1
|
Rea M, Lisa LD, Pagnotta G, Gallo N, Salvatore L, D’Amico F, Campilio N, Baena JM, Marchal JA, Cicero AF, Borghi C, Focarete ML. Establishing a Bioink Assessment Protocol: GelMA and Collagen in the Bioprinting of a Potential In Vitro Intestinal Model. ACS Biomater Sci Eng 2025; 11:2456-2467. [PMID: 40131228 PMCID: PMC12001187 DOI: 10.1021/acsbiomaterials.5c00034] [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: 01/05/2025] [Revised: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 03/26/2025]
Abstract
Collagen and gelatin methacryloyl (GelMA) are widely studied biomaterials for extrusion-based bioprinting (EBB) due to their excellent biological properties and ability to mimic the extracellular matrix of native tissues. This study aims to establish a preliminary workflow for approaching EBB by assessing collagen and GelMA printability and biological performance. GelMA was selected for its cost-effectiveness and ease of synthesis, while our collagen formulation was specifically optimized for printability, which is a challenging aspect of bioprinting. A parallel evaluation of their printability and biological performance is provided to develop a preliminary 3D intestinal model replicating the submucosa, lamina propria, and epithelial layer. Rheological analyses demonstrated that both materials exhibit a shear-thinning behavior. Collagen (u-CI) displayed a shear-thinning parameter p = 0.1 and a consistency index C = 80.62 Pa·s, while GelMA (u-GI) exhibited a more pronounced shear-thinning effect and enhanced shape retention (p = 0.06, C = 286.6 Pa·s). Post-extrusion recovery was higher for collagen (85%), compared to GelMA (45%), indicating its greater mechanical resilience. Photo-crosslinking improved hydrogel stability, with an increase in storage modulus G' for both materials. Printing tests confirmed the suitability of both hydrogels for bioprinting, with GelMA demonstrating higher print fidelity than collagen. Dimensional stability assessments under incubating conditions revealed that collagen constructs maintained their shape for 14 days before degradation, whereas GelMA constructs exhibited a gradual decrease in diameter over 21 days. Cell culture studies showed that human skin fibroblasts (HSFs) and human colon adenocarcinoma cells (HCT-8) could be successfully cocultured in an optimized RPMI 1640-based medium. AlamarBlue assays and Live/Dead staining confirmed high cell viability and proliferation within both hydrogel matrices. Notably, HSFs in GelMA exhibited more elongated morphologies, likely due to the material's lower stiffness (380 Pa) compared to collagen (585 Pa). HCT-8 cells adhered more rapidly to GelMA constructs, forming colonies within 7 days, whereas on collagen, colony formation was delayed to 14 days. Finally, a layered intestinal model was fabricated, and immunostaining confirmed the expression of tight junction (ZO-1) and adhesion (E-cadherin) proteins, validating the epithelial monolayer integrity. These findings highlight the potential of collagen and GelMA in 3D bioprinting applications for gut tissue engineering and pave the way for future developments of in vitro intestinal models.
Collapse
Grants
- National Recovery and Resilience Plan (NRRP), Mission 04 Component 2 Investment 1.5 â NextGenerationEU, Call for tender n. 3277 dated 30/12/2021,
- European Union - NextGenerationEU through the Italian Ministry of University and Research under PNRR âMission 4 Component 2, Investment 3.3 ââPartnerships extended to universities, research centers, companies and funding of basic research projectsââ D.M. 352/2021 â CUP J33C22001330009
- ConsejerÃa de EconomÃa, Conocimiento, Empresas y Universidad de la Junta de AndalucÃa (FEDER Funds, Projects B-CTS-230-UGR18, A-CTS-180-UGR20 and PYC20 RE 015 UGR)
- Chair ''Doctors Galera-Requena in cancer stem cell research'' (CMC-CTS963)
Collapse
Affiliation(s)
- Mariangela Rea
- Department
of Chemistry ‘Giacomo Ciamician’ and INSTM UdR of Bologna, University of Bologna, 40129 Bologna, Italy
| | - Luana Di Lisa
- Department
of Chemistry ‘Giacomo Ciamician’ and INSTM UdR of Bologna, University of Bologna, 40129 Bologna, Italy
| | - Giorgia Pagnotta
- Department
of Chemistry ‘Giacomo Ciamician’ and INSTM UdR of Bologna, University of Bologna, 40129 Bologna, Italy
| | - Nunzia Gallo
- Department
of Engineering for Innovation, University
of Salento, 73100 Lecce, Italy
- Typeone
Biomaterials S.r.l., Via Europa 167, 73021 Calimera, Lecce, Italy
| | - Luca Salvatore
- Typeone
Biomaterials S.r.l., Via Europa 167, 73021 Calimera, Lecce, Italy
| | - Federica D’Amico
- Department
of Pharmacy and Biotechnology, University
of Bologna, 40126 Bologna, Italy
| | | | - José Manuel Baena
- REGEMAT
3D S.L., 18016 Granada, Spain
- BRECA
Health Care S.L., 18016 Granada, Spain
- Biofabrication
group, Department of Pharmacy, School of Health Sciences, Universidad
Cardenal Herrera-CEU, CEU Universities, 46115 Alfara
de Patriarca, Valencia, Spain
| | - Juan Antonio Marchal
- Department
of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
- BioFab
i3D Lab, Centre for Biomedical Research (CIBM), University of Granada, 18016 Granada, Spain
- Instituto
de Investigación Biosanitaria ibs.GRANADA, 18016 Granada, Spain
- Excellence Research Unit “Modeling
Nature” (MNat),
University of Granada, 18071 Granada, Spain
| | - Arrigo F.G. Cicero
- Medical
and Surgery Sciences Department, University
of Bologna, 40138 Bologna, Italy
- Cardiovascular
Medicine Unit, IRCCS AOU di Bologna, 40138 Bologna, Italy
| | - Claudio Borghi
- Medical
and Surgery Sciences Department, University
of Bologna, 40138 Bologna, Italy
- Cardiovascular
Medicine Unit, IRCCS AOU di Bologna, 40138 Bologna, Italy
| | - Maria Letizia Focarete
- Department
of Chemistry ‘Giacomo Ciamician’ and INSTM UdR of Bologna, University of Bologna, 40129 Bologna, Italy
- Interdepartmental
Center for Industrial Research in Health Sciences and Technologies, University of Bologna, Via Tolara di Sopra, 41/E, 40064 Ozzano Emilia, Bologna, Italy
| |
Collapse
|
2
|
Mei X, Uribe Estrada MF, Rizwan M, Lukin I, Sanchez Gonzalez B, Marin Canchola JG, Velarde Jarquín V, Salazar Parraguez X, Del Valle Rodríguez F, Garciamendez-Mijares CE, Lin Z, Guo J, Wang Z, Maharjan S, Orive G, Zhang YS. A bioprinted animal patient-derived breast cancer model for anti-cancer drug screening. Mater Today Bio 2025; 31:101449. [PMID: 39896287 PMCID: PMC11782996 DOI: 10.1016/j.mtbio.2025.101449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/23/2024] [Accepted: 01/03/2025] [Indexed: 02/04/2025] Open
Abstract
Animal models are commonly used for drug screening before clinical trials. However, developing these models is time-consuming, and the results obtained from these models may differ from clinical outcomes due to the differences between animals and humans. To this end, 3D bioprinting offers several advantages for drug screening, such as high reproducibility and improved throughput, in addition to the human cells that can be used to generate these models. Here, we report the development of an animal patient-derived in vitro breast cancer model for drug screening using digital light processing (DLP) bioprinting. These bioprinted models demonstrated good cytocompatibility and preserved phenotypes of the cells. DLP enabled rapid fabrication with blood vessel-like channels to replicate, to a good extent, the tumor microenvironment. Our findings suggested that the improved microenvironment, provided by vascular structures within the bioprinted models, played a crucial role in reducing the chemoresistance of drugs. In addition, the correlation of the in vitro and in vivo drug-screening results was preliminarily performed to evaluate the predictive feasibility of this bioprinted model, suggesting a potential strategy for the design of future drug-testing platforms.
Collapse
Affiliation(s)
- Xuan Mei
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge 02139, MA, USA
| | - Maria Fernanda Uribe Estrada
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge 02139, MA, USA
| | - Muhammad Rizwan
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge 02139, MA, USA
- Cancer Genetics & Epigenetics Research Group, Department of Biosciences, COMSATS University Islamabad, Park Road Islamabad 45550, Pakistan
| | - Izeia Lukin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge 02139, MA, USA
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz 01009, Spain
| | - Begoña Sanchez Gonzalez
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge 02139, MA, USA
| | - Jose Gerardo Marin Canchola
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge 02139, MA, USA
| | - Valeria Velarde Jarquín
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge 02139, MA, USA
| | - Ximena Salazar Parraguez
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge 02139, MA, USA
| | - Francisco Del Valle Rodríguez
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge 02139, MA, USA
| | - Carlos Ezio Garciamendez-Mijares
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge 02139, MA, USA
| | - Zeng Lin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge 02139, MA, USA
| | - Jie Guo
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge 02139, MA, USA
| | - Zhenwu Wang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge 02139, MA, USA
| | - Sushila Maharjan
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge 02139, MA, USA
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz 01009, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
- University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria-Gasteiz, 01007, Spain
- Singapore Eye Research Institute, Singapore 169856, Singapore
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge 02139, MA, USA
| |
Collapse
|
3
|
Song YE, Eckman N, Sen S, Jons CK, Saouaf OM, Appel EA. Highly Extensible Physically Crosslinked Hydrogels for High-Speed 3D Bioprinting. Adv Healthc Mater 2025; 14:e2404988. [PMID: 39955737 PMCID: PMC12004426 DOI: 10.1002/adhm.202404988] [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: 12/28/2024] [Revised: 02/04/2025] [Indexed: 02/17/2025]
Abstract
Hydrogels have emerged as promising materials for bioprinting and many other biomedical applications due to their high degree of biocompatibility and ability to support and/or modulate cell viability and function. Yet, many hydrogel bioinks have suffered from low efficiency due to limitations on accessible printing speeds, often limiting cell viability and/or the constructs which can be generated. In this study, a highly extensible bioink system created by modulating the rheology of physically crosslinked hydrogels comprising hydrophobically-modified cellulosics and either surfactants or cyclodextrins is reported. It is demonstrated that these hydrogels are highly shear-thinning with broadly tunable viscoelasticity and stress-relaxation through simple modulation of the composition. Rheological experiments demonstrate that increasing concentration of rheology-modifying additives yields hydrogel materials exhibiting extensional strain-to-break values up to 2000%, which is amongst the most extensible examples of physically crosslinked hydrogels of this type. The potential of these hydrogels for use as bioinks is demonstrated by evaluating the relationship between extensibility and printability, demonstrating that greater hydrogel extensibility enables faster print speeds and smaller print features. The findings suggest that optimizing hydrogel extensibility can enhance high-speed 3D bioprinting capabilities, reporting over 5000 fold enhancement in speed index compared to existing works reported for hydrogel-based bioinks in extrusion-based printing.
Collapse
Affiliation(s)
- Ye Eun Song
- Department of Materials Science & EngineeringStanford UniversityStanfordCA94305USA
| | - Noah Eckman
- Department of Chemical EngineeringStanford UniversityStanfordCA94305USA
| | - Samya Sen
- Department of Materials Science & EngineeringStanford UniversityStanfordCA94305USA
| | - Carolyn K. Jons
- Department of Materials Science & EngineeringStanford UniversityStanfordCA94305USA
| | - Olivia M. Saouaf
- Department of Materials Science & EngineeringStanford UniversityStanfordCA94305USA
| | - Eric A. Appel
- Department of Materials Science & EngineeringStanford UniversityStanfordCA94305USA
- Department of BioengineeringStanford UniversityStanfordCA94305USA
- ChEM‐H InstituteStanford UniversityStanfordCA94305USA
- Woods Institute for the EnvironmentStanford UniversityStanfordCA94305USA
- Department of Pediatrics–EndocrinologyStanford University School of MedicineStanfordCA94305USA
| |
Collapse
|
4
|
Bilginer-Kartal R, Çoban B, Yildirim-Semerci Ö, Arslan-Yildiz A. Recent Advances in Hydrogel-Based 3D Disease Modeling and Drug Screening Platforms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025. [PMID: 40095242 DOI: 10.1007/5584_2025_851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Three-dimensional (3D) disease modeling and drug screening systems have become important in tissue engineering, drug screening, and development. The newly developed systems support cell and extracellular matrix (ECM) interactions, which are necessary for the formation of the tissue or an accurate model of a disease. Hydrogels are favorable biomaterials due to their properties: biocompatibility, high swelling capacity, tunable viscosity, mechanical properties, and their ability to biomimic the structure and function of ECM. They have been used to model various diseases such as tumors, cancer diseases, neurodegenerative diseases, cardiac diseases, and cardiovascular diseases. Additive manufacturing approaches, such as 3D printing/bioprinting, stereolithography (SLA), selective laser sintering (SLS), and fused deposition modeling (FDM), enable the design of scaffolds with high precision; thus, increasing the accuracy of the disease models. In addition, the aforementioned methodologies improve the design of the hydrogel-based scaffolds, which resemble the complicated structure and intricate microenvironment of tissues or tumors, further advancing the development of therapeutic agents and strategies. Thus, 3D hydrogel-based disease models fabricated through additive manufacturing approaches provide an enhanced 3D microenvironment that empowers personalized medicine toward targeted therapeutics, in accordance with 3D drug screening platforms.
Collapse
Affiliation(s)
| | - Başak Çoban
- Department of Bioengineering, Izmir Institute of Technology (IZTECH), Izmir, Turkey
| | | | - Ahu Arslan-Yildiz
- Department of Bioengineering, Izmir Institute of Technology (IZTECH), Izmir, Turkey.
| |
Collapse
|
5
|
Burstow R, Andrés D, Jiménez N, Camarena F, Thanou M, Pouliopoulos AN. Acoustic holography in biomedical applications. Phys Med Biol 2025; 70:06TR01. [PMID: 39978080 DOI: 10.1088/1361-6560/adb89a] [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: 10/15/2024] [Accepted: 02/20/2025] [Indexed: 02/22/2025]
Abstract
Acoustic holography can be used to construct an arbitrary wavefront at a desired 2D plane or 3D volume by beam shaping an emitted field and is a relatively new technique in the field of biomedical applications. Acoustic holography was first theorized in 1985 following Gabor's work in creating optical holograms in the 1940s. Recent developments in 3D printing have led to an easier and faster way to manufacture monolithic acoustic holographic lenses that can be attached to single-element transducers. As ultrasound passes through the lens material, a phase shift is applied to the waves, causing an interference pattern at the 2D image plane or 3D volume, which forms the desired pressure field. This technology has many applications already in use and has become of increasing interest for the biomedical community, particularly for treating regions that are notoriously difficult to operate on, such as the brain. Acoustic holograms could provide a non-invasive, precise, and patient specific way to deliver drugs, induce hyperthermia, or create tissue cell patterns. However, there are still limitations in acoustic holography, such as the difficulties in creating 3D holograms and the passivity of monolithic lenses. This review aims to outline the biomedical applications of acoustic holograms reported to date and discuss their current limitations and the future work that is needed for them to reach their full potential in the biomedical community.
Collapse
Affiliation(s)
- Rachel Burstow
- Department of Surgical & Interventional Engineering, School of Biomedical Engineering Imaging Sciences, King's College London, London, United Kingdom
| | - Diana Andrés
- Instituto de Instrumentación para Imagen Molecular (I3M), CSIC-Universitat Politècnica de València, Valencia, Spain
| | - Noé Jiménez
- Instituto de Instrumentación para Imagen Molecular (I3M), CSIC-Universitat Politècnica de València, Valencia, Spain
| | - Francisco Camarena
- Instituto de Instrumentación para Imagen Molecular (I3M), CSIC-Universitat Politècnica de València, Valencia, Spain
| | - Maya Thanou
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Antonios N Pouliopoulos
- Department of Surgical & Interventional Engineering, School of Biomedical Engineering Imaging Sciences, King's College London, London, United Kingdom
| |
Collapse
|
6
|
Michelutti L, Tel A, Robiony M, Vinayahalingam S, Agosti E, Ius T, Gagliano C, Zeppieri M. The Properties and Applicability of Bioprinting in the Field of Maxillofacial Surgery. Bioengineering (Basel) 2025; 12:251. [PMID: 40150715 PMCID: PMC11939734 DOI: 10.3390/bioengineering12030251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
Perhaps the most innovative branch of medicine is represented by regenerative medicine. It deals with regenerating or replacing tissues damaged by disease or aging. The innovative frontier of this branch is represented by bioprinting. This technology aims to reconstruct tissues, organs, and anatomical structures, such as those in the head and neck region. This would mean revolutionizing therapeutic and surgical approaches in the management of multiple conditions in which a conspicuous amount of tissue is lost. The application of bioprinting for the reconstruction of anatomical areas removed due to the presence of malignancy would represent a revolutionary new step in personalized and precision medicine. This review aims to investigate recent advances in the use of biomaterials for the reconstruction of anatomical structures of the head-neck region, particularly those of the oral cavity. The characteristics and properties of each biomaterial currently available will be presented, as well as their potential applicability in the reconstruction of areas affected by neoplasia damaged after surgery. In addition, this study aims to examine the current limitations and challenges and to analyze the future prospects of this technology in maxillofacial surgery.
Collapse
Affiliation(s)
- Luca Michelutti
- Clinic of Maxillofacial Surgery, Head-Neck and NeuroScience Department, University Hospital of Udine, p.le S. Maria della Misericordia 15, 33100 Udine, Italy; (L.M.); (A.T.)
| | - Alessandro Tel
- Clinic of Maxillofacial Surgery, Head-Neck and NeuroScience Department, University Hospital of Udine, p.le S. Maria della Misericordia 15, 33100 Udine, Italy; (L.M.); (A.T.)
| | - Massimo Robiony
- Clinic of Maxillofacial Surgery, Head-Neck and NeuroScience Department, University Hospital of Udine, p.le S. Maria della Misericordia 15, 33100 Udine, Italy; (L.M.); (A.T.)
| | | | - Edoardo Agosti
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Piazza Spedali Civili 1, 25123 Brescia, Italy
| | - Tamara Ius
- Academic Neurosurgery, Department of Neurosciences, University of Padova, 35121 Padova, Italy
| | - Caterina Gagliano
- Department of Medicine and Surgery, University of Enna “Kore”, Piazza dell’Università, 94100 Enna, Italy
- Mediterranean Foundation “G.B. Morgagni”, 95125 Catania, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, 33100 Udine, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34100 Trieste, Italy
| |
Collapse
|
7
|
Sousa AC, Alvites R, Lopes B, Sousa P, Moreira A, Coelho A, Santos JD, Atayde L, Alves N, Maurício AC. Three-Dimensional Printing/Bioprinting and Cellular Therapies for Regenerative Medicine: Current Advances. J Funct Biomater 2025; 16:28. [PMID: 39852584 PMCID: PMC11765675 DOI: 10.3390/jfb16010028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/26/2025] Open
Abstract
The application of three-dimensional (3D) printing/bioprinting technologies and cell therapies has garnered significant attention due to their potential in the field of regenerative medicine. This paper aims to provide a comprehensive overview of 3D printing/bioprinting technology and cell therapies, highlighting their results in diverse medical applications, while also discussing the capabilities and limitations of their combined use. The synergistic combination of 3D printing and cellular therapies has been recognised as a promising and innovative approach, and it is expected that these technologies will progressively assume a crucial role in the treatment of various diseases and conditions in the foreseeable future. This review concludes with a forward-looking perspective on the future impact of these technologies, highlighting their potential to revolutionize regenerative medicine through enhanced tissue repair and organ replacement strategies.
Collapse
Affiliation(s)
- Ana Catarina Sousa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Rui Alvites
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
- Instituto Universitário de Ciências da Saúde (CESPU), Instituto Universitário de Ciências da Saúde (IUCS), Avenida Central de Gandra 1317, Gandra, 4585-116 Paredes, Portugal
| | - Bruna Lopes
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Patrícia Sousa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Alícia Moreira
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - André Coelho
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - José Domingos Santos
- REQUIMTE-LAQV, Departamento de Engenharia Metalúrgica e Materiais, Faculdade de Engenharia, UP, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal;
| | - Luís Atayde
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Nuno Alves
- Centre for Rapid and Sustainable Product Development (CDRSP), Polytechnic Institute of Leiria, Rua de Portugal—Zona Industrial, 2430-028 Marinha Grande, Portugal;
| | - Ana Colette Maurício
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| |
Collapse
|
8
|
Mathur V, Dsouza V, Srinivasan V, Vasanthan KS. Volumetric Additive Manufacturing for Cell Printing: Bridging Industry Adaptation and Regulatory Frontiers. ACS Biomater Sci Eng 2025; 11:156-181. [PMID: 39746181 PMCID: PMC11733917 DOI: 10.1021/acsbiomaterials.4c01837] [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: 10/03/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025]
Abstract
Volumetric additive manufacturing (VAM) is revolutionizing the field of cell printing by enabling the rapid creation of complex three-dimensional cellular structures that mimic natural tissues. This paper explores the advantages and limitations of various VAM techniques, such as holographic lithography, digital light processing, and volumetric projection, while addressing their suitability across diverse industrial applications. Despite the significant potential of VAM, challenges related to regulatory compliance and scalability persist, particularly in the context of bioprinted tissues. In India, the lack of clear regulatory guidelines and intellectual property protections poses additional hurdles for companies seeking to navigate the evolving landscape of bioprinting. This study emphasizes the importance of collaboration among industry stakeholders, regulatory agencies, and academic institutions to establish tailored frameworks that promote innovation while ensuring safety and efficacy. By bridging the gap between technological advancement and regulatory oversight, VAM can unlock new opportunities in regenerative medicine and tissue engineering, transforming patient care and therapeutic outcomes.
Collapse
Affiliation(s)
- Vidhi Mathur
- Manipal
Centre for Biotherapeutics Research, Manipal
Academy of Higher Education, Manipal, 576104 Karnataka, India
| | - Vinita Dsouza
- Manipal
Centre for Biotherapeutics Research, Manipal
Academy of Higher Education, Manipal, 576104 Karnataka, India
| | - Varadharajan Srinivasan
- Department
of Civil Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, 576104 Karnataka, India
| | - Kirthanashri S Vasanthan
- Manipal
Centre for Biotherapeutics Research, Manipal
Academy of Higher Education, Manipal, 576104 Karnataka, India
| |
Collapse
|
9
|
An JH, Kim HY. Scaffolds Bioink for Three-Dimensional (3D) Bioprinting. Food Sci Anim Resour 2025; 45:126-144. [PMID: 39840242 PMCID: PMC11743847 DOI: 10.5851/kosfa.2024.e120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 01/23/2025] Open
Abstract
Rapid population growth and a corresponding increase in the demand for animal-derived proteins have led to food supply challenges and the need for alternative and sustainable meat production methods. Therefore, this study explored the importance of cell engineering technology-based three-dimensional bioprinting and bioinks, which play key roles in cultured meat production. In cultured meat production, bioinks have a significant effect on cell growth, differentiation, and mechanical stability. Hence, in this study, the characteristics of animal-, plant-, and marine-based bioinks were compared and analyzed, and the impact of each bioink on cultured meat production was evaluated. In particular, animal-based bioinks have the potential to produce cultured meat that is similar to conventional meat and are considered the most suitable bioinks for commercialization. Although plant- and marine-based bioinks are ecofriendly and have fewer religious restrictions, they are limited in terms of mechanical stability and consumer acceptance. Therefore, further research is required to develop and apply optimal animal-based bioinks for commercialization of cultured meat, particularly to improve its mechanical compatibility.
Collapse
Affiliation(s)
- Jin-Hee An
- Department of Animal Resources Science, Kongju National University, Yesan 32439, Korea
| | - Hack-Youn Kim
- Department of Animal Resources Science, Kongju National University, Yesan 32439, Korea
- Resources Science Research Institute, Yesan 32439, Korea
| |
Collapse
|
10
|
Derman ID, Moses JC, Rivera T, Ozbolat IT. Understanding the cellular dynamics, engineering perspectives and translation prospects in bioprinting epithelial tissues. Bioact Mater 2025; 43:195-224. [PMID: 39386221 PMCID: PMC11462153 DOI: 10.1016/j.bioactmat.2024.09.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/04/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024] Open
Abstract
The epithelium is one of the important tissues in the body as it plays a crucial barrier role serving as a gateway into and out of the body. Most organs in the body contain an epithelial tissue component, where the tightly connected, organ-specific epithelial cells organize into cysts, invaginations, or tubules, thereby performing distinct to endocrine or exocrine secretory functions. Despite the significance of epithelium, engineering functional epithelium in vitro has remained a challenge due to it is special architecture, heterotypic composition of epithelial tissues, and most importantly, difficulty in attaining the apico-basal and planar polarity of epithelial cells. Bioprinting has brought a paradigm shift in fabricating such apico-basal polarized tissues. In this review, we provide an overview of epithelial tissues and provide insights on recapitulating their cellular arrangement and polarization to achieve epithelial function. We describe the different bioprinting techniques that have been successful in engineering polarized epithelium, which can serve as in vitro models for understanding homeostasis and studying diseased conditions. We also discuss the different attempts that have been investigated to study these 3D bioprinted engineered epithelium for preclinical use. Finally, we highlight the challenges and the opportunities that need to be addressed for translation of 3D bioprinted epithelial tissues towards paving way for personalized healthcare in the future.
Collapse
Affiliation(s)
- Irem Deniz Derman
- 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
| | - Joseph Christakiran Moses
- 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
| | - Taino Rivera
- Biomedical Engineering Department, Penn State University, University Park, PA, 16802, 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
- Cancer Institute, Penn State University, University Park, PA, 16802, USA
- Neurosurgery Department, Penn State University, University Park, PA, 16802, USA
- Department of Medical Oncology, Cukurova University, Adana, 01330, Turkey
| |
Collapse
|
11
|
Li H, Chen H, Du C, Liu Y, Wan L, Ai F, Zhou K. Effect of Hydroxyapatite Nanowires on Formation and Bioactivity of Osteoblastic Cell Spheroid. ACS Biomater Sci Eng 2024; 10:7413-7428. [PMID: 39403768 DOI: 10.1021/acsbiomaterials.4c01159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Compared with traditional high-density cell spheroids, which are more prone to core necrosis, nanowires effectively improve the biological activity of core cells in spheroids, emanating more innovations for optimizing the internal cell survival environment and providing differentiation signals. In this study, hydroxyapatite nanowires (HAW), which provide numerous material exchange channels for internal cells by interpenetrating into cell spheroids, were added to osteoblast precursor (MC3T3-E1) cell spheroids. HAW, synthesized using the hydrothermal method, was used as a regulatory material to prepare uniformly sized 3D composite spheroids with good biological activity. Subsequently, material characterization and biocompatibility tests were performed on HAW, and the biological activity and osteogenic differentiation ability of the cell spheroids were tested. Notably, in 2D coculture, HAW displayed a certain attraction to MC3T3-E1 cells and promoted cell aggregation toward it. The content of HAW determined whether composite cell spheroids can form aggregated spherical structures, and incorporation of HAW alleviated core necrosis and enhanced the osteogenic phenotype. In summary, these findings indicate that the prepared HAW-bone cell composite spheroids can potentially be used as building blocks for the construction of large high-density biomimetic tissues and organoids using 3D bioprinting technology.
Collapse
Affiliation(s)
- Hanjing Li
- School of Advanced Manufacturing, Nanchang University, Nanchang 330031, People's Republic of China
| | - Hongwei Chen
- School of Advanced Manufacturing, Nanchang University, Nanchang 330031, People's Republic of China
| | - Chunyuan Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Yucheng Liu
- School of Advanced Manufacturing, Nanchang University, Nanchang 330031, People's Republic of China
| | - Li Wan
- School of Advanced Manufacturing, Nanchang University, Nanchang 330031, People's Republic of China
| | - Fanrong Ai
- School of Advanced Manufacturing, Nanchang University, Nanchang 330031, People's Republic of China
- Nanchang Municipal Key Laboratory of 3D Bioprinting Technology and Equipment, Nanchang University, Nanchang 330031, People's Republic of China
| | - Kui Zhou
- School of Advanced Manufacturing, Nanchang University, Nanchang 330031, People's Republic of China
- Nanchang Municipal Key Laboratory of 3D Bioprinting Technology and Equipment, Nanchang University, Nanchang 330031, People's Republic of China
- State Key Lab of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China
| |
Collapse
|
12
|
Jamróz P, Świeży A, Noworyta M, Starzak K, Środa P, Wielgus W, Szymaszek P, Tyszka-Czochara M, Ortyl J. Photocurable biomaterials labeled with luminescent sensors dedicated to bioprinting. J Biotechnol 2024; 395:122-140. [PMID: 39349123 DOI: 10.1016/j.jbiotec.2024.09.017] [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: 06/22/2024] [Revised: 08/20/2024] [Accepted: 09/24/2024] [Indexed: 10/02/2024]
Abstract
In the present study, we focused on the development and characterization of formulations that function as biological inks. These inks were doped with coumarin derivatives to act as molecular luminescent sensors that allow the monitoring of the kinetics of in situ photopolymerization in 3D (DLP) printing and bioprinting using pneumatic extrusion techniques, making it possible to study the changes in the system in real time. The efficiency of the systems was tested on compositions containing monomers: poly(ethylene glycol) diacrylates and photoinitiators: 2,4,6-trimethylbenzoyldi-phenylphosphinate and lithium phenyl-2,4,6-trimethylbenzoylphosphinate. The selected formulations were spectroscopically characterized and examined for their photopolymerization kinetics and rheological properties. This is important because of the fact that spectroscopic characterization, examination of photopolymerization kinetics, and rheological properties provide valuable insights into the behaviour of photocurable resin dedicated for 3D printing processes. The next step involved printing tests on commercially available 3D printers. In turn, printing carried out as part of the work on commercially available 3D printers further verified the effectiveness of the formulations. Moreover the formulation components and the resulting 3D objects were tested for their antiproliferative effects on the selected Chinese hamster ovary cell line, CHO-K1.
Collapse
Affiliation(s)
- Paweł Jamróz
- Cracow University of Technology, Faculty of Chemical Engineering and Technology, Warszawska 24, Cracow 31-155, Poland
| | - Andrzej Świeży
- Cracow University of Technology, Faculty of Chemical Engineering and Technology, Warszawska 24, Cracow 31-155, Poland; Photo HiTech Ltd., Bobrzyńskiego 14, Cracow 30-348, Poland
| | - Małgorzata Noworyta
- Cracow University of Technology, Faculty of Chemical Engineering and Technology, Warszawska 24, Cracow 31-155, Poland
| | - Katarzyna Starzak
- Cracow University of Technology, Faculty of Chemical Engineering and Technology, Warszawska 24, Cracow 31-155, Poland
| | - Patrycja Środa
- Cracow University of Technology, Faculty of Chemical Engineering and Technology, Warszawska 24, Cracow 31-155, Poland; Photo HiTech Ltd., Bobrzyńskiego 14, Cracow 30-348, Poland
| | - Weronika Wielgus
- Cracow University of Technology, Faculty of Chemical Engineering and Technology, Warszawska 24, Cracow 31-155, Poland
| | - Patryk Szymaszek
- Cracow University of Technology, Faculty of Chemical Engineering and Technology, Warszawska 24, Cracow 31-155, Poland
| | | | - Joanna Ortyl
- Cracow University of Technology, Faculty of Chemical Engineering and Technology, Warszawska 24, Cracow 31-155, Poland; Photo HiTech Ltd., Bobrzyńskiego 14, Cracow 30-348, Poland; Photo4Chem Ltd., Lea 114, Cracow 30-133, Poland.
| |
Collapse
|
13
|
Son KH, Kim DH, Park S, Kim HJ, Park M, Kim SJ, Lee SJ, Ahn K, Lee JW. Spherical Shell Bioprinting to Produce Uniform Spheroids with Controlled Sizes. J Funct Biomater 2024; 15:350. [PMID: 39590553 PMCID: PMC11595458 DOI: 10.3390/jfb15110350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/14/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
Conventional cell spheroid production methods are largely manual, leading to variations in size and shape that compromise consistency and reliability for use in cell-based therapeutic applications. To enhance spheroid production, a spherical shell bioprinting system was implemented, enabling the high-throughput generation of uniform cell spheroids with precisely controlled sizes. The system encapsulates cells within thin alginate hydrogel shells formed through bioprinting and ion crosslinking reactions. Alginate-calcium ion crosslinking created alginate shells that contained gelatin-based bioinks with embedded cells, facilitating spontaneous cell aggregation within the shells and eliminating the need for plastic wells. By adjusting cell concentrations in the alginate-gelatin bioink, we achieved precise control over spheroid size, maintaining a sphericity above 0.94 and size deviations within ±10 µm. This method has been successfully applied to various cell types including cancer cells, fibroblasts, chondrocytes, and epithelial cells, demonstrating its versatility. This scalable approach enhances the reliability of cell therapy and drug screening, offering a robust platform for future biomedical applications.
Collapse
Affiliation(s)
- Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gil Medical Center, College of Medicine, Gachon University, 21, Namdong-daero 774 Beon-gil, Namdong-gu, Incheon 21565, Republic of Korea;
| | - Dong-Ha Kim
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Seunghye Park
- Department of Health Sciences and Technology, GAIHST, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Republic of Korea;
| | - Hyun Jae Kim
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Mira Park
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Seung-Jin Kim
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA;
| | - Keunsun Ahn
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Jin Woo Lee
- Department of Health Sciences and Technology, GAIHST, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Republic of Korea;
- Department of Molecular Medicine, College of Medicine, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Republic of Korea
| |
Collapse
|
14
|
Vecchiotti D, Di Vito Nolfi M, Veglianti F, Dall’Aglio F, Khan HN, Flati I, Verzella D, Capece D, Alesse E, Angelucci A, Zazzeroni F. A 3D Bioprinting Approach to Studying Retinal Müller Cells. Genes (Basel) 2024; 15:1414. [PMID: 39596614 PMCID: PMC11593586 DOI: 10.3390/genes15111414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Bioprinting is an innovative technology in tissue engineering, enabling the creation of complex biological structures. This study aims to develop a three-dimensional (3D) bioprinted model of Müller cells (MCs) to enhance our understanding of their physiological and pathological roles in the retina. Methods: We investigated two different hydrogels for their ability to support the viability and differentiation of rMC-1 cells, an immortalized retinal cell line. Using 3D bioprinting technology, we assessed cell viability, differentiation, and functional characteristics through various assays, including live/dead assays and western blot analysis. Results: The collagen-based hydrogel significantly improved the viability of rMC-1 cells and facilitated the formation of spheroid aggregates, more accurately mimicking in vivo conditions compared to traditional two-dimensional (2D) culture systems. Moreover, 3D bioprinted MCs exhibited reduced markers of gliosis and oxidative stress compared to 2D cultures. Molecular analysis revealed decreased expression of GFAP and phosphorylated ERK in the 3D setting, indicating a less stressed cellular phenotype. Conclusions: Our findings demonstrate that 3D bioprinting technologies provide a more predictive platform for studying the biology of retinal MCs, which can help in the development of targeted therapeutic strategies for retinal diseases.
Collapse
Affiliation(s)
- Davide Vecchiotti
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy
| | - Mauro Di Vito Nolfi
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy
| | - Francesca Veglianti
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy
| | - Francesca Dall’Aglio
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy
| | - Hafiz Nadeem Khan
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy
| | - Irene Flati
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Daniela Verzella
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy
| | - Daria Capece
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy
| | - Edoardo Alesse
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy
| | - Adriano Angelucci
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy
| | - Francesca Zazzeroni
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy
| |
Collapse
|
15
|
Teixeira MC, Lameirinhas NS, Carvalho JPF, Luís J, Oliveira H, Oliveira JM, Silvestre AJD, Vilela C, Freire CSR. Biobased hydrogel bioinks of pectin, nanocellulose and lysozyme nanofibrils for the bioprinting of A375 melanoma cell-laden 3D in vitro platforms. Int J Biol Macromol 2024; 282:136958. [PMID: 39490881 DOI: 10.1016/j.ijbiomac.2024.136958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/24/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Melanoma is one of the most aggressive types of skin cancer, and the need for advanced platforms to study this disease and to develop new treatments is rising. 3D bioprinted tumor models are emerging as advanced tools to tackle these needs, with the design of adequate bioinks being a fundamental step to address this challenging process. Thus, this work explores the synergy between two biobased nanofibers, nanofibrillated cellulose (NFC) and lysozyme amyloid nanofibrils (LNFs), to create pectin nanocomposite hydrogel bioinks for the 3D bioprinting of A375 melanoma cell-laden living constructs. The incorporation of LNFs (5, 10 or 15 wt%) on a pectin-NFC suspension originates inks with enhanced rheological properties (shear viscosity and yield point) and proper shear-thinning behavior. The crosslinked hydrogels mimic the stiffness of melanoma tissues, being stable under physiological and cell-culture conditions, and non-cytotoxic towards A375 melanoma cells. P-NFC-LNFs (10 %) reveals good printability (Pr = 0.89) and printing accuracy (51 ± 2 %), and when loaded with A375 cells (3 × 106 cells mL-1) the bioink originates 3D-constructs with high cell viability (92 ± 1 %) after 14 days. The potential of the constructs as 3D in vitro platforms is corroborated by a drug-screening test with doxorubicin, where cells within the model displayed high sensitivity to the drug.
Collapse
Affiliation(s)
- Maria C Teixeira
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Nicole S Lameirinhas
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - João P F Carvalho
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Jorge Luís
- CICECO - Aveiro Institute of Materials, EMaRT Group - Emerging: Materials, Research, Technology, School of Design, Management and Production Technologies Northern Aveiro, University of Aveiro, 3720-509 Oliveira de Azeméis, Portugal
| | - Helena Oliveira
- Department of Biology & CESAM, University of Aveiro, 3810-193 Aveiro, Portugal
| | - José Martinho Oliveira
- CICECO - Aveiro Institute of Materials, EMaRT Group - Emerging: Materials, Research, Technology, School of Design, Management and Production Technologies Northern Aveiro, University of Aveiro, 3720-509 Oliveira de Azeméis, Portugal
| | - Armando J D Silvestre
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Carla Vilela
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Carmen S R Freire
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|
16
|
Greco I, Machrafi H, Iorio CS. Double-Network Hydrogel 3D BioPrinting Biocompatible with Fibroblast Cells for Tissue Engineering Applications. Gels 2024; 10:684. [PMID: 39590040 PMCID: PMC11594167 DOI: 10.3390/gels10110684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
The present study examines the formulation of a biocompatible hydrogel bioink for 3D bioprinting, integrating poly(ethylene glycol) diacrylate (PEGDA) and sodium alginate (SA) using a double-network approach. These materials were chosen for their synergistic qualities, with PEGDA contributing to mechanical integrity and SA ensuring biocompatibility. Fibroblast cells were included in the bioink and printed with a Reg4Life bioprinter employing micro-extrusion technology. The optimisation of printing parameters included needle size and flow velocities. This led to precise structure development and yielded results with a negligible deviation in printed angles and better control of line widths. The rheological characteristics of the bioink were evaluated, demonstrating appropriate viscosity and shear-thinning behaviour for efficient extrusion. The mechanical characterisation revealed an average compressive modulus of 0.38 MPa, suitable for tissue engineering applications. The printability of the bioink was further confirmed through the evaluations of morphology and diffusion rates, confirming structural integrity. Biocompatibility assessments demonstrated a high cell viability rate of 82.65% following 48 h of incubation, supporting the bioink's suitability for facilitating cell survival. This study introduced a reliable technique for producing tissue-engineered scaffolds that exhibit outstanding mechanical characteristics and cell viability, highlighting the promise of PEGDA-SA hydrogels in bioprinting applications.
Collapse
Affiliation(s)
- Immacolata Greco
- Center for Research and Engineering in Space Technologies, Université Libre de Bruxelles, 1050 Brussels, Belgium; (I.G.); (C.S.I.)
| | - Hatim Machrafi
- Center for Research and Engineering in Space Technologies, Université Libre de Bruxelles, 1050 Brussels, Belgium; (I.G.); (C.S.I.)
- GIGA-In Silico Medicine, Université de Liège, 4000 Liège, Belgium
| | - Carlo S. Iorio
- Center for Research and Engineering in Space Technologies, Université Libre de Bruxelles, 1050 Brussels, Belgium; (I.G.); (C.S.I.)
| |
Collapse
|
17
|
Boretti G, Amirfallah A, Edmunds KJ, Hamzehpour H, Sigurjónsson ÓE. Advancing Cartilage Tissue Engineering: A Review of 3D Bioprinting Approaches and Bioink Properties. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 39381849 DOI: 10.1089/ten.teb.2024.0168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Articular cartilage is crucial in human physiology, and its degeneration poses a significant public health challenge. While recent advancements in 3D bioprinting and tissue engineering show promise for cartilage regeneration, there remains a gap between research findings and clinical application. This review critically examines the mechanical and biological properties of hyaline cartilage, along with current 3D manufacturing methods and analysis techniques. Moreover, we provide a quantitative synthesis of bioink properties used in cartilage tissue engineering. After screening 181 initial works, 33 studies using extrusion bioprinting were analyzed and synthesized, presenting results that indicate the main materials, cells, and methods utilized for mechanical and biological evaluation. Altogether, this review motivates the standardization of mechanical analyses and biomaterial assessments of 3D bioprinted constructs to clarify their chondrogenic potential.
Collapse
Affiliation(s)
- Gabriele Boretti
- School of Science and Engineering, Reykjavík University, Reykjavík, Iceland
| | - Arsalan Amirfallah
- The Blood Bank, Landspitali, The National University Hospital of Iceland, Reykjavík, Iceland
| | - Kyle J Edmunds
- School of Science and Engineering, Reykjavík University, Reykjavík, Iceland
| | - Helena Hamzehpour
- Faculty of Pharmaceutical Sciences, University of Iceland, Reykjavik, Iceland
| | - Ólafur E Sigurjónsson
- School of Science and Engineering, Reykjavík University, Reykjavík, Iceland
- The Blood Bank, Landspitali, The National University Hospital of Iceland, Reykjavík, Iceland
| |
Collapse
|
18
|
Taylor S, Mueller E, Jones LR, Makela AV, Ashammakhi N. Translational Aspects of 3D and 4D Printing and Bioprinting. Adv Healthc Mater 2024; 13:e2400463. [PMID: 38979857 DOI: 10.1002/adhm.202400463] [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: 02/06/2024] [Revised: 05/22/2024] [Indexed: 07/10/2024]
Abstract
Three-dimensional (3D) printed medical devices include orthopedic and craniofacial implants, surgical tools, and external prosthetics that have been directly used in patients. While the advances of additive manufacturing techniques in the production of medical devices have been on the rise, clinical translation of living cellular constructs face significant limitations in terms of regulatory affairs, process technology, and materials development. In this perspective, the current status-quo of 3D and four-dimensional (4D) (bio)printing is summarized, current advancements are discussed and the challenges that need to be addressed for improved industrial translation and clinical applications of bioprinting are highlighted. It is focused on a multidisciplinary approach in discussing the key translational considerations, from the perspective of industry, regulatory bodies, funding strategies, and future directions.
Collapse
Affiliation(s)
| | - Eva Mueller
- Ricoh 3D for Healthcare, Ricoh USA, Winston-Salem, NC 27101, USA
| | - Lamont R Jones
- Department of Otolaryngology, Henry Ford Heath, Detroit, MI 48322, USA
| | - Ashley V Makela
- Institute for Quantitative Health Science & Engineering and Department of Engineering, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science & Engineering and Department of Engineering, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
19
|
Mihaylova A, Shopova D, Parahuleva N, Yaneva A, Bakova D. (3D) Bioprinting-Next Dimension of the Pharmaceutical Sector. Pharmaceuticals (Basel) 2024; 17:797. [PMID: 38931464 PMCID: PMC11206453 DOI: 10.3390/ph17060797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 05/26/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
To create a review of the published scientific literature on the benefits and potential perspectives of the use of 3D bio-nitrification in the field of pharmaceutics. This work was performed in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines for reporting meta-analyses and systematic reviews. The scientific databases PubMed, Scopus, Google Scholar, and ScienceDirect were used to search and extract data using the following keywords: 3D bioprinting, drug research and development, personalized medicine, pharmaceutical companies, clinical trials, drug testing. The data points to several aspects of the application of bioprinting in pharmaceutics were reviewed. The main applications of bioprinting are in the development of new drug molecules as well as in the preparation of personalized drugs, but the greatest benefits are in terms of drug screening and testing. Growth in the field of 3D printing has facilitated pharmaceutical applications, enabling the development of personalized drug screening and drug delivery systems for individual patients. Bioprinting presents the opportunity to print drugs on demand according to the individual needs of the patient, making the shape, structure, and dosage suitable for each of the patient's physical conditions, i.e., print specific drugs for controlled release rates; print porous tablets to reduce swallowing difficulties; make transdermal microneedle patches to reduce patient pain; and so on. On the other hand, bioprinting can precisely control the distribution of cells and biomaterials to build organoids, or an Organ-on-a-Chip, for the testing of drugs on printed organs mimicking specified disease characteristics instead of animal testing and clinical trials. The development of bioprinting has the potential to offer customized drug screening platforms and drug delivery systems meeting a range of individualized needs, as well as prospects at different stages of drug development and patient therapy. The role of bioprinting in preclinical and clinical testing of drugs is also of significant importance in terms of shortening the time to launch a medicinal product on the market.
Collapse
Affiliation(s)
- Anna Mihaylova
- Department of Healthcare Management, Faculty of Public Health, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
| | - Dobromira Shopova
- Department of Prosthetic Dentistry, Faculty of Dental Medicine, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
| | - Nikoleta Parahuleva
- Department of Obstetrics and Gynecology, Faculty of Medicine, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
| | - Antoniya Yaneva
- Department of Medical Informatics, Biostatistics and eLearning, Faculty of Public Health, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
| | - Desislava Bakova
- Department of Healthcare Management, Faculty of Public Health, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
| |
Collapse
|
20
|
Khorsandi D, Rezayat D, Sezen S, Ferrao R, Khosravi A, Zarepour A, Khorsandi M, Hashemian M, Iravani S, Zarrabi A. Application of 3D, 4D, 5D, and 6D bioprinting in cancer research: what does the future look like? J Mater Chem B 2024; 12:4584-4612. [PMID: 38686396 DOI: 10.1039/d4tb00310a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
The application of three- and four-dimensional (3D/4D) printing in cancer research represents a significant advancement in understanding and addressing the complexities of cancer biology. 3D/4D materials provide more physiologically relevant environments compared to traditional two-dimensional models, allowing for a more accurate representation of the tumor microenvironment that enables researchers to study tumor progression, drug responses, and interactions with surrounding tissues under conditions similar to in vivo conditions. The dynamic nature of 4D materials introduces the element of time, allowing for the observation of temporal changes in cancer behavior and response to therapeutic interventions. The use of 3D/4D printing in cancer research holds great promise for advancing our understanding of the disease and improving the translation of preclinical findings to clinical applications. Accordingly, this review aims to briefly discuss 3D and 4D printing and their advantages and limitations in the field of cancer. Moreover, new techniques such as 5D/6D printing and artificial intelligence (AI) are also introduced as methods that could be used to overcome the limitations of 3D/4D printing and opened promising ways for the fast and precise diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Danial Khorsandi
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90024, USA
| | - Dorsa Rezayat
- Center for Global Design and Manufacturing, College of Engineering and Applied Science, University of Cincinnati, 2901 Woodside Drive, Cincinnati, OH 45221, USA
| | - Serap Sezen
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla 34956 Istanbul, Türkiye
- Nanotechnology Research and Application Center, Sabanci University, Tuzla 34956 Istanbul, Türkiye
| | - Rafaela Ferrao
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90024, USA
- University of Coimbra, Institute for Interdisciplinary Research, Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), Portugal
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul 34959, Türkiye
| | - Atefeh Zarepour
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai - 600 077, India
| | - Melika Khorsandi
- Department of Cellular and Molecular Biology, Najafabad Branch, Islamic Azad University, Isfahan, Iran
| | - Mohammad Hashemian
- Department of Cellular and Molecular Biology, Najafabad Branch, Islamic Azad University, Isfahan, Iran
| | - Siavash Iravani
- Independent Researcher, W Nazar ST, Boostan Ave, Isfahan, Iran.
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Türkiye.
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan
| |
Collapse
|
21
|
Zoco de la Fuente A, García-García A, Pérez-Álvarez L, Moreno-Benítez I, Larrea-Sebal A, Martin C, Vilas-Vilela JL. Evaluation of Various Types of Alginate Inks for Light-Mediated Extrusion 3D Printing. Polymers (Basel) 2024; 16:986. [PMID: 38611244 PMCID: PMC11014002 DOI: 10.3390/polym16070986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/25/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024] Open
Abstract
Naturally derived biopolymers modifying or combining with other components are excellent candidates to promote the full potential of additive manufacturing in biomedicine, cosmetics, and the food industry. This work aims to develop new photo-cross-linkable alginate-based inks for extrusion 3D printing. Specifically, this work is focused on the effect of the addition of cross-linkers with different chemical structures (polyethylene glycol diacrylate (PEGDA), N,N'-methylenebisacrylamide (NMBA), and acrylic acid (AA)) in the potential printability and physical properties of methacrylated alginate (AlgMe) hydrogels. Although all inks showed maximum photo-curing conversions and gelation times less than 2 min, only those structures printed with the inks incorporating cross-linking agents with flexible and long chain structure (PEGDA and AA) displayed acceptable size accuracy (~0.4-0.5) and printing index (Pr ~1.00). The addition of these cross-linking agents leads to higher Young's moduli (from 1.6 to 2.0-2.6 KPa) in the hydrogels, and their different chemical structures results in variations in their mechanical and rheological properties. However, similar swelling ability (~15 swelling factor), degradability (~45 days 100% weight loss), and cytocompatibility (~100%) were assessed in all the systems, which is of great importance for the final applicability of these hydrogels.
Collapse
Affiliation(s)
- Aitana Zoco de la Fuente
- Macromolecular Chemistry Group (LABQUIMAC), Physical Chemistry Department, Faculty of Science and Technology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (A.Z.d.l.F.); (A.G.-G.); (J.L.V.-V.)
| | - Ane García-García
- Macromolecular Chemistry Group (LABQUIMAC), Physical Chemistry Department, Faculty of Science and Technology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (A.Z.d.l.F.); (A.G.-G.); (J.L.V.-V.)
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain
| | - Leyre Pérez-Álvarez
- Macromolecular Chemistry Group (LABQUIMAC), Physical Chemistry Department, Faculty of Science and Technology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (A.Z.d.l.F.); (A.G.-G.); (J.L.V.-V.)
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain
| | - Isabel Moreno-Benítez
- Macromolecular Chemistry Group (LABQUIMAC), Organic Chemistry Department, Faculty of Science and Technology, University of the Basque CountryUPV/EHU, 48940 Leioa, Spain;
| | - Asier Larrea-Sebal
- Biofisika Institute (UPV/EHU, CSIC), UPV/EHU Science Park, 48940 Leioa, Spain; (A.L.-S.); (C.M.)
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
- Fundación Biofisika Bizkaia, Barrio Sarriena s/n, 48940 Leioa, Spain
| | - Cesar Martin
- Biofisika Institute (UPV/EHU, CSIC), UPV/EHU Science Park, 48940 Leioa, Spain; (A.L.-S.); (C.M.)
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
| | - Jose Luis Vilas-Vilela
- Macromolecular Chemistry Group (LABQUIMAC), Physical Chemistry Department, Faculty of Science and Technology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (A.Z.d.l.F.); (A.G.-G.); (J.L.V.-V.)
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain
| |
Collapse
|
22
|
Teng F, Wang W, Wang ZQ, Wang GX. Analysis of bioprinting strategies for skin diseases and injuries through structural and temporal dynamics: historical perspectives, research hotspots, and emerging trends. Biofabrication 2024; 16:025019. [PMID: 38350130 DOI: 10.1088/1758-5090/ad28f0] [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: 10/29/2023] [Accepted: 02/13/2024] [Indexed: 02/15/2024]
Abstract
This study endeavors to investigate the progression, research focal points, and budding trends in the realm of skin bioprinting over the past decade from a structural and temporal dynamics standpoint. Scholarly articles on skin bioprinting were obtained from WoSCC. A series of bibliometric tools comprising R software, CiteSpace, HistCite, and an alluvial generator were employed to discern historical characteristics, evolution of active topics, and upcoming tendencies in the area of skin bioprinting. Over the past decade, there has been a consistent rise in research interest in skin bioprinting, accompanied by an extensive array of meaningful scientific collaborations. Concurrently, diverse dynamic topics have emerged during various periods, as substantiated by an aggregate of 22 disciplines, 74 keywords, and 187 references demonstrating citation bursts. Four burgeoning research subfields were discerned through keyword clustering-namely, #3 'in situbioprinting', #6 'vascular', #7 'xanthan gum', and #8 'collagen hydrogels'. The keyword alluvial map reveals that Module 1, including 'transplantation' etc, has primarily dominated the research module over the previous decade, maintaining enduring relevance despite annual shifts in keyword focus. Additionally, we mapped out the top six key modules from 2023 being 'silk fibroin nanofiber', 'system', 'ionic liquid', 'mechanism', and 'foot ulcer'. Three recent research subdivisions were identified via timeline visualization of references, particularly Clusters #0 'wound healing', #4 'situ mineralization', and #5 '3D bioprinter'. Insights derived from bibliometric analyses illustrate present conditions and trends in skin bioprinting research, potentially aiding researchers in pinpointing central themes and pioneering novel investigative approaches in this field.
Collapse
Affiliation(s)
- Fei Teng
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, People's Republic of China
| | - Wei Wang
- Department of Ultrasound, University-Town Hospital of Chongqing Medical University, Chongqing 400042, People's Republic of China
| | - Zhi-Qiang Wang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, People's Republic of China
| | - Gui-Xue Wang
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Modern Life Science Experiment Teaching Center at Bioengineering College of Chongqing University, Chongqing 400030, People's Republic of China
| |
Collapse
|
23
|
Lv X, Zhang C, Liu X, Li P, Yang Y. 3D bioprinting technology to construct bone reconstruction research model and its feasibility evaluation. Front Bioeng Biotechnol 2024; 12:1328078. [PMID: 38314351 PMCID: PMC10834755 DOI: 10.3389/fbioe.2024.1328078] [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: 10/26/2023] [Accepted: 01/09/2024] [Indexed: 02/06/2024] Open
Abstract
Objective: To explore and construct a 3D bone remodeling research model displaying stability, repeatability, and precise simulation of the physiological and biochemical environment in vivo. Methods: In this study, 3D bioprinting was used to construct a bone reconstruction model. Sodium alginate (SA), hydroxyapatite (HA) and gelatin (Gel) were mixed into hydrogel as scaffold material. The osteoblast precursor cells MC3T3-E1 and osteoclast precursor cells RAW264.7 were used as seed cells, which may or may not be separated by polycarbonate membrane. The cytokines osteoprotegerin (OPG) and receptor activator of NF-κB ligand (RANKL) were used to induce cell differentiation. The function of scaffolds in the process of bone remodeling was analyzed by detecting the related markers of osteoblasts (alkaline phosphatase, ALP) and osteoclasts (tartrate resistant acid phosphatase, TRAP). Results: The scaffold showed good biocompatibility and low toxicity. The surface morphology aided cell adhesion and growth. The scaffold had optimum degradability, water absorption capacity and porosity, which are in line with the conditions of biological experiments. The effect of induced differentiation of cells was the best when cultured alone. After direct contact between the two types of cells at 2D or 3D level, the induced differentiation of cells was inhibited to varying degrees, although they still showed osteogenesis and osteoclast. After the cells were induced by indirect contact culture, the effect of induced differentiation improved when compared with direct contact culture, although it was still not as good as that of single culture. On the whole, the effect of inducing differentiation at 3D level was the same as that at 2D level, and its relative gene expression and enzyme activity were higher than that in the control group. Hence the scaffold used in this study could induce osteogenesis as well as osteoclast, thereby rendering it more effective in inducing new bone formation. Conclusion: This method can be used to construct the model of 3D bone remodeling mechanism.
Collapse
Affiliation(s)
- Xiao Lv
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Chenyang Zhang
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Xingzhu Liu
- West China Hospital, Sichuan University, Hangzhou, China
| | - Ping Li
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Yadong Yang
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
24
|
Jakob Y, Kern J, Gvaramia D, Fisch P, Magritz R, Reutter S, Rotter N. Suitability of Ex Vivo-Expanded Microtic Perichondrocytes for Auricular Reconstruction. Cells 2024; 13:141. [PMID: 38247833 PMCID: PMC10814984 DOI: 10.3390/cells13020141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024] Open
Abstract
Tissue engineering (TE) techniques offer solutions for tissue regeneration but require large quantities of cells. For microtia patients, TE methods represent a unique opportunity for therapies with low donor-site morbidity and reliance on the surgeon's individual expertise. Microtia-derived chondrocytes and perichondrocytes are considered a valuable cell source for autologous reconstruction of the pinna. The aim of this study was to investigate the suitability of perichondrocytes from microtia patients for autologous reconstruction in comparison to healthy perichondrocytes and microtia chondrocytes. Perichondrocytes were isolated via two different methods: explant culture and enzymatic digestion. The isolated cells were analyzed in vitro for their chondrogenic cell properties. We examined migration activity, colony-forming ability, expression of mesenchymal stem cell markers, and gene expression profile. We found that microtic perichondrocytes exhibit similar chondrogenic properties compared to chondrocytes in vitro. We investigated the behavior in three-dimensional cell cultures (spheroids and scaffold-based 3D cell cultures) and assessed the expression of cartilage-specific proteins via immunohistochemistry, e.g., collagen II, which was detected in all samples. Our results show that perichondrocytes from microtia patients are comparable to healthy perichondrocytes and chondrocytes in terms of chondrogenic cell properties and could therefore be a promising cell source for auricular reconstruction.
Collapse
Affiliation(s)
- Yvonne Jakob
- Department of Otorhinolaryngology Head and Neck Surgery, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany; (J.K.); (D.G.); (N.R.)
| | - Johann Kern
- Department of Otorhinolaryngology Head and Neck Surgery, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany; (J.K.); (D.G.); (N.R.)
| | - David Gvaramia
- Department of Otorhinolaryngology Head and Neck Surgery, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany; (J.K.); (D.G.); (N.R.)
| | - Philipp Fisch
- Tissue Engineering and Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zurich, Otto-Stern-Weg 7, CH-8093 Zurich, Switzerland;
| | - Ralph Magritz
- Clinic for Otorhinolaryngology, Oberhavel-Kliniken GmbH, Klinik Henningsdorf, Marwitzer Strasse 91, D-16761 Henningsdorf, Germany;
| | - Sven Reutter
- Department of Otorhinolaryngology Head and Neck Surgery, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany; (J.K.); (D.G.); (N.R.)
| | - Nicole Rotter
- Department of Otorhinolaryngology Head and Neck Surgery, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany; (J.K.); (D.G.); (N.R.)
| |
Collapse
|
25
|
Amondarain M, Gallego I, Puras G, Saenz-Del-Burgo L, Luzzani C, Pedraz JL. The role of microfluidics and 3D-bioprinting in the future of exosome therapy. Trends Biotechnol 2023; 41:1343-1359. [PMID: 37302911 DOI: 10.1016/j.tibtech.2023.05.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 04/28/2023] [Accepted: 05/18/2023] [Indexed: 06/13/2023]
Abstract
Exosome-based strategies constitute a promising tool for therapeutics, avoiding potential immunogenic and tumorigenic side-effects of cell therapies. However, the collection of a suitable exosome pool, and the need for high doses with conventional administration approaches, hamper their clinical translation. To overcome these challenges, versatile exosome collection strategies together with advanced delivery platforms may represent major progress in this field. Microfluidics enables large-scale gathering of both natural and synthetic exosomes for their implementation into bioinks, while 3D-bioprinting holds great promise in regenerative medicine with the use of exosome-loaded scaffolds that mimic the target tissue with controlled pharmacokinetics and pharmacodynamics. Hence, the combination of both strategies might become the key for the translation of exosome therapies to clinical practice.
Collapse
Affiliation(s)
- Mikele Amondarain
- CONICET - Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Laboratorio de Investigación Aplicada a Neurociencias (LIAN), Buenos Aires, Argentina
| | - Idoia Gallego
- Laboratory of Pharmaceutics, NanoBioCel Group, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
| | - Gustavo Puras
- Laboratory of Pharmaceutics, NanoBioCel Group, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Laura Saenz-Del-Burgo
- Laboratory of Pharmaceutics, NanoBioCel Group, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Carlos Luzzani
- CONICET - Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Laboratorio de Investigación Aplicada a Neurociencias (LIAN), Buenos Aires, Argentina
| | - José Luis Pedraz
- Laboratory of Pharmaceutics, NanoBioCel Group, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
| |
Collapse
|
26
|
Dutta SD, Ganguly K, Patil TV, Randhawa A, Lim KT. Unraveling the potential of 3D bioprinted immunomodulatory materials for regulating macrophage polarization: State-of-the-art in bone and associated tissue regeneration. Bioact Mater 2023; 28:284-310. [PMID: 37303852 PMCID: PMC10248805 DOI: 10.1016/j.bioactmat.2023.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/29/2023] [Accepted: 05/20/2023] [Indexed: 06/13/2023] Open
Abstract
Macrophage-assisted immunomodulation is an alternative strategy in tissue engineering, wherein the interplay between pro-inflammatory and anti-inflammatory macrophage cells and body cells determines the fate of healing or inflammation. Although several reports have demonstrated that tissue regeneration depends on spatial and temporal regulation of the biophysical or biochemical microenvironment of the biomaterial, the underlying molecular mechanism behind immunomodulation is still under consideration for developing immunomodulatory scaffolds. Currently, most fabricated immunomodulatory platforms reported in the literature show regenerative capabilities of a particular tissue, for example, endogenous tissue (e.g., bone, muscle, heart, kidney, and lungs) or exogenous tissue (e.g., skin and eye). In this review, we briefly introduced the necessity of the 3D immunomodulatory scaffolds and nanomaterials, focusing on material properties and their interaction with macrophages for general readers. This review also provides a comprehensive summary of macrophage origin and taxonomy, their diverse functions, and various signal transduction pathways during biomaterial-macrophage interaction, which is particularly helpful for material scientists and clinicians for developing next-generation immunomodulatory scaffolds. From a clinical standpoint, we briefly discussed the role of 3D biomaterial scaffolds and/or nanomaterial composites for macrophage-assisted tissue engineering with a special focus on bone and associated tissues. Finally, a summary with expert opinion is presented to address the challenges and future necessity of 3D bioprinted immunomodulatory materials for tissue engineering.
Collapse
Affiliation(s)
- Sayan Deb Dutta
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Keya Ganguly
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Tejal V. Patil
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Aayushi Randhawa
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| |
Collapse
|
27
|
Sabra DM, Krin A, Romeral AB, Frieß JL, Jeremias G. Anthrax revisited: how assessing the unpredictable can improve biosecurity. Front Bioeng Biotechnol 2023; 11:1215773. [PMID: 37795173 PMCID: PMC10546327 DOI: 10.3389/fbioe.2023.1215773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/24/2023] [Indexed: 10/06/2023] Open
Abstract
B. anthracis is one of the most often weaponized pathogens. States had it in their bioweapons programs and criminals and terrorists have used or attempted to use it. This study is motivated by the narrative that emerging and developing technologies today contribute to the amplification of danger through greater easiness, accessibility and affordability of steps in the making of an anthrax weapon. As states would have way better preconditions if they would decide for an offensive bioweapons program, we focus on bioterrorism. This paper analyzes and assesses the possible bioterrorism threat arising from advances in synthetic biology, genome editing, information availability, and other emerging, and converging sciences and enabling technologies. Methodologically we apply foresight methods to encourage the analysis of contemporary technological advances. We have developed a conceptual six-step foresight science framework approach. It represents a synthesis of various foresight methodologies including literature review, elements of horizon scanning, trend impact analysis, red team exercise, and free flow open-ended discussions. Our results show a significant shift in the threat landscape. Increasing affordability, widespread distribution, efficiency, as well as ease of use of DNA synthesis, and rapid advances in genome-editing and synthetic genomic technologies lead to an ever-growing number and types of actors who could potentially weaponize B. anthracis. Understanding the current and future capabilities of these technologies and their potential for misuse critically shapes the current and future threat landscape and underlines the necessary adaptation of biosecurity measures in the spheres of multi-level political decision making and in the science community.
Collapse
Affiliation(s)
- Dunja Manal Sabra
- Carl Friedrich von Weizsäcker-Centre for Science and Peace Research (ZNF), University of Hamburg, Bogenallee, Hamburg, Germany
| | | | | | | | | |
Collapse
|
28
|
Deniz Derman I, Yeo M, Castaneda DC, Callender M, Horvath M, Mo Z, Xiong R, Fleming E, Chen P, Peeples ME, Palucka K, Oh J, Ozbolat IT. High-throughput bioprinting of the nasal epithelium using patient-derived nasal epithelial cells. Biofabrication 2023; 15:044103. [PMID: 37536321 PMCID: PMC10424246 DOI: 10.1088/1758-5090/aced23] [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: 02/02/2023] [Revised: 07/20/2023] [Accepted: 08/03/2023] [Indexed: 08/05/2023]
Abstract
Progenitor human nasal epithelial cells (hNECs) are an essential cell source for the reconstruction of the respiratory pseudostratified columnar epithelium composed of multiple cell types in the context of infection studies and disease modeling. Hitherto, manual seeding has been the dominant method for creating nasal epithelial tissue models through biofabrication. However, this approach has limitations in terms of achieving the intricate three-dimensional (3D) structure of the natural nasal epithelium. 3D bioprinting has been utilized to reconstruct various epithelial tissue models, such as cutaneous, intestinal, alveolar, and bronchial epithelium, but there has been no attempt to use of 3D bioprinting technologies for reconstruction of the nasal epithelium. In this study, for the first time, we demonstrate the reconstruction of the nasal epithelium with the use of primary hNECs deposited on Transwell inserts via droplet-based bioprinting (DBB), which enabled high-throughput fabrication of the nasal epithelium in Transwell inserts of 24-well plates. DBB of progenitor hNECs ranging from one-tenth to one-half of the cell seeding density employed during the conventional cell seeding approach enabled a high degree of differentiation with the presence of cilia and tight-junctions over a 4 weeks air-liquid interface culture. Single cell RNA sequencing of these cultures identified five major epithelial cells populations, including basal, suprabasal, goblet, club, and ciliated cells. These cultures recapitulated the pseudostratified columnar epithelial architecture present in the native nasal epithelium and were permissive to respiratory virus infection. These results denote the potential of 3D bioprinting for high-throughput fabrication of nasal epithelial tissue models not only for infection studies but also for other purposes, such as disease modeling, immunological studies, and drug screening.
Collapse
Affiliation(s)
- I Deniz Derman
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, United States of America
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, United States of America
| | - Miji Yeo
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, United States of America
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, United States of America
| | | | - Megan Callender
- The Jackson Laboratory, Farmington, CT 06032, United States of America
| | - Mian Horvath
- The Jackson Laboratory, Farmington, CT 06032, United States of America
| | - Zengshuo Mo
- The Jackson Laboratory, Farmington, CT 06032, United States of America
| | - Ruoyun Xiong
- The Jackson Laboratory, Farmington, CT 06032, United States of America
| | - Elizabeth Fleming
- The Jackson Laboratory, Farmington, CT 06032, United States of America
| | - Phylip Chen
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, United States of America
| | - Mark E Peeples
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, United States of America
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH 43210, United States of America
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, United States of America
| | - Karolina Palucka
- The Jackson Laboratory, Farmington, CT 06032, United States of America
| | - Julia Oh
- The Jackson Laboratory, Farmington, CT 06032, United States of America
| | - Ibrahim T Ozbolat
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, United States of America
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, United States of America
- Biomedical Engineering Department, Penn State University, University Park, PA 16802, United States of America
- Materials Research Institute, Penn State University, University Park, PA 16802, United States of America
- Cancer Institute, Penn State University, University Park, PA 16802, United States of America
- Neurosurgery Department, Penn State University, University Park, PA 16802, United States of America
- Department of Medical Oncology, Cukurova University, Adana, Turkey
- Biotechnology Research and Application Center, Cukurova University, Adana, Turkey
| |
Collapse
|
29
|
Alqahtani AM. Guided Tissue and Bone Regeneration Membranes: A Review of Biomaterials and Techniques for Periodontal Treatments. Polymers (Basel) 2023; 15:3355. [PMID: 37631412 PMCID: PMC10457807 DOI: 10.3390/polym15163355] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
This comprehensive review provides an in-depth analysis of the use of biomaterials in the processes of guided tissue and bone regeneration, and their indispensable role in dental therapeutic interventions. These interventions serve the critical function of restoring both structural integrity and functionality to the dentition that has been lost or damaged. The basis for this review is laid through the exploration of various relevant scientific databases such as Scopus, PubMed, Web of science and MEDLINE. From a meticulous selection, relevant literature was chosen. This review commences by examining the different types of membranes used in guided bone regeneration procedures and the spectrum of biomaterials employed in these operations. It then explores the manufacturing technologies for the scaffold, delving into their significant impact on tissue and bone regenerations. At the core of this review is the method of guided bone regeneration, which is a crucial technique for counteracting bone loss induced by tooth extraction or periodontal disease. The discussion advances by underscoring the latest innovations and strategies in the field of tissue regeneration. One key observation is the critical role that membranes play in guided reconstruction; they serve as a barrier, preventing the entry of non-ossifying cells, thereby promoting the successful growth and regeneration of bone and tissue. By reviewing the existing literature on biomaterials, membranes, and scaffold manufacturing technologies, this paper illustrates the vast potential for innovation and growth within the field of dental therapeutic interventions, particularly in guided tissue and bone regeneration.
Collapse
Affiliation(s)
- Ali M Alqahtani
- Department of Restorative Dental Sciences, College of Dentistry, King Khalid University, Al Fara, Abha 62223, Saudi Arabia
| |
Collapse
|
30
|
Wu CA, Zhu Y, Woo YJ. Advances in 3D Bioprinting: Techniques, Applications, and Future Directions for Cardiac Tissue Engineering. Bioengineering (Basel) 2023; 10:842. [PMID: 37508869 PMCID: PMC10376421 DOI: 10.3390/bioengineering10070842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Cardiovascular diseases are the leading cause of morbidity and mortality in the United States. Cardiac tissue engineering is a direction in regenerative medicine that aims to repair various heart defects with the long-term goal of artificially rebuilding a full-scale organ that matches its native structure and function. Three-dimensional (3D) bioprinting offers promising applications through its layer-by-layer biomaterial deposition using different techniques and bio-inks. In this review, we will introduce cardiac tissue engineering, 3D bioprinting processes, bioprinting techniques, bio-ink materials, areas of limitation, and the latest applications of this technology, alongside its future directions for further innovation.
Collapse
Affiliation(s)
- Catherine A Wu
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Yuanjia Zhu
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
31
|
Veeravalli RS, Vejandla B, Savani S, Nelluri A, Peddi NC. Three-Dimensional Bioprinting in Medicine: A Comprehensive Overview of Current Progress and Challenges Faced. Cureus 2023; 15:e41624. [PMID: 37565118 PMCID: PMC10410602 DOI: 10.7759/cureus.41624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2023] [Indexed: 08/12/2023] Open
Abstract
The shortage of organs for transplantation is a global crisis, with an increasing demand and an inadequate supply of organ donors. The convergence of biology and engineering has led to the emergence of 3D bioprinting, which enables the precise and customizable construction of biological structures. Various 3D bioprinting techniques include inkjet printing, extrusion printing, and laser-assisted bioprinting (LAB). Although it has the potential for many benefits, 3D bioprinting comes with its own set of challenges and requirements, specifically associated with the bioprinting of various tissues. The challenges of bioprinting include issues with cells, bioinks, and bioprinters, as well as ethical concerns, clinical efficacy, and cost-effectiveness, making it difficult to integrate 3D bioprinting into widespread clinical practice. Three-dimensional bioprinting holds great promise in addressing the organ shortage crisis, and its applications extend beyond organ transplantation to include drug screening, disease modeling, and regenerative medicine. However, further research is needed to overcome the technical, biological, and ethical challenges associated with 3D bioprinting, paving the way for its widespread clinical implementation. This article discusses the processes and challenges of bioprinting as well as the current research direction in the field.
Collapse
Affiliation(s)
| | | | - Sarah Savani
- Medicine, Loyola University Chicago, Chicago, USA
| | | | | |
Collapse
|
32
|
Manikandan C, Jaiswal AK. Scaffold-based spheroid models of glioblastoma multiforme and its use in drug screening. Biotechnol Bioeng 2023. [PMID: 37366303 DOI: 10.1002/bit.28481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/30/2023] [Accepted: 06/12/2023] [Indexed: 06/28/2023]
Abstract
Among several types of brain cancers, glioblastoma multiforme (GBM) is a terminal and aggressive disease with a median survival of 15 months despite the most intensive surgery and chemotherapy. Preclinical models that accurately reproduce the tumor microenvironment are vital for developing new therapeutic alternatives. Understanding the complicated interactions between cells and their surroundings is essential to comprehend the tumor's microenvironment, however the monolayer cell culture approach falls short. Numerous approaches are used to develop GBM cells into tumor spheroids, while scaffold-based spheroids provides the opportunity to investigate the synergies between cells as well as cells and the matrix. This review summarizes the development of various scaffold-based GBM spheroid models and the prospective for their use as drug testing systems.
Collapse
Affiliation(s)
- Ceera Manikandan
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
- Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology, Vellore, India
| | - Amit Kumar Jaiswal
- Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
33
|
Fang W, Yang M, Liu M, Jin Y, Wang Y, Yang R, Wang Y, Zhang K, Fu Q. Review on Additives in Hydrogels for 3D Bioprinting of Regenerative Medicine: From Mechanism to Methodology. Pharmaceutics 2023; 15:1700. [PMID: 37376148 PMCID: PMC10302687 DOI: 10.3390/pharmaceutics15061700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
The regeneration of biological tissues in medicine is challenging, and 3D bioprinting offers an innovative way to create functional multicellular tissues. One common way in bioprinting is bioink, which is one type of the cell-loaded hydrogel. For clinical application, however, the bioprinting still suffers from satisfactory performance, e.g., in vascularization, effective antibacterial, immunomodulation, and regulation of collagen deposition. Many studies incorporated different bioactive materials into the 3D-printed scaffolds to optimize the bioprinting. Here, we reviewed a variety of additives added to the 3D bioprinting hydrogel. The underlying mechanisms and methodology for biological regeneration are important and will provide a useful basis for future research.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kaile Zhang
- Department of Urology, Affiliated Sixth People’s Hospital, Shanghai Jiaotong University, No. 600 Yi-Shan Road, Shanghai 200233, China; (W.F.); (M.Y.)
| | - Qiang Fu
- Department of Urology, Affiliated Sixth People’s Hospital, Shanghai Jiaotong University, No. 600 Yi-Shan Road, Shanghai 200233, China; (W.F.); (M.Y.)
| |
Collapse
|
34
|
Dabaghi M, Carpio MB, Saraei N, Moran-Mirabal JM, Kolb MR, Hirota JA. A roadmap for developing and engineering in vitro pulmonary fibrosis models. BIOPHYSICS REVIEWS 2023; 4:021302. [PMID: 38510343 PMCID: PMC10903385 DOI: 10.1063/5.0134177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 04/03/2023] [Indexed: 03/22/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a severe form of pulmonary fibrosis. IPF is a fatal disease with no cure and is challenging to diagnose. Unfortunately, due to the elusive etiology of IPF and a late diagnosis, there are no cures for IPF. Two FDA-approved drugs for IPF, nintedanib and pirfenidone, slow the progression of the disease, yet fail to cure or reverse it. Furthermore, most animal models have been unable to completely recapitulate the physiology of human IPF, resulting in the failure of many drug candidates in preclinical studies. In the last few decades, the development of new IPF drugs focused on changes at the cellular level, as it was believed that the cells were the main players in IPF development and progression. However, recent studies have shed light on the critical role of the extracellular matrix (ECM) in IPF development, where the ECM communicates with cells and initiates a positive feedback loop to promote fibrotic processes. Stemming from this shift in the understanding of fibrosis, there is a need to develop in vitro model systems that mimic the human lung microenvironment to better understand how biochemical and biomechanical cues drive fibrotic processes in IPF. However, current in vitro cell culture platforms, which may include substrates with different stiffness or natural hydrogels, have shortcomings in recapitulating the complexity of fibrosis. This review aims to draw a roadmap for developing advanced in vitro pulmonary fibrosis models, which can be leveraged to understand better different mechanisms involved in IPF and develop drug candidates with improved efficacy. We begin with a brief overview defining pulmonary fibrosis and highlight the importance of ECM components in the disease progression. We focus on fibroblasts and myofibroblasts in the context of ECM biology and fibrotic processes, as most conventional advanced in vitro models of pulmonary fibrosis use these cell types. We transition to discussing the parameters of the 3D microenvironment that are relevant in pulmonary fibrosis progression. Finally, the review ends by summarizing the state of the art in the field and future directions.
Collapse
Affiliation(s)
- Mohammadhossein Dabaghi
- Firestone Institute for Respiratory Health—Division of Respirology, Department of Medicine, McMaster University, St. Joseph's Healthcare Hamilton, 50 Charlton Avenue East, Hamilton, Ontario L8N 4A6, Canada
| | - Mabel Barreiro Carpio
- Department of Chemistry and Chemical Biology, McMaster University, Arthur N. Bourns Science Building, 1280 Main Street West, Hamilton, Ontario L8S 4M1, Canada
| | - Neda Saraei
- School of Biomedical Engineering, McMaster University, Engineering Technology Building, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | | | - Martin R. Kolb
- Firestone Institute for Respiratory Health—Division of Respirology, Department of Medicine, McMaster University, St. Joseph's Healthcare Hamilton, 50 Charlton Avenue East, Hamilton, Ontario L8N 4A6, Canada
| | | |
Collapse
|
35
|
He N, Wang X, Shi L, Li J, Mo L, Chen F, Huang Y, Liu H, Zhu X, Zhu W, Mao Y, Han X. Photoinhibiting via simultaneous photoabsorption and free-radical reaction for high-fidelity light-based bioprinting. Nat Commun 2023; 14:3063. [PMID: 37244910 DOI: 10.1038/s41467-023-38838-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 05/17/2023] [Indexed: 05/29/2023] Open
Abstract
Light-based 3D bioprinting is now employed widely to fabricate geometrically complex constructs for various biomedical applications. However, the inherent light scattering defect creates significant challenges in patterning dilute hydrogels to form high-fidelity structures with fine-scale features. Herein, we introduce a photoinhibiting approach that can effectively suppress the light scattering effect via a mechanism of simultaneous photoabsorption and free-radical reaction. This biocompatible approach significantly improves the printing resolution (~1.2 - ~2.1 pixels depending on swelling) and shape fidelity (geometric error less than 5%), while minimising the costly trial-and-error procedures. The capability in patterning 3D complex constructs using different hydrogels is demonstrated by manufacturing various scaffolds featuring intricate multi-sized channels and thin-walled networks. Importantly, cellularised gyroid scaffolds (HepG2) are fabricated successfully, exhibiting high cell proliferation and functionality. The strategy established in this study promotes the printability and operability of light-based 3D bioprinting systems, allowing numerous new applications for tissue engineering.
Collapse
Affiliation(s)
- Ning He
- National Engineering Research Centre for High Efficiency Grinding, Hunan University, 410082, Changsha, China
- State Key Laboratory of Advanced Design and Manufacture for Vehicle Body, Hunan University, 410082, Changsha, China
| | - Xiaonan Wang
- College of Biology, Hunan University, 410082, Changsha, China
| | - Liyang Shi
- College of Biology, Hunan University, 410082, Changsha, China
| | - Jing Li
- National Engineering Research Centre for High Efficiency Grinding, Hunan University, 410082, Changsha, China
| | - Lan Mo
- College of Food Science and Technology, Hunan Agricultural University, 410128, Changsha, China
| | - Feng Chen
- National Engineering Research Centre for High Efficiency Grinding, Hunan University, 410082, Changsha, China.
| | - Yuting Huang
- College of Material Science and Engineering, Hunan University, 410082, Changsha, China
| | - Hairong Liu
- College of Material Science and Engineering, Hunan University, 410082, Changsha, China
| | - Xiaolong Zhu
- National Engineering Research Centre for High Efficiency Grinding, Hunan University, 410082, Changsha, China
| | - Wei Zhu
- National Engineering Research Centre for High Efficiency Grinding, Hunan University, 410082, Changsha, China
| | - Yiqi Mao
- National Engineering Research Centre for High Efficiency Grinding, Hunan University, 410082, Changsha, China
| | - Xiaoxiao Han
- National Engineering Research Centre for High Efficiency Grinding, Hunan University, 410082, Changsha, China.
- State Key Laboratory of Advanced Design and Manufacture for Vehicle Body, Hunan University, 410082, Changsha, China.
| |
Collapse
|
36
|
Abstract
Bioprinting as an extension of 3D printing offers capabilities for printing tissues and organs for application in biomedical engineering. Conducting bioprinting in space, where the gravity is zero, can enable new frontiers in tissue engineering. Fabrication of soft tissues, which usually collapse under their own weight, can be accelerated in microgravity conditions as the external forces are eliminated. Furthermore, human colonization in space can be supported by providing critical needs of life and ecosystems by 3D bioprinting without relying on cargos from Earth, e.g., by development and long-term employment of living engineered filters (such as sea sponges-known as critical for initiating and maintaining an ecosystem). This review covers bioprinting methods in microgravity along with providing an analysis on the process of shipping bioprinters to space and presenting a perspective on the prospects of zero-gravity bioprinting.
Collapse
Affiliation(s)
- Misagh Rezapour Sarabi
- Mechanical Engineering Department, School of Engineering, Koç University, Istanbul, Turkey 34450
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, Stuttgart, Germany 70569
| | - Ali K Yetisen
- Department of Chemical Engineering, Imperial College London, London SW7 2AZ, U.K
| | - Savas Tasoglu
- Mechanical Engineering Department, School of Engineering, Koç University, Istanbul, Turkey 34450
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, Stuttgart, Germany 70569
- Koç University Translational Medicine Research Center (KUTTAM), Koç University, Istanbul, Turkey 34450
- Koç University Arçelik Research Center for Creative Industries (KUAR), Koç University, Istanbul, Turkey 34450
- Boğaziçi Institute of Biomedical Engineering, Boğaziçi University, Istanbul, Turkey 34684
| |
Collapse
|
37
|
Liu C, Tong YW. Interfacial Polyelectrolyte Complexation-Inspired Bioprinting of Vascular Constructs. ACS APPLIED MATERIALS & INTERFACES 2023; 15:20712-20725. [PMID: 37071430 DOI: 10.1021/acsami.3c01199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Bioprinting is a precise layer-by-layer manufacturing technology utilizing biomaterials, cells, and sometimes growth factors for the fabrication of customized three-dimensional (3D) biological constructs. In recent years, it has gained considerable interest in various biomedical studies. However, the translational application of bioprinting is currently impeded by the lack in efficient techniques for blood vessel fabrications. In this report, by systematically studying the previously reported phenomenon, interfacial polyelectrolyte complexation, an efficient blood vessel bioprinting technique based on the phenomenon, was proposed and subsequently investigated. In this technique, anionic hyaluronate and cationic lysine-based peptide amphiphiles were placed concentrically to bioprint with human umbilical endothelial cells for the fabrication of biological tubular constructs. These constructs demonstrated clear vascular features, which made them highly resemble blood vessels. In addition, to optimize the bioactivity of the printed constructs, this report also, for the first time, studied peptide sequencing's effect on the biocompatibility of the polyelectrolyte-peptide amphiphile complex. All these studies conducted in the report are highly relevant and interesting for research in vascular structure fabrication, which will eventually be beneficial for translational application development of bioprinting.
Collapse
Affiliation(s)
- Chixuan Liu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585
| | - Yen Wah Tong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585
| |
Collapse
|
38
|
Duarte AC, Costa EC, Filipe HAL, Saraiva SM, Jacinto T, Miguel SP, Ribeiro MP, Coutinho P. Animal-derived products in science and current alternatives. BIOMATERIALS ADVANCES 2023; 151:213428. [PMID: 37146527 DOI: 10.1016/j.bioadv.2023.213428] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/08/2023] [Accepted: 04/11/2023] [Indexed: 05/07/2023]
Abstract
More than fifty years after the 3Rs definition and despite the continuous implementation of regulatory measures, animals continue to be widely used in basic research. Their use comprises not only in vivo experiments with animal models, but also the production of a variety of supplements and products of animal origin for cell and tissue culture, cell-based assays, and therapeutics. The animal-derived products most used in basic research are fetal bovine serum (FBS), extracellular matrix proteins such as Matrigel™, and antibodies. However, their production raises several ethical issues regarding animal welfare. Additionally, their biological origin is associated with a high risk of contamination, resulting, frequently, in poor scientific data for clinical translation. These issues support the search for new animal-free products able to replace FBS, Matrigel™, and antibodies in basic research. In addition, in silico methodologies play an important role in the reduction of animal use in research by refining the data previously to in vitro and in vivo experiments. In this review, we depicted the current available animal-free alternatives in in vitro research.
Collapse
Affiliation(s)
- Ana C Duarte
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal; CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - Elisabete C Costa
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal
| | - Hugo A L Filipe
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal
| | - Sofia M Saraiva
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal
| | - Telma Jacinto
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal
| | - Sónia P Miguel
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal; CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - Maximiano P Ribeiro
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal; CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - Paula Coutinho
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal; CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal.
| |
Collapse
|
39
|
Raees S, Ullah F, Javed F, Akil HM, Jadoon Khan M, Safdar M, Din IU, Alotaibi MA, Alharthi AI, Bakht MA, Ahmad A, Nassar AA. Classification, processing, and applications of bioink and 3D bioprinting: A detailed review. Int J Biol Macromol 2023; 232:123476. [PMID: 36731696 DOI: 10.1016/j.ijbiomac.2023.123476] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/12/2023] [Accepted: 01/25/2023] [Indexed: 02/02/2023]
Abstract
With the advancement in 3D bioprinting technology, cell culture methods can design 3D environments which are both, complex and physiologically relevant. The main component in 3D bioprinting, bioink, can be split into various categories depending on the criterion of categorization. Although the choice of bioink and bioprinting process will vary greatly depending on the application, general features such as material properties, biological interaction, gelation, and viscosity are always important to consider. The foundation of 3D bioprinting is the exact layer-by-layer implantation of biological elements, biochemicals, and living cells with the spatial control of the implantation of functional elements onto the biofabricated 3D structure. Three basic strategies underlie the 3D bioprinting process: autonomous self-assembly, micro tissue building blocks, and biomimicry or biomimetics. Tissue engineering can benefit from 3D bioprinting in many ways, but there are still numerous obstacles to overcome before functional tissues can be produced and used in clinical settings. A better comprehension of the physiological characteristics of bioink materials and a higher level of ability to reproduce the intricate biologically mimicked and physiologically relevant 3D structures would be a significant improvement for 3D bioprinting to overcome the limitations.
Collapse
Affiliation(s)
- Sania Raees
- Department of Biosciences, COMSATS University Islamabad, Park Road, 45520 Islamabad, Pakistan
| | - Faheem Ullah
- Department of Biological Sciences, National University of Medical Sciences, NUMS, Rawalpindi 46000, Pakistan; School of Materials and Mineral Resources Engineering, Engineering Campus, Universiti Sains Malaysia, Seri Ampangan, 14300 Nibong Tebal, Pulau Pinang, Malaysia
| | - Fatima Javed
- Department of Chemistry, Shaheed Benazir Bhutto Women University, Peshawar 25000, KPK, Pakistan
| | - Hazizan Md Akil
- School of Materials and Mineral Resources Engineering, Engineering Campus, Universiti Sains Malaysia, Seri Ampangan, 14300 Nibong Tebal, Pulau Pinang, Malaysia
| | - Muhammad Jadoon Khan
- Department of Biosciences, COMSATS University Islamabad, Park Road, 45520 Islamabad, Pakistan
| | - Muhammad Safdar
- Department of Pharmacy, Gomal University D. I Khan, KPK, Pakistan
| | - Israf Ud Din
- Department of Chemistry, College of Science and Humanities, Prince Sattam bin Abdulaziz University, 16278 Al-Kharj, Saudi Arabia.
| | - Mshari A Alotaibi
- Department of Chemistry, College of Science and Humanities, Prince Sattam bin Abdulaziz University, 16278 Al-Kharj, Saudi Arabia
| | - Abdulrahman I Alharthi
- Department of Chemistry, College of Science and Humanities, Prince Sattam bin Abdulaziz University, 16278 Al-Kharj, Saudi Arabia
| | - M Afroz Bakht
- Department of Chemistry, College of Science and Humanities, Prince Sattam bin Abdulaziz University, 16278 Al-Kharj, Saudi Arabia
| | - Akil Ahmad
- Department of Chemistry, College of Science and Humanities, Prince Sattam bin Abdulaziz University, 16278 Al-Kharj, Saudi Arabia
| | - Amal A Nassar
- Department of Chemistry, College of Science and Humanities, Prince Sattam bin Abdulaziz University, 16278 Al-Kharj, Saudi Arabia
| |
Collapse
|
40
|
Derman ID, Yeo M, Castaneda DC, Callender M, Horvath M, Mo Z, Xiong R, Fleming E, Chen P, Peeples ME, Palucka K, Oh J, Ozbolat IT. High-Throughput Bioprinting of the Nasal Epithelium using Patient-derived Nasal Epithelial Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.29.534723. [PMID: 37034627 PMCID: PMC10081172 DOI: 10.1101/2023.03.29.534723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Human nasal epithelial cells (hNECs) are an essential cell source for the reconstruction of the respiratory pseudostratified columnar epithelium composed of multiple cell types in the context of infection studies and disease modeling. Hitherto, manual seeding has been the dominant method for creating nasal epithelial tissue models. However, the manual approach is slow, low-throughput and has limitations in terms of achieving the intricate 3D structure of the natural nasal epithelium in a uniform manner. 3D Bioprinting has been utilized to reconstruct various epithelial tissue models, such as cutaneous, intestinal, alveolar, and bronchial epithelium, but there has been no attempt to use of 3D bioprinting technologies for reconstruction of the nasal epithelium. In this study, for the first time, we demonstrate the reconstruction of the nasal epithelium with the use of primary hNECs deposited on Transwell inserts via droplet-based bioprinting (DBB), which enabled high-throughput fabrication of the nasal epithelium in Transwell inserts of 24-well plates. DBB of nasal progenitor cells ranging from one-tenth to one-half of the cell seeding density employed during the conventional cell seeding approach enabled a high degree of differentiation with the presence of cilia and tight-junctions over a 4-week air-liquid interface culture. Single cell RNA sequencing of these cultures identified five major epithelial cells populations, including basal, suprabasal, goblet, club, and ciliated cells. These cultures recapitulated the pseudostratified columnar epithelial architecture present in the native nasal epithelium and were permissive to respiratory virus infection. These results denote the potential of 3D bioprinting for high-throughput fabrication of nasal epithelial tissue models not only for infection studies but also for other purposes such as disease modeling, immunological studies, and drug screening.
Collapse
|
41
|
Harding A, Pramanik A, Basak A, Prakash C, Shankar S. Application of additive manufacturing in the biomedical field- A review. ANNALS OF 3D PRINTED MEDICINE 2023. [DOI: 10.1016/j.stlm.2023.100110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023] Open
|
42
|
Liu S, Cheng L, Liu Y, Zhang H, Song Y, Park JH, Dashnyam K, Lee JH, Khalak FAH, Riester O, Shi Z, Ostrovidov S, Kaji H, Deigner HP, Pedraz JL, Knowles JC, Hu Q, Kim HW, Ramalingam M. 3D Bioprinting tissue analogs: Current development and translational implications. J Tissue Eng 2023; 14:20417314231187113. [PMID: 37464999 PMCID: PMC10350769 DOI: 10.1177/20417314231187113] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/25/2023] [Indexed: 07/20/2023] Open
Abstract
Three-dimensional (3D) bioprinting is a promising and rapidly evolving technology in the field of additive manufacturing. It enables the fabrication of living cellular constructs with complex architectures that are suitable for various biomedical applications, such as tissue engineering, disease modeling, drug screening, and precision regenerative medicine. The ultimate goal of bioprinting is to produce stable, anatomically-shaped, human-scale functional organs or tissue substitutes that can be implanted. Although various bioprinting techniques have emerged to develop customized tissue-engineering substitutes over the past decade, several challenges remain in fabricating volumetric tissue constructs with complex shapes and sizes and translating the printed products into clinical practice. Thus, it is crucial to develop a successful strategy for translating research outputs into clinical practice to address the current organ and tissue crises and improve patients' quality of life. This review article discusses the challenges of the existing bioprinting processes in preparing clinically relevant tissue substitutes. It further reviews various strategies and technical feasibility to overcome the challenges that limit the fabrication of volumetric biological constructs and their translational implications. Additionally, the article highlights exciting technological advances in the 3D bioprinting of anatomically shaped tissue substitutes and suggests future research and development directions. This review aims to provide readers with insight into the state-of-the-art 3D bioprinting techniques as powerful tools in engineering functional tissues and organs.
Collapse
Affiliation(s)
- Suihong Liu
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Rapid Manufacturing Engineering Center, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai, China
- Shanghai Key Laboratory of Intelligent Manufacturing and Robotics, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai, China
- National Demonstration Center for Experimental Engineering Training Education, Shanghai University, Shanghai, China
| | - Lijia Cheng
- School of Basic Medical Sciences, Clinical Medical College and Affiliated Hospital, Chengdu University, Chengdu, China
| | - Yakui Liu
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Haiguang Zhang
- Rapid Manufacturing Engineering Center, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai, China
- Shanghai Key Laboratory of Intelligent Manufacturing and Robotics, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai, China
- National Demonstration Center for Experimental Engineering Training Education, Shanghai University, Shanghai, China
| | - Yongteng Song
- Rapid Manufacturing Engineering Center, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai, China
| | - Jeong-Hui Park
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, South Korea
- Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, South Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, South Korea
| | - Khandmaa Dashnyam
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, South Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, South Korea
- Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, South Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, South Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, South Korea
| | - Fouad Al-Hakim Khalak
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, Madrid, Spain
| | - Oliver Riester
- Institute of Precision Medicine, Furtwangen University, Jakob-Kienzle-Strasse 17, Villingen-Schwenningen, Germany
| | - Zheng Shi
- School of Basic Medical Sciences, Clinical Medical College and Affiliated Hospital, Chengdu University, Chengdu, China
| | - Serge Ostrovidov
- Department of Diagnostic and Therapeutic Systems Engineering, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hirokazu Kaji
- Department of Diagnostic and Therapeutic Systems Engineering, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hans-Peter Deigner
- Institute of Precision Medicine, Furtwangen University, Jakob-Kienzle-Strasse 17, Villingen-Schwenningen, Germany
| | - José Luis Pedraz
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, Madrid, Spain
| | - Jonathan C Knowles
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, South Korea
- Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, South Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, South Korea
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, London, UK
| | - Qingxi Hu
- Rapid Manufacturing Engineering Center, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai, China
- Shanghai Key Laboratory of Intelligent Manufacturing and Robotics, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai, China
- National Demonstration Center for Experimental Engineering Training Education, Shanghai University, Shanghai, China
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, South Korea
- Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, South Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, South Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, South Korea
| | - Murugan Ramalingam
- School of Basic Medical Sciences, Clinical Medical College and Affiliated Hospital, Chengdu University, Chengdu, China
- Institute of Precision Medicine, Furtwangen University, Jakob-Kienzle-Strasse 17, Villingen-Schwenningen, Germany
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- Joint Research Laboratory on Advanced Pharma Development Initiative, A Joined Venture of TECNALIA and School of Pharmacy, University of the Basque Country (UPV/ EHU), Vitoria-Gasteiz, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
- Bioprinting Laboratory, Centro de investigación Lascaray Ikergunea, Avenida Miguel de Unamuno, Vitoria-Gasteiz, Spain
- Department of Metallurgical and Materials Engineering, Atilim University, Ankara, Turkey
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, China
| |
Collapse
|
43
|
Liang S, Su Y, Yao R. 3D Bioprinting of Induced Pluripotent Stem Cells and Disease Modeling. Handb Exp Pharmacol 2023; 281:29-56. [PMID: 36882603 DOI: 10.1007/164_2023_646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Patient-derived induced pluripotent stem cells (iPSCs), carrying the genetic information of the disease and capable of differentiating into multilineages in vitro, are valuable for disease modeling. 3D bioprinting enables the assembly of the cell-laden hydrogel into hierarchically three-dimensional architectures that recapitulate the natural tissues and organs. Investigation of iPSC-derived physiological and pathological models constructed by 3D bioprinting is a fast-growing field still in its infancy. Distinctly from cell lines and adult stem cells, iPSCs and iPSC-derived cells are more susceptible to external stimuli which can disturb the differentiation, maturation, and organization of iPSCs and their progeny. Here we discuss the fitness of iPSCs and 3D bioprinting from the perspective of bioinks and printing technologies. We provide a timely review of the progress of 3D bioprinting iPSC-derived physiological and pathological models by exemplifying the relatively prosperous cardiac and neurological fields. We also discuss scientific rigors and highlight the remaining issues to offer a guiding framework for bioprinting-assisted personalized medicine.
Collapse
Affiliation(s)
- Shaojun Liang
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering,, Tsinghua University, Beijing, China
| | - Yijun Su
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering,, Tsinghua University, Beijing, China
| | - Rui Yao
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering,, Tsinghua University, Beijing, China.
- State Key Laboratory of Tribology in Advanced Equipment, Tsinghua University, Beijing, China.
| |
Collapse
|
44
|
Aleman J, Young CD, Karam SD, Wang XJ. Revisiting laminin and extracellular matrix remodeling in metastatic squamous cell carcinoma: What have we learned after more than four decades of research? Mol Carcinog 2023; 62:5-23. [PMID: 35596706 PMCID: PMC9676410 DOI: 10.1002/mc.23417] [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: 03/01/2022] [Accepted: 04/18/2022] [Indexed: 02/06/2023]
Abstract
Patients with squamous cell carcinoma (SCC) have significantly lower survival upon the development of distant metastases. The extracellular matrix (ECM) is a consistent yet dynamic influence on the metastatic capacity of SCCs. The ECM encompasses a milieu of structural proteins, signaling molecules, and enzymes. Just over 40 years ago, the fibrous ECM glycoprotein laminin was identified. Roughly four decades of research have revealed a pivotal role of laminins in metastasis. However, trends in ECM alterations in some cancers have been applied broadly to all metastatic diseases, despite evidence that these characteristics vary by tumor type. We will summarize how laminins influence the SCC metastatic process exclusively. Enhanced laminin protein deposition occurs at the invasive edge of SCC tumors, which correlates with elevated levels of laminin-binding β1 integrins on SCC cells, increased MMP-3 presence, worse prognosis, and lymphatic dissemination. Although these findings are significant, gaps in knowledge of the formation of a premetastatic niche, the processes of intra- and extravasation, and the contributions of the ECM to SCC metastatic cell dormancy persist. Bridging these gaps requires novel in vitro systems and animal models that reproduce tumor-stromal interactions and spontaneous metastasis seen in the clinic. These advances will allow accurate assessment of laminins to predict responders to transforming growth factor-β inhibitors and immunotherapy, as well as potential combinatorial therapies with the standard of care. Such clinical interventions may drastically improve quality of life and patient survival by explicitly targeting SCC metastasis.
Collapse
Affiliation(s)
- John Aleman
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Christian D. Young
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sana D. Karam
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Xiao-Jing Wang
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
- Veterans Affairs Medical Center, VA Eastern Colorado Health Care System, Aurora, Colorado, USA
| |
Collapse
|
45
|
Message in a Scaffold: Natural Biomaterials for Three-Dimensional (3D) Bioprinting of Human Brain Organoids. Biomolecules 2022; 13:biom13010025. [PMID: 36671410 PMCID: PMC9855696 DOI: 10.3390/biom13010025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/07/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Brain organoids are invaluable tools for pathophysiological studies or drug screening, but there are still challenges to overcome in making them more reproducible and relevant. Recent advances in three-dimensional (3D) bioprinting of human neural organoids is an emerging approach that may overcome the limitations of self-organized organoids. It requires the development of optimal hydrogels, and a wealth of research has improved our knowledge about biomaterials both in terms of their intrinsic properties and their relevance on 3D culture of brain cells and tissue. Although biomaterials are rarely biologically neutral, few articles have reviewed their roles on neural cells. We here review the current knowledge on unmodified biomaterials amenable to support 3D bioprinting of neural organoids with a particular interest in their impact on cell homeostasis. Alginate is a particularly suitable bioink base for cell encapsulation. Gelatine is a valuable helper agent for 3D bioprinting due to its viscosity. Collagen, fibrin, hyaluronic acid and laminin provide biological support to adhesion, motility, differentiation or synaptogenesis and optimize the 3D culture of neural cells. Optimization of specialized hydrogels to direct differentiation of stem cells together with an increased resolution in phenotype analysis will further extend the spectrum of possible bioprinted brain disease models.
Collapse
|
46
|
Trikalitis VD, Kroese NJJ, Kaya M, Cofiño-Fabres C, Ten Den S, Khalil ISM, Misra S, Koopman BFJM, Passier R, Schwach V, Rouwkema J. Embedded 3D printing of dilute particle suspensions into dense complex tissue fibers using shear thinning xanthan baths. Biofabrication 2022; 15. [PMID: 36347040 DOI: 10.1088/1758-5090/aca124] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 11/08/2022] [Indexed: 11/09/2022]
Abstract
In order to fabricate functional organoids and microtissues, a high cell density is generally required. As such, the placement of cell suspensions in molds or microwells to allow for cell concentration by sedimentation is the current standard for the production of organoids and microtissues. Even though molds offer some level of control over the shape of the resulting microtissue, this control is limited as microtissues tend to compact towards a sphere after sedimentation of the cells. 3D bioprinting on the other hand offers complete control over the shape of the resulting structure. Even though the printing of dense cell suspensions in the ink has been reported, extruding dense cellular suspensions is challenging and generally results in high shear stresses on the cells and a poor shape fidelity of the print. As such, additional materials such as hydrogels are added in the bioink to limit shear stresses, and to improve shape fidelity and resolution. The maximum cell concentration that can be incorporated in a hydrogel-based ink before the ink's rheological properties are compromised, is significantly lower than the concentration in a tissue equivalent. Additionally, the hydrogel components often interfere with cellular self-assembly processes. To circumvent these limitations, we report a simple and inexpensive xanthan bath based embedded printing method to 3D print dense functional linear tissues using dilute particle suspensions consisting of cells, spheroids, hydrogel beads, or combinations thereof. Using this method, we demonstrated the self-organization of functional cardiac tissue fibers with a layer of epicardial cells surrounding a body of cardiomyocytes.
Collapse
Affiliation(s)
- Vasileios D Trikalitis
- Department of Biomechanical Engineering, Vascularization Lab, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| | - Niels J J Kroese
- Department of Applied Stem Cell Technologies, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| | - Mert Kaya
- Surgical Robotics Laboratory, Department of Biomechanical Engineering, University of Twente, TechMed Center, MESA+ Institute, 7500AE Enschede, The Netherlands.,Surgical Robotics Laboratory, Department of Biomedical Engineering, University of Groningen and University Medical Centre Groningen, 9713AV Groningen, The Netherlands
| | - Carla Cofiño-Fabres
- Department of Applied Stem Cell Technologies, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| | - Simone Ten Den
- Department of Applied Stem Cell Technologies, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| | - Islam S M Khalil
- Surgical Robotics Laboratory, Department of Biomechanical Engineering, University of Twente, TechMed Center, MESA+ Institute, 7500AE Enschede, The Netherlands
| | - Sarthak Misra
- Surgical Robotics Laboratory, Department of Biomechanical Engineering, University of Twente, TechMed Center, MESA+ Institute, 7500AE Enschede, The Netherlands.,Surgical Robotics Laboratory, Department of Biomedical Engineering, University of Groningen and University Medical Centre Groningen, 9713AV Groningen, The Netherlands
| | - Bart F J M Koopman
- Department of Biomechanical Engineering, Vascularization Lab, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| | - Robert Passier
- Department of Applied Stem Cell Technologies, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| | - Verena Schwach
- Department of Applied Stem Cell Technologies, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| | - Jeroen Rouwkema
- Department of Biomechanical Engineering, Vascularization Lab, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| |
Collapse
|
47
|
Akide Ndunge OB, Kilian N, Salman MM. Cerebral Malaria and Neuronal Implications of Plasmodium Falciparum Infection: From Mechanisms to Advanced Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202944. [PMID: 36300890 PMCID: PMC9798991 DOI: 10.1002/advs.202202944] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 09/22/2022] [Indexed: 06/01/2023]
Abstract
Reorganization of host red blood cells by the malaria parasite Plasmodium falciparum enables their sequestration via attachment to the microvasculature. This artificially increases the dwelling time of the infected red blood cells within inner organs such as the brain, which can lead to cerebral malaria. Cerebral malaria is the deadliest complication patients infected with P. falciparum can experience and still remains a major public health concern despite effective antimalarial therapies. Here, the current understanding of the effect of P. falciparum cytoadherence and their secreted proteins on structural features of the human blood-brain barrier and their involvement in the pathogenesis of cerebral malaria are highlighted. Advanced 2D and 3D in vitro models are further assessed to study this devastating interaction between parasite and host. A better understanding of the molecular mechanisms leading to neuronal and cognitive deficits in cerebral malaria will be pivotal in devising new strategies to treat and prevent blood-brain barrier dysfunction and subsequent neurological damage in patients with cerebral malaria.
Collapse
Affiliation(s)
- Oscar Bate Akide Ndunge
- Department of Internal MedicineSection of Infectious DiseasesYale University School of Medicine300 Cedar StreetNew HavenCT06510USA
| | - Nicole Kilian
- Centre for Infectious Diseases, ParasitologyHeidelberg University HospitalIm Neuenheimer Feld 32469120HeidelbergGermany
| | - Mootaz M. Salman
- Department of PhysiologyAnatomy and GeneticsUniversity of OxfordOxfordOX1 3QUUK
- Kavli Institute for NanoScience DiscoveryUniversity of OxfordOxfordUK
- Oxford Parkinson's Disease CentreUniversity of OxfordOxfordUK
| |
Collapse
|
48
|
Ngomi N, Khayeka-Wandabwa C, Egondi T, Marinda PA, Haregu TN. Determinants of inequality in health care seeking for childhood illnesses: insights from Nairobi informal settlements. GLOBAL HEALTH JOURNAL 2022. [DOI: 10.1016/j.glohj.2022.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
49
|
Javaid M, Haleem A, Singh RP, Suman R. 3D printing applications for healthcare research and development. GLOBAL HEALTH JOURNAL 2022. [DOI: 10.1016/j.glohj.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
50
|
Banerjee D, Singh YP, Datta P, Ozbolat V, O'Donnell A, Yeo M, Ozbolat IT. Strategies for 3D bioprinting of spheroids: A comprehensive review. Biomaterials 2022; 291:121881. [DOI: 10.1016/j.biomaterials.2022.121881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/04/2022] [Accepted: 10/23/2022] [Indexed: 11/17/2022]
|