sbirkov Y, Molander D, Milet C, Bodurov I, Atanasov B, Penkov R, Belev N, Forraz N, McGuckin C, Sarafian V. A Colorectal Cancer 3D Bioprinting Workflow as a Platform for Disease Modeling and Chemotherapeutic Screening. Front Bioeng Biotechnol 2021; 9:755563. [PMID: 34869264 PMCID: PMC8638705 DOI: 10.3389/fbioe.2021.755563] [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: 08/09/2021] [Accepted: 09/20/2021] [Indexed: 12/23/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common malignancy and has recently moved up to the second leading cause of death among carcinomas. Prognosis, especially for advanced diseases or certain molecular subtypes of CRC, remains poor, which highlights the urgent need for better therapeutic strategies. However, currently, as little as 0.1% of all drugs make it from bench to bedside because of the inherently high false-positive and false-negative rates of current preclinical and clinical drug testing data. Therefore, the success of developing novel treatment agents lies in the introduction of improved preclinical disease models which resemble in vivo carcinomas closer, possess higher predictive properties, and offer opportunities for individualized therapies. Aiming to address these needs, we have established an affordable, flexible, and highly reproducible 3D bioprinted CRC model. The histological assessment of Caco-2 cells in 3D bioprints revealed the formation of glandular-like structures which show greater pathomorphological resemblance to tumors than monolayer cultures do. RNA expression profiles in 3D bioprinted cells were marked by upregulation of genes involved in cell adhesion, hypoxia, EGFR/KRAS signaling, and downregulation of cell cycle programs. Testing this 3D experimental platform with three of the most commonly used chemotherapeutics in CRC (5-fluoruracil, oxaliplatin, and irinotecan) revealed overall increased resistance compared to 2D cell cultures. Last, we demonstrate that our workflow can be successfully extended to primary CRC samples. Thereby, we describe a novel accessible platform for disease modeling and drug testing, which may present an innovative opportunity for personalized therapeutic screening.
Collapse
Affiliation(s)
- Yordan Sbirkov
- Department of Medical Biology, Medical University of Plovdiv, Plovdiv, Bulgaria
- Research Institute at Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Diana Molander
- Department of Medical Biology, Medical University of Plovdiv, Plovdiv, Bulgaria
- Research Institute at Medical University of Plovdiv, Plovdiv, Bulgaria
| | | | - Ilia Bodurov
- UMHAT-Eurohospital, Clinical Pathology Department, Plovdiv, Bulgaria
| | - Boyko Atanasov
- UMHAT-Eurohospital, Surgical Department, Plovdiv, Bulgaria
- Department of Propaedeutics of Surgical Diseases, Medical University of Plovdiv, Plovdiv, Bulgaria
| | | | - Nikolay Belev
- Research Institute at Medical University of Plovdiv, Plovdiv, Bulgaria
- UMHAT-Eurohospital, Surgical Department, Plovdiv, Bulgaria
- Medical Simulation Training Centre, Medical University of Plovdiv, Plovdiv, Bulgaria
| | | | | | - Victoria Sarafian
- Department of Medical Biology, Medical University of Plovdiv, Plovdiv, Bulgaria
- Research Institute at Medical University of Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
19
|
Engberg A, Stelzl C, Eriksson O, O'Callaghan P, Kreuger J. An open source extrusion bioprinter based on the E3D motion system and tool changer to enable FRESH and multimaterial bioprinting. Sci Rep 2021; 11:21547. [PMID: 34732783 PMCID: PMC8566469 DOI: 10.1038/s41598-021-00931-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/20/2021] [Indexed: 12/29/2022] Open
Abstract
Bioprinting is increasingly used to create complex tissue constructs for an array of research applications, and there are also increasing efforts to print tissues for transplantation. Bioprinting may also prove valuable in the context of drug screening for personalized medicine for treatment of diseases such as cancer. However, the rapidly expanding bioprinting research field is currently limited by access to bioprinters. To increase the availability of bioprinting technologies we present here an open source extrusion bioprinter based on the E3D motion system and tool changer to enable high-resolution multimaterial bioprinting. As proof of concept, the bioprinter is used to create collagen constructs using freeform reversible embedding of suspended hydrogels (FRESH) methodology, as well as multimaterial constructs composed of distinct sections of laminin and collagen. Data is presented demonstrating that the bioprinted constructs support growth of cells either seeded onto printed constructs or included in the bioink prior to bioprinting. This open source bioprinter is easily adapted for different bioprinting applications, and additional tools can be incorporated to increase the capabilities of the system.
Collapse
Affiliation(s)
- Adam Engberg
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Christina Stelzl
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Olle Eriksson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Paul O'Callaghan
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Johan Kreuger
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
20
|
Bioprintable Lung Extracellular Matrix Hydrogel Scaffolds for 3D Culture of Mesenchymal Stromal Cells. Polymers (Basel) 2021; 13:polym13142350. [PMID: 34301107 PMCID: PMC8309540 DOI: 10.3390/polym13142350] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stromal cell (MSC)-based cell therapy in acute respiratory diseases is based on MSC secretion of paracrine factors. Several strategies have proposed to improve this are being explored including pre-conditioning the MSCs prior to administration. We here propose a strategy for improving the therapeutic efficacy of MSCs based on cell preconditioning by growing them in native extracellular matrix (ECM) derived from the lung. To this end, a bioink with tunable stiffness based on decellularized porcine lung ECM hydrogels was developed and characterized. The bioink was suitable for 3D culturing of lung-resident MSCs without the need for additional chemical or physical crosslinking. MSCs showed good viability, and contraction assays showed the existence of cell–matrix interactions in the bioprinted scaffolds. Adhesion capacity and length of the focal adhesions formed were increased for the cells cultured within the lung hydrogel scaffolds. Also, there was more than a 20-fold increase of the expression of the CXCR4 receptor in the 3D-cultured cells compared to the cells cultured in plastic. Secretion of cytokines when cultured in an in vitro model of lung injury showed a decreased secretion of pro-inflammatory mediators for the cells cultured in the 3D scaffolds. Moreover, the morphology of the harvested cells was markedly different with respect to conventionally (2D) cultured MSCs. In conclusion, the developed bioink can be used to bioprint structures aimed to improve preconditioning MSCs for therapeutic purposes.
Collapse
|
21
|
Parihar A, Pandita V, Kumar A, Parihar DS, Puranik N, Bajpai T, Khan R. 3D Printing: Advancement in Biogenerative Engineering to Combat Shortage of Organs and Bioapplicable Materials. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2021; 8:173-199. [PMID: 34230892 PMCID: PMC8252697 DOI: 10.1007/s40883-021-00219-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/26/2021] [Accepted: 06/08/2021] [Indexed: 02/06/2023]
Abstract
ABSTRACT Organ or cell transplantation is medically evaluated for end-stage failure saving or extending the lives of thousands of patients who are suffering from organ failure disorders. The unavailability of adequate organs for transplantation to meet the existing demand is a major challenge in the medical field. This led to day-day-increase in the number of patients on transplant waiting lists as well as in the number of patients dying while on the queue. Recently, technological advancements in the field of biogenerative engineering have the potential to regenerate tissues and, in some cases, create new tissues and organs. In this context, major advances and innovations are being made in the fields of tissue engineering and regenerative medicine which have a huge impact on the scientific community is three-dimensional bioprinting (3D bioprinting) of tissues and organs. Besides this, the decellularization of organs and using this as a scaffold for generating new organs through the recellularization process shows promising results. This review discussed about current approaches for tissue and organ engineering including methods of scaffold designing, recent advances in 3D bioprinting, organs regenerated successfully using 3D printing, and extended application of 3D bioprinting technique in the field of medicine. Besides this, information about commercially available 3D printers has also been included in this article. LAY SUMMARY Today's need for organs for the transplantation process in order to save a patient's life or to enhance the survival rate of diseased one is the prime concern among the scientific community. Recent, advances in the field of biogenerative engineering have the potential to regenerate tissues and create organs compatible with the patient's body. In this context, major advances and innovations are being made in the fields of tissue engineering and regenerative medicine which have a huge impact on the scientific community is three-dimensional bioprinting (3D bioprinting) of tissues and organs. Besides this, the decellularization of organs and using this as a scaffold for generating new organs through the recellularization process shows promising results. This review dealt with the current approaches for tissue and organ engineering including methods of scaffold designing, recent advances in 3D bioprinting, organs regenerated successfully using 3D printing, and extended application of 3D bioprinting technique in the field of medicine. Furthermore, information about commercially available 3D printers has also been included in this article.
Collapse
Affiliation(s)
- Arpana Parihar
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, Madhya Pradesh 462026 India
- Microfluidics & MEMS Centre, CSIR-Advanced Materials and Processes Research Institute (AMPRI), Hoshangabad Road Bhopal, 462026 India
| | - Vasundhara Pandita
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, Madhya Pradesh 462026 India
| | - Avinash Kumar
- Department of Mechanical Engineering, Indian Institute of Information Technology, Design & Manufacturing (IIITD&M), Kancheepuram, 600127 India
| | - Dipesh Singh Parihar
- Engineering College Tuwa , At. & Post. Tuwa, Taluka Godhra, Dist. Panchmahal, Godhra, Gujarat 388713 India
| | - Nidhi Puranik
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, Madhya Pradesh 462026 India
| | - Tapas Bajpai
- Department of Mechanical Engineering, Malaviya National Institute of Technology, Jaipur, 302017 India
| | - Raju Khan
- Microfluidics & MEMS Centre, CSIR-Advanced Materials and Processes Research Institute (AMPRI), Hoshangabad Road Bhopal, 462026 India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-AMPRI, Bhopal, 462026 India
| |
Collapse
|
22
|
Kronemberger GS, Miranda GASC, Tavares RSN, Montenegro B, Kopke ÚDA, Baptista LS. Recapitulating Tumorigenesis in vitro: Opportunities and Challenges of 3D Bioprinting. Front Bioeng Biotechnol 2021; 9:682498. [PMID: 34239860 PMCID: PMC8258101 DOI: 10.3389/fbioe.2021.682498] [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: 03/18/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer is considered one of the most predominant diseases in the world and one of the principal causes of mortality per year. The cellular and molecular mechanisms involved in the development and establishment of solid tumors can be defined as tumorigenesis. Recent technological advances in the 3D cell culture field have enabled the recapitulation of tumorigenesis in vitro, including the complexity of stromal microenvironment. The establishment of these 3D solid tumor models has a crucial role in personalized medicine and drug discovery. Recently, spheroids and organoids are being largely explored as 3D solid tumor models for recreating tumorigenesis in vitro. In spheroids, the solid tumor can be recreated from cancer cells, cancer stem cells, stromal and immune cell lineages. Organoids must be derived from tumor biopsies, including cancer and cancer stem cells. Both models are considered as a suitable model for drug assessment and high-throughput screening. The main advantages of 3D bioprinting are its ability to engineer complex and controllable 3D tissue models in a higher resolution. Although 3D bioprinting represents a promising technology, main challenges need to be addressed to improve the results in cancer research. The aim of this review is to explore (1) the principal cell components and extracellular matrix composition of solid tumor microenvironment; (2) the recapitulation of tumorigenesis in vitro using spheroids and organoids as 3D culture models; and (3) the opportunities, challenges, and applications of 3D bioprinting in this area.
Collapse
Affiliation(s)
- Gabriela S. Kronemberger
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro Xerém, Duque de Caxias, Brazil
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
- Post-graduation Program of Translational Biomedicine (Biotrans), Unigranrio, Duque de Caxias, Brazil
| | - Guilherme A. S. C. Miranda
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro Xerém, Duque de Caxias, Brazil
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
- Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| | - Renata S. N. Tavares
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| | - Bianca Montenegro
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro Xerém, Duque de Caxias, Brazil
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
- Post-graduation Program of Translational Biomedicine (Biotrans), Unigranrio, Duque de Caxias, Brazil
| | - Úrsula de A. Kopke
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro Xerém, Duque de Caxias, Brazil
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| | - Leandra S. Baptista
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro Xerém, Duque de Caxias, Brazil
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
- Post-graduation Program of Translational Biomedicine (Biotrans), Unigranrio, Duque de Caxias, Brazil
- Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| |
Collapse
|
23
|
Chen EP, Toksoy Z, Davis BA, Geibel JP. 3D Bioprinting of Vascularized Tissues for in vitro and in vivo Applications. Front Bioeng Biotechnol 2021; 9:664188. [PMID: 34055761 PMCID: PMC8158943 DOI: 10.3389/fbioe.2021.664188] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/06/2021] [Indexed: 12/23/2022] Open
Abstract
With a limited supply of organ donors and available organs for transplantation, the aim of tissue engineering with three-dimensional (3D) bioprinting technology is to construct fully functional and viable tissue and organ replacements for various clinical applications. 3D bioprinting allows for the customization of complex tissue architecture with numerous combinations of materials and printing methods to build different tissue types, and eventually fully functional replacement organs. The main challenge of maintaining 3D printed tissue viability is the inclusion of complex vascular networks for nutrient transport and waste disposal. Rapid development and discoveries in recent years have taken huge strides toward perfecting the incorporation of vascular networks in 3D printed tissue and organs. In this review, we will discuss the latest advancements in fabricating vascularized tissue and organs including novel strategies and materials, and their applications. Our discussion will begin with the exploration of printing vasculature, progress through the current statuses of bioprinting tissue/organoids from bone to muscles to organs, and conclude with relevant applications for in vitro models and drug testing. We will also explore and discuss the current limitations of vascularized tissue engineering and some of the promising future directions this technology may bring.
Collapse
Affiliation(s)
- Earnest P Chen
- Department of Surgery, School of Medicine, Yale University, New Haven, CT, United States.,Yale College, Yale University, New Haven, CT, United States
| | - Zeren Toksoy
- Department of Surgery, School of Medicine, Yale University, New Haven, CT, United States.,Yale College, Yale University, New Haven, CT, United States
| | - Bruce A Davis
- Department of Surgery, School of Medicine, Yale University, New Haven, CT, United States.,Department of Cellular and Molecular Physiology, School of Medicine, Yale University, New Haven, CT, United States
| | - John P Geibel
- Department of Surgery, School of Medicine, Yale University, New Haven, CT, United States.,Department of Cellular and Molecular Physiology, School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
24
|
Svozilová H, Plichta Z, Proks V, Studená R, Baloun J, Doubek M, Pospíšilová Š, Horák D. RGDS-Modified Superporous Poly(2-Hydroxyethyl Methacrylate)-Based Scaffolds as 3D In Vitro Leukemia Model. Int J Mol Sci 2021; 22:ijms22052376. [PMID: 33673496 PMCID: PMC7956824 DOI: 10.3390/ijms22052376] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 01/05/2023] Open
Abstract
Superporous poly(2-hydroxyethyl methacrylate-co-2-aminoethyl methacrylate) (P(HEMA-AEMA)) hydrogel scaffolds are designed for in vitro 3D culturing of leukemic B cells. Hydrogel porosity, which influences cell functions and growth, is introduced by adding ammonium oxalate needle-like crystals in the polymerization mixture. To improve cell vitality, cell-adhesive Arg-Gly-Asp-Ser (RGDS) peptide is immobilized on the N-(γ-maleimidobutyryloxy)succinimide-activated P(HEMA-AEMA) hydrogels via reaction of SH with maleimide groups. This modification is especially suitable for the survival of primary chronic lymphocytic leukemia cells (B-CLLs) in 3D cell culture. No other tested stimuli (interleukin-4, CD40 ligand, or shaking) can further improve B-CLL survival or metabolic activity. Both unmodified and RGDS-modified P(HEMA-AEMA) scaffolds serve as a long-term (70 days) 3D culture platforms for HS-5 and M2-10B4 bone marrow stromal cell lines and MEC-1 and HG-3 B-CLL cell lines, although the adherent cells retain their physiological morphologies, preferably on RGDS-modified hydrogels. Moreover, the porosity of hydrogels allows direct cell lysis, followed by efficient DNA isolation from the 3D-cultured cells. P(HEMA-AEMA)-RGDS thus serves as a suitable 3D in vitro leukemia model that enables molecular and metabolic assays and allows imaging of cell morphology, interactions, and migration by confocal microscopy. Such applications can prospectively assist in testing of drugs to treat this frequently recurring or refractory cancer.
Collapse
Affiliation(s)
- Hana Svozilová
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; (H.S.); (R.S.); (J.B.); (M.D.); (Š.P.)
- Department of Internal Medicine—Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Jihlavská 20, 625 00 Brno, Czech Republic
| | - Zdeněk Plichta
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06 Prague, Czech Republic; (Z.P.); (V.P.)
| | - Vladimír Proks
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06 Prague, Czech Republic; (Z.P.); (V.P.)
| | - Radana Studená
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; (H.S.); (R.S.); (J.B.); (M.D.); (Š.P.)
| | - Jiří Baloun
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; (H.S.); (R.S.); (J.B.); (M.D.); (Š.P.)
| | - Michael Doubek
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; (H.S.); (R.S.); (J.B.); (M.D.); (Š.P.)
- Department of Internal Medicine—Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Jihlavská 20, 625 00 Brno, Czech Republic
| | - Šárka Pospíšilová
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; (H.S.); (R.S.); (J.B.); (M.D.); (Š.P.)
- Department of Internal Medicine—Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Jihlavská 20, 625 00 Brno, Czech Republic
| | - Daniel Horák
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06 Prague, Czech Republic; (Z.P.); (V.P.)
- Correspondence: ; Tel.: +420-296-809-260
| |
Collapse
|
25
|
Morales X, Cortés-Domínguez I, Ortiz-de-Solorzano C. Modeling the Mechanobiology of Cancer Cell Migration Using 3D Biomimetic Hydrogels. Gels 2021; 7:17. [PMID: 33673091 PMCID: PMC7930983 DOI: 10.3390/gels7010017] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/29/2021] [Accepted: 02/09/2021] [Indexed: 02/06/2023] Open
Abstract
Understanding how cancer cells migrate, and how this migration is affected by the mechanical and chemical composition of the extracellular matrix (ECM) is critical to investigate and possibly interfere with the metastatic process, which is responsible for most cancer-related deaths. In this article we review the state of the art about the use of hydrogel-based three-dimensional (3D) scaffolds as artificial platforms to model the mechanobiology of cancer cell migration. We start by briefly reviewing the concept and composition of the extracellular matrix (ECM) and the materials commonly used to recreate the cancerous ECM. Then we summarize the most relevant knowledge about the mechanobiology of cancer cell migration that has been obtained using 3D hydrogel scaffolds, and relate those discoveries to what has been observed in the clinical management of solid tumors. Finally, we review some recent methodological developments, specifically the use of novel bioprinting techniques and microfluidics to create realistic hydrogel-based models of the cancer ECM, and some of their applications in the context of the study of cancer cell migration.
Collapse
Affiliation(s)
| | | | - Carlos Ortiz-de-Solorzano
- IDISNA, Ciberonc and Solid Tumors and Biomarkers Program, Center for Applied Medical Research, University of Navarra, 31008 Pamplona, Spain; (X.M.); (I.C.-D.)
| |
Collapse
|
26
|
Qian L, TCW J. Human iPSC-Based Modeling of Central Nerve System Disorders for Drug Discovery. Int J Mol Sci 2021; 22:1203. [PMID: 33530458 PMCID: PMC7865494 DOI: 10.3390/ijms22031203] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 02/07/2023] Open
Abstract
A high-throughput drug screen identifies potentially promising therapeutics for clinical trials. However, limitations that persist in current disease modeling with limited physiological relevancy of human patients skew drug responses, hamper translation of clinical efficacy, and contribute to high clinical attritions. The emergence of induced pluripotent stem cell (iPSC) technology revolutionizes the paradigm of drug discovery. In particular, iPSC-based three-dimensional (3D) tissue engineering that appears as a promising vehicle of in vitro disease modeling provides more sophisticated tissue architectures and micro-environmental cues than a traditional two-dimensional (2D) culture. Here we discuss 3D based organoids/spheroids that construct the advanced modeling with evolved structural complexity, which propels drug discovery by exhibiting more human specific and diverse pathologies that are not perceived in 2D or animal models. We will then focus on various central nerve system (CNS) disease modeling using human iPSCs, leading to uncovering disease pathogenesis that guides the development of therapeutic strategies. Finally, we will address new opportunities of iPSC-assisted drug discovery with multi-disciplinary approaches from bioengineering to Omics technology. Despite technological challenges, iPSC-derived cytoarchitectures through interactions of diverse cell types mimic patients' CNS and serve as a platform for therapeutic development and personalized precision medicine.
Collapse
Affiliation(s)
- Lu Qian
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Ronald Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Julia TCW
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Ronald Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
27
|
|
28
|
Gordeev EG, Ananikov VP. Widely accessible 3D printing technologies in chemistry, biochemistry and pharmaceutics: applications, materials and prospects. RUSSIAN CHEMICAL REVIEWS 2020. [DOI: 10.1070/rcr4980] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
29
|
Pahlevanzadeh F, Mokhtari H, Bakhsheshi-Rad HR, Emadi R, Kharaziha M, Valiani A, Poursamar SA, Ismail AF, RamaKrishna S, Berto F. Recent Trends in Three-Dimensional Bioinks Based on Alginate for Biomedical Applications. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E3980. [PMID: 32911867 PMCID: PMC7557490 DOI: 10.3390/ma13183980] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/28/2020] [Accepted: 09/01/2020] [Indexed: 02/06/2023]
Abstract
Three-dimensional (3D) bioprinting is an appealing and revolutionary manufacturing approach for the accurate placement of biologics, such as living cells and extracellular matrix (ECM) components, in the form of a 3D hierarchical structure to fabricate synthetic multicellular tissues. Many synthetic and natural polymers are applied as cell printing bioinks. One of them, alginate (Alg), is an inexpensive biomaterial that is among the most examined hydrogel materials intended for vascular, cartilage, and bone tissue printing. It has also been studied pertaining to the liver, kidney, and skin, due to its excellent cell response and flexible gelation preparation through divalent ions including calcium. Nevertheless, Alg hydrogels possess certain negative aspects, including weak mechanical characteristics, poor printability, poor structural stability, and poor cell attachment, which may restrict its usage along with the 3D printing approach to prepare artificial tissue. In this review paper, we prepare the accessible materials to be able to encourage and boost new Alg-based bioink formulations with superior characteristics for upcoming purposes in drug delivery systems. Moreover, the major outcomes are discussed, and the outstanding concerns regarding this area and the scope for upcoming examination are outlined.
Collapse
Affiliation(s)
- Farnoosh Pahlevanzadeh
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
- Department of Anatomical Science, School of Medicine, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| | - Hamidreza Mokhtari
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Hamid Reza Bakhsheshi-Rad
- Advanced Materials Research Center, Department of Materials Engineering, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Rahmatollah Emadi
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Mahshid Kharaziha
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Ali Valiani
- Department of Anatomical Science, School of Medicine, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| | - S Ali Poursamar
- Biomaterials, Nanotechnology, and Tissue Engineering Group, Advanced Medical Technology Department, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| | - Ahmad Fauzi Ismail
- Advanced Membrane Technology Research Center (AMTEC), Universiti Teknologi Malaysia, Skudai 81310, Johor Bahru, Johor, Malaysia
| | - Seeram RamaKrishna
- Department of Mechanical Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Filippo Berto
- Department of Mechanical and Industrial Engineering, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| |
Collapse
|
30
|
Datta S, Jana S, Das A, Chakraborty A, Chowdhury AR, Datta P. Bioprinting of radiopaque constructs for tissue engineering and understanding degradation behavior by use of Micro-CT. Bioact Mater 2020; 5:569-576. [PMID: 32373763 PMCID: PMC7195521 DOI: 10.1016/j.bioactmat.2020.04.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 04/05/2020] [Accepted: 04/22/2020] [Indexed: 12/17/2022] Open
Abstract
Bioprinting has emerged as a potential technique to fabricate tissue engineering constructs and in vitro models directly using living cells as a raw material for fabrication, conforming to the heterogeneity and architectural complexity of the tissues. In several of tissue engineering and in vitro disease modelling or surgical planning applications, it is desirable to have radiopaque constructs for monitoring and evaluation. In the present work, enhanced radiopaque constructs are generated by substituting Calcium ions with Barium ions for crosslinking of alginate hydrogels. The constructs are characterized for their structural integrity and followed by cell culture studies to evaluate their biocompatibility. This was followed by the radiopacity evaluation. The radiological images obtained by micro-CT technique was further applied to investigate the degradation behavior of the scaffolds. In conclusion, it is observed that barium crosslinking can provide a convenient means to obtain radiopaque constructs with potential for multi-faceted applications.
Collapse
Affiliation(s)
- Sudipto Datta
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, 711103, WB, India
| | - Shuvodeep Jana
- Indian Institute of Technology, Kharagpur, West Bengal, India
| | - Ankita Das
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, 711103, WB, India
| | - Arindam Chakraborty
- Department of Aerospace Engineering and Applied Mechanics, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, 711103, WB, India
| | - Amit Roy Chowdhury
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, 711103, WB, India
- Department of Aerospace Engineering and Applied Mechanics, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, 711103, WB, India
| | - Pallab Datta
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, 711103, WB, India
| |
Collapse
|
31
|
Ayan B, Wu Y, Karuppagounder V, Kamal F, Ozbolat IT. Aspiration-assisted bioprinting of the osteochondral interface. Sci Rep 2020; 10:13148. [PMID: 32753630 PMCID: PMC7403300 DOI: 10.1038/s41598-020-69960-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/22/2020] [Indexed: 11/09/2022] Open
Abstract
Osteochondral defects contain damage to both the articular cartilage and underlying subchon- dral bone, which remains a significant challenge in orthopedic surgery. Layered structure of bone, cartilage and the bone-cartilage interface must be taken into account in the case of biofabrication of the osteochondral (OC) interface. In this study, a dual layered OC interface was bioprinted using a newly developed aspiration-assisted bioprinting (AAB) technique, which has been the first time that scaffold-free bioprinting was applied to OC interface engineering. Tissue spheroids, made of human adipose-derived stem cells (ADSCs), were differentiated in three dimensions (3D) into chondrogenic and osteogenic spheroids, which were confirmed by immunostaining and histology qualitatively, and biochemistry assays and gene expression, quantitatively. Remarkably, the OC interface was bioprinted by accurate positioning of a layer of osteogenic spheroids onto a sacrificial alginate support followed by another layer of chondrogenic spheroids overlaid by the same support. Spheroids in individual zones fused and the maintenance of phenotypes in both zones confirmed the successful biofabrication of the histomorphologically-relevant OC interface. The biofabrication of OC tissue model without the use of polymeric scaffolds unveils great potential not only in regenerative medicine but also in drug testing and disease modeling for osteoarthritis.
Collapse
Affiliation(s)
- Bugra Ayan
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
| | - Yang Wu
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Vengadeshprabhu Karuppagounder
- Center for Orthopedic Research and Translational Sciences, Department of Orthopedics and Re-Habilitation, Penn State University, Hershey, PA, 17033, USA
| | - Fadia Kamal
- Center for Orthopedic Research and Translational Sciences, Department of Orthopedics and Re-Habilitation, Penn State University, Hershey, PA, 17033, USA
| | - Ibrahim T Ozbolat
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA.
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA.
- Biomedical Engineering Department, Penn State University, University Park, PA, 16802, USA.
- Materials Research Institute, Penn State University, University Park, PA, 16802, USA.
- Department of Neurosurgery, Penn State University, Hershey, PA, 17033, USA.
| |
Collapse
|
32
|
Elemoso A, Shalunov G, Balakhovsky YM, Ostrovskiy AY, Khesuani YD. 3D Bioprinting: The Roller Coaster Ride to Commercialization. Int J Bioprint 2020; 6:301. [PMID: 33088989 PMCID: PMC7557343 DOI: 10.18063/ijb.v6i3.301] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 06/23/2020] [Indexed: 01/30/2023] Open
Abstract
Three-dimensional (3D) bioprinting as a technology is being researched and applied since 2003. It is actually several technologies (inkjet, extrusion, laser, magnetic bioprinting, etc.) under an umbrella term "3D bioprinting." The versatility of this technology allows widespread applications in several; however, after almost 20 years of research, there is still a limited number of cases of commercialized applications. This article discusses the potential for 3D bioprinting in regenerative medicine, drug discovery, and food industry, as well as the existing cases of companies that create commercialized products and services in the aforementioned areas and even in fashion, including their go-to-market route and financing received. We also address the main barriers to creating practical applications of 3D bioprinting within each sphere the technology that is being studied for.
Collapse
Affiliation(s)
- Anton Elemoso
- Laboratory of Biotechnical Research 3D Bioprinting Solutions, Moscow, Russian Federation
| | - Grigoriy Shalunov
- Laboratory of Biotechnical Research 3D Bioprinting Solutions, Moscow, Russian Federation
| | | | - Alexander Yu Ostrovskiy
- Vivax Bio, LLC, New York, NY, USA.,Independent Laboratory IN VITRO, Moscow, Russian Federation
| | - Yusef D Khesuani
- Laboratory of Biotechnical Research 3D Bioprinting Solutions, Moscow, Russian Federation.,Vivax Bio, LLC, New York, NY, USA
| |
Collapse
|
33
|
Ramzy GM, Koessler T, Ducrey E, McKee T, Ris F, Buchs N, Rubbia-Brandt L, Dietrich PY, Nowak-Sliwinska P. Patient-Derived In Vitro Models for Drug Discovery in Colorectal Carcinoma. Cancers (Basel) 2020; 12:cancers12061423. [PMID: 32486365 PMCID: PMC7352800 DOI: 10.3390/cancers12061423] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 02/07/2023] Open
Abstract
Lack of relevant preclinical models that reliably recapitulate the complexity and heterogeneity of human cancer has slowed down the development and approval of new anti-cancer therapies. Even though two-dimensional in vitro culture models remain widely used, they allow only partial cell-to-cell and cell-to-matrix interactions and therefore do not represent the complex nature of the tumor microenvironment. Therefore, better models reflecting intra-tumor heterogeneity need to be incorporated in the drug screening process to more reliably predict the efficacy of drug candidates. Classic methods of modelling colorectal carcinoma (CRC), while useful for many applications, carry numerous limitations. In this review, we address the recent advances in in vitro CRC model systems, ranging from conventional CRC patient-derived models, such as conditional reprogramming-based cell cultures, to more experimental and state-of-the-art models, such as cancer-on-chip platforms or liquid biopsy.
Collapse
Affiliation(s)
- George M. Ramzy
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland; (G.M.R.); (E.D.)
- Translational Research Center in Oncohaematology, University of Geneva, 1211 Geneva, Switzerland
| | - Thibaud Koessler
- Department of Oncology, Geneva University Hospitals, 1211 Geneva, Switzerland; (T.K.); (P.-Y.D.)
| | - Eloise Ducrey
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland; (G.M.R.); (E.D.)
- Translational Research Center in Oncohaematology, University of Geneva, 1211 Geneva, Switzerland
| | - Thomas McKee
- Division of Clinical Pathology, Diagnostic Department, University Hospitals of Geneva (HUG), 1211 Geneva, Switzerland; (T.M.); (L.R.-B.)
| | - Frédéric Ris
- Translational Department of Digestive and Transplant Surgery, Faculty of Medicine, Geneva University Hospitals, 1211 Geneva, Switzerland; (F.R.); (N.B.)
| | - Nicolas Buchs
- Translational Department of Digestive and Transplant Surgery, Faculty of Medicine, Geneva University Hospitals, 1211 Geneva, Switzerland; (F.R.); (N.B.)
| | - Laura Rubbia-Brandt
- Division of Clinical Pathology, Diagnostic Department, University Hospitals of Geneva (HUG), 1211 Geneva, Switzerland; (T.M.); (L.R.-B.)
| | - Pierre-Yves Dietrich
- Department of Oncology, Geneva University Hospitals, 1211 Geneva, Switzerland; (T.K.); (P.-Y.D.)
| | - Patrycja Nowak-Sliwinska
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland; (G.M.R.); (E.D.)
- Translational Research Center in Oncohaematology, University of Geneva, 1211 Geneva, Switzerland
- Correspondence: ; Tel.: +41-22-379-3352
| |
Collapse
|
34
|
Chameettachal S, Yeleswarapu S, Sasikumar S, Shukla P, Hibare P, Bera AK, Bojedla SSR, Pati F. 3D Bioprinting: Recent Trends and Challenges. J Indian Inst Sci 2019. [DOI: 10.1007/s41745-019-00113-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
35
|
Chang EA, Jin SW, Nam MH, Kim SD. Human Induced Pluripotent Stem Cells : Clinical Significance and Applications in Neurologic Diseases. J Korean Neurosurg Soc 2019; 62:493-501. [PMID: 31392877 PMCID: PMC6732359 DOI: 10.3340/jkns.2018.0222] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 04/22/2019] [Indexed: 02/07/2023] Open
Abstract
The generation of human induced pluripotent stem cells (iPSCs) from somatic cells using gene transfer opens new areas for precision medicine with personalized cell therapy and encourages the discovery of essential platforms for targeted drug development. iPSCs retain the genome of the donor, may regenerate indefinitely, and undergo differentiation into virtually any cell type of interest using a range of published protocols. There has been enormous interest among researchers regarding the application of iPSC technology to regenerative medicine and human disease modeling, in particular, modeling of neurologic diseases using patient-specific iPSCs. For instance, Parkinson’s disease, Alzheimer’s disease, and spinal cord injuries may be treated with iPSC therapy or replacement tissues obtained from iPSCs. In this review, we discuss the work so far on generation and characterization of iPSCs and focus on recent advances in the use of human iPSCs in clinical setting.
Collapse
Affiliation(s)
- Eun-Ah Chang
- Department of Laboratory Medicine, Korea University Ansan Hospital, Ansan, Korea
| | - Sung-Won Jin
- Department of Neurosurgery, Korea University Ansan Hospital, Ansan, Korea
| | - Myung-Hyun Nam
- Department of Laboratory Medicine, Korea University Ansan Hospital, Ansan, Korea
| | - Sang-Dae Kim
- Department of Neurosurgery, Korea University Ansan Hospital, Ansan, Korea
| |
Collapse
|