1
|
Yang T, Jin Y, Smith LM, Dahotre NB, Neogi A. Real-time in-situ ultrasound monitoring of soft hydrogel 3D printing with subwavelength resolution. COMMUNICATIONS ENGINEERING 2024; 3:162. [PMID: 39521874 PMCID: PMC11550851 DOI: 10.1038/s44172-024-00318-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
3D bioprinting has excellent potential in tissue engineering, regenerative medicine, and drug delivery systems due to the ability to fabricate intricate structures that are challenging to make with conventional manufacturing methods. However, the complexity of parametric combinations and lack of product quality control have restricted soft hydrogel bioprinting from practical applications. Here we show an in-situ ultrasound monitoring system that reveals the alginate-gelatin hydrogel's additive manufacturing process. We use this technique to understand the parameters that influenced transient printing behaviors and material properties in approximately real-time. This unique monitoring process can facilitate the detection of minor errors/flaws during the printing. By analyzing the ultrasonic reflected signals in both time and frequency domains, transient printing information can be obtained from 3D printed soft hydrogels during the processes with a depth subwavelength resolution approaching 0.78 λ . This in-situ technique monitors the printing behaviors regarding the constructed film, interlayer bonding, transient effective elastic constant, layer-wise surface roughness (elastic or plastic), nozzle indentation/scratching, and gravitational spreading. The simulation-verified experimental methods monitored fully infilled printing and gridded pattern printing conditions. Furthermore, the proposed ultrasound system also experimentally monitored the post-crosslinking process of alginate-gelatin hydrogel in CaCl2 solution. The results can optimize crosslinking time by balancing the hydrogel's stiffness enhancement and geometrical distortion.
Collapse
Affiliation(s)
- Teng Yang
- Department of Materials Science and Engineering, University of North Texas, Denton, TX, USA
- Department of Physics, University of North Texas, Denton, TX, USA
| | - Yuqi Jin
- Department of Physics, University of North Texas, Denton, TX, USA.
- Department of Mechanical Engineering, University of North Texas, Denton, TX, USA.
| | - Lee Miller Smith
- Department of Mechanical Engineering, University of North Texas, Denton, TX, USA
| | - Narendra B Dahotre
- Department of Materials Science and Engineering, University of North Texas, Denton, TX, USA
| | - Arup Neogi
- Department of Physics, University of North Texas, Denton, TX, USA.
| |
Collapse
|
2
|
Zhang X, Zhao G, Ma T, Simmons CA, Santerre JP. A critical review on advances and challenges of bioprinted cardiac patches. Acta Biomater 2024; 189:1-24. [PMID: 39374681 DOI: 10.1016/j.actbio.2024.09.056] [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: 06/14/2024] [Revised: 09/10/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024]
Abstract
Myocardial infarction (MI), which causes irreversible myocardium necrosis, affects 0.25 billion people globally and has become one of the most significant epidemics of our time. Over the past few years, bioprinting has moved beyond a concept of simply incorporating cells into biomaterials, to strategically defining the microenvironment (e.g., architecture, biomolecular signalling, mechanical stimuli, etc.) within which the cells are printed. Among the different bioprinting applications, myocardial repair is a field that has seen some of the most significant advances towards the management of the repaired tissue microenvironment. This review critically assesses the most recent biomedical innovations being carried out in cardiac patch bioprinting, with specific considerations given to the biomaterial design parameters, growth factors/cytokines, biomechanical and bioelectrical conditioning, as well as innovative biomaterial-based "4D" bioprinting (3D scaffold structure + temporal morphology changes) of myocardial tissues, immunomodulation and sustained delivery systems used in myocardium bioprinting. Key challenges include the ability to generate large quantities of cardiac cells, achieve high-density capillary networks, establish biomaterial designs that are comparable to native cardiac extracellular matrix, and manage the sophisticated systems needed for combining cardiac tissue microenvironmental cues while simultaneously establishing bioprinting technologies yielding both high-speed and precision. This must be achieved while considering quality assurance towards enabling reproducibility and clinical translation. Moreover, this manuscript thoroughly discussed the current clinical translational hurdles and regulatory issues associated with the post-bioprinting evaluation, storage, delivery and implantation of the bioprinted myocardial patches. Overall, this paper provides insights into how the clinical feasibility and important regulatory concerns may influence the design of the bioink (biomaterials, cell sources), fabrication and post-fabrication processes associated with bioprinting of the cardiac patches. This paper emphasizes that cardiac patch bioprinting requires extensive collaborations from imaging and 3D modelling technical experts, biomaterial scientists, additive manufacturing experts and healthcare professionals. Further, the work can also guide the field of cardiac patch bioprinting moving forward, by shedding light on the potential use of robotics and automation to increase productivity, reduce financial cost, and enable standardization and true commercialization of bioprinted cardiac patches. STATEMENT OF SIGNIFICANCE: The manuscript provides a critical review of important themes currently pursued for heart patch bioprinting, including critical biomaterial design parameters, physiologically-relevant cardiac tissue stimulations, and newly emerging cardiac tissue bioprinting strategies. This review describes the limited number of studies, to date in the literature, that describe systemic approaches to combine multiple design parameters, including capabilities to yield high-density capillary networks, establish biomaterial composite designs similar to native cardiac extracellular matrix, and incorporate cardiac tissue microenvironmental cues, while simultaneously establishing bioprinting technologies that yield high-speed and precision. New tools such as artificial intelligence may provide the analytical power to consider multiple design parameters and identify an optimized work-flow(s) for enabling the clinical translation of bioprinted cardiac patches.
Collapse
Affiliation(s)
- Xiaoqing Zhang
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong 264003, China; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario M5G 1M1, Canada.
| | - Guangtao Zhao
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong 264003, China
| | - Tianyi Ma
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam 999077, Hong Kong Special Administrative Region of China
| | - Craig A Simmons
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong 264003, China; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario M5G 1M1, Canada.
| | - J Paul Santerre
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong 264003, China; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario M5G 1M1, Canada.
| |
Collapse
|
3
|
Subramanian A, Mohanbabu J, Srinivasan T, T T, Subramaniyan V, V M, Sekar M, Wong LS. Reviewing the literature of 3D printing of bones and cartilage: Evidence and practice. ANNALS OF 3D PRINTED MEDICINE 2024; 16:100180. [DOI: 10.1016/j.stlm.2024.100180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024] Open
|
4
|
Albrecht FB, Schick AK, Klatt A, Schmidt FF, Nellinger S, Kluger PJ. Exploring Morphological and Molecular Properties of Different Adipose Cell Models: Monolayer, Spheroids, Gellan Gum-Based Hydrogels, and Explants. Macromol Biosci 2024:e2400320. [PMID: 39450850 DOI: 10.1002/mabi.202400320] [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: 09/18/2024] [Revised: 10/04/2024] [Indexed: 10/26/2024]
Abstract
White adipose tissue (WAT) plays a crucial role in energy homeostasis and secretes numerous adipokines with far-reaching effects. WAT is linked to diseases such as diabetes, cardiovascular disease, and cancer. There is a high demand for suitable in vitro models to study diseases and tissue metabolism. Most of these models are covered by 2D-monolayer cultures. This study aims to evaluate the performance of different WAT models to better derive potential applications. The stability of adipocyte characteristics in spheroids and two 3D gellan gum hydrogels with ex situ lobules and 2D-monolayer culture is analyzed. First, the differentiation to achieve adipocyte-like characteristics is determined. Second, to evaluate the maintenance of differentiated ASC-based models, an adipocyte-based model, and explants over 3 weeks, viability, intracellular lipid content, perilipin A expression, adipokine, and gene expression are analyzed. Several advantages are supported using each of the models. Including, but not limited to, the strong differentiation in 2D-monolayers, the self-assembling within spheroids, the long-term stability of the stem cell-containing hydrogels, and the mature phenotype within adipocyte-containing hydrogels and the lobules. This study highlights the advantages of 3D models due to their more in vivo-like behavior and provides an overview of the different adipose cell models.
Collapse
Affiliation(s)
- Franziska B Albrecht
- Reutlingen Research Institute, Reutlingen University, Alteburgstraße 150, 72762, Reutlingen, Germany
- Faculty of Natural Science, University of Hohenheim, Schloss Hohenheim 1, 70599, Stuttgart, Germany
| | - Ann-Kathrin Schick
- Faculty of Science, Energy and Building Services, Esslingen University, Kanalstraße 33, 73728, Esslingen, Germany
| | - Annemarie Klatt
- Reutlingen Research Institute, Reutlingen University, Alteburgstraße 150, 72762, Reutlingen, Germany
| | - Freia F Schmidt
- Reutlingen Research Institute, Reutlingen University, Alteburgstraße 150, 72762, Reutlingen, Germany
| | - Svenja Nellinger
- Reutlingen Research Institute, Reutlingen University, Alteburgstraße 150, 72762, Reutlingen, Germany
| | - Petra J Kluger
- School of Life Sciences, Reutlingen University, Alteburgstraße 150, 72762, Reutlingen, Germany
| |
Collapse
|
5
|
Arslan H, Davuluri A, Nguyen HH, So BR, Lee J, Jeon J, Yum K. 3D Bioprinting Using Universal Fugitive Network Bioinks. ACS APPLIED BIO MATERIALS 2024; 7:7040-7050. [PMID: 39291381 DOI: 10.1021/acsabm.4c01220] [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: 09/19/2024]
Abstract
Three-dimensional (3D) bioprinting has emerged with potential for creating functional 3D tissues with customized geometries. However, the limited availability of bioinks capable of printing 3D structures with both high-shape fidelity and desired biological environments for encapsulated cells remains a key challenge. Here, we present a 3D bioprinting approach that uses universal fugitive network bioinks prepared by loading cells and hydrogel precursors (bioink base materials) into a 3D printable fugitive carrier. This approach constructs 3D structures of cell-encapsulated hydrogels by printing 3D structures using fugitive network bioinks, followed by cross-linking printed structures and removing the carrier from them. The use of the fugitive carrier decouples the 3D printability of bioinks from the material properties of bioink base materials, making this approach readily applicable to a range of hydrogel systems. The decoupling also enables the design of bioinks for the biological functionality of the final printed constructs without compromising the 3D printability. We demonstrate the generalizable 3D printability by printing self-supporting 3D structures of various hydrogels, including conventionally non-3D printable hydrogels and those with a low polymer content. We conduct preprinting screening of bioink base materials through 3D cell culture to select bioinks with high cell compatibility. The selected bioinks produce 3D constructs of cell-encapsulated hydrogels with both high-shape fidelity and high cell viability and proliferation. The universal fugitive network bioink platform could significantly expand 3D printable bioinks with customizable biological functionalities and the adoption of 3D bioprinting in diverse research and applied settings.
Collapse
Affiliation(s)
- Hakan Arslan
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas 76019, United States
- Department of Mechanical and Aerospace Engineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Aneela Davuluri
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Hiep H Nguyen
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Byung Ran So
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Juhyun Lee
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Junha Jeon
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Kyungsuk Yum
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| |
Collapse
|
6
|
Khan AA, Kim JH. Recent advances in materials and manufacturing of implantable devices for continuous health monitoring. Biosens Bioelectron 2024; 261:116461. [PMID: 38850737 DOI: 10.1016/j.bios.2024.116461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/30/2024] [Accepted: 06/01/2024] [Indexed: 06/10/2024]
Abstract
Implantable devices are vital in healthcare, enabling continuous monitoring, early disease detection, informed decision-making, enhanced outcomes, cost reduction, and chronic condition management. These devices provide real-time data, allowing proactive healthcare interventions, and contribute to overall improvements in patient care and quality of life. The success of implantable devices relies on the careful selection of materials and manufacturing methods. Recent materials research and manufacturing advancements have yielded implantable devices with enhanced biocompatibility, reliability, and functionality, benefiting human healthcare. This paper provides a comprehensive overview of the latest developments in implantable medical devices, emphasizing the importance of material selection and manufacturing methods, including biocompatibility, self-healing capabilities, corrosion resistance, mechanical properties, and conductivity. It explores various manufacturing techniques such as microfabrication, 3D printing, laser micromachining, electrospinning, screen printing, inkjet printing, and nanofabrication. The paper also discusses challenges and limitations in the field, including biocompatibility concerns, privacy and data security issues, and regulatory hurdles for implantable devices.
Collapse
Affiliation(s)
- Akib Abdullah Khan
- School of Engineering and Computer Science, Washington State University, Vancouver, WA, 98686, USA
| | - Jong-Hoon Kim
- School of Engineering and Computer Science, Washington State University, Vancouver, WA, 98686, USA; Department of Mechanical Engineering, University of Washington, WA, 98195, USA.
| |
Collapse
|
7
|
Skopinska-Wisniewska J, Tuszynska M, Kaźmierski Ł, Bartniak M, Bajek A. Gelatin-Sodium Alginate Hydrogels Cross-Linked by Squaric Acid and Dialdehyde Starch as a Potential Bio-Ink. Polymers (Basel) 2024; 16:2560. [PMID: 39339023 PMCID: PMC11435377 DOI: 10.3390/polym16182560] [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: 07/16/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
Hydrogels as biomaterials possess appropriate physicochemical and mechanical properties that enable the formation of a three-dimensional, stable structure used in tissue engineering and 3D printing. The integrity of the hydrogel composition is due to the presence of covalent or noncovalent cross-linking bonds. Using various cross-linking methods and agents is crucial for adjusting the properties of the hydrogel to specific biomedical applications, e.g., for direct bioprinting. The research subject was mixtures of gel-forming polymers: sodium alginate and gelatin. The polymers were cross-linked ionically with the addition of CaCl2 solutions of various concentrations (10%, 5%, 2.5%, and 1%) and covalently using squaric acid (SQ) and dialdehyde starch (DAS). Initially, the polymer mixture's composition and the hydrogel cross-linking procedure were determined. The obtained materials were characterized by mechanical property tests, swelling degree, FTIR, SEM, thermal analysis, and biological research. It was found that the tensile strength of hydrogels cross-linked with 1% and 2.5% CaCl2 solutions was higher than after using a 10% solution (130 kPa and 80 kPa, respectively), and at the same time, the elongation at break increased (to 75%), and the stiffness decreased (Young Modulus is 169 kPa and 104 kPa, respectively). Moreover, lowering the concentration of the CaCl2 solution from 10% to 1% reduced the final material's toxicity. The hydrogels cross-linked with 1% CaCl2 showed lower degradation temperatures and higher weight losses than those cross-linked with 2.5% CaCl2 and therefore were less thermally stable. Additional cross-linking using SQ and DAS had only a minor effect on the strength of the hydrogels, but especially the use of 1% DAS increased the material's elasticity. All tested hydrogels possess a 3D porous structure, with pores of irregular shape and heterogenic size, and their swelling degree initially increased sharply to the value of approx. 1000% during the first 6 h, and finally, it stabilized at a level of 1200-1600% after 24 h. The viscosity of 6% gelatin and 2% alginate solutions with and without cross-linking agents was similar, and they were only slightly shear-thinning. It was concluded that a mixture containing 2% sodium alginate and 6% gelatin presented optimal properties after gel formation and lowering the concentration of the CaCl2 solution to 1% improved the hydrogel's biocompatibility and positively influenced the cross-linking efficiency. Moreover, chemical cross-linking by DAS or SQ additionally improved the final hydrogel's properties and the mixture's printability. In conclusion, among the tested systems, the cross-linking of 6% gelatin-2% alginate mixtures by 1% DAS addition and 1% CaCl2 solution is optimal for tissue engineering applications and potentially suitable for 3D printing.
Collapse
Affiliation(s)
- Joanna Skopinska-Wisniewska
- Chair of Biomaterials and Cosmetics Chemistry, Faculty of Chemistry, Nicolaus Copernicus University in Torun, Gagarina 7 Street, 87-100 Torun, Poland
| | - Marta Tuszynska
- Chair of Biomaterials and Cosmetics Chemistry, Faculty of Chemistry, Nicolaus Copernicus University in Torun, Gagarina 7 Street, 87-100 Torun, Poland
- Department of Tissue Engineering, Chair of Urology and Andrology, Ludwik Rydygier Collegium Medicum in Bydgoszcz Nicolaus Copernicus University in Torun, Karlowicza 24 Street, 85-092 Bydgoszcz, Poland
| | - Łukasz Kaźmierski
- Department of Tissue Engineering, Chair of Urology and Andrology, Ludwik Rydygier Collegium Medicum in Bydgoszcz Nicolaus Copernicus University in Torun, Karlowicza 24 Street, 85-092 Bydgoszcz, Poland
| | - Mateusz Bartniak
- Faculty of Mechanical Engineering, Institute of Materials Science and Engineering, Lodz University of Technology, Stefanowskiego Str. 1/15, 90-537 Lodz, Poland
| | - Anna Bajek
- Department of Oncology, Ludwik Rydygier Collegium Medicum in Bydgoszcz Nicolaus Copernicus University in Torun, Lukasiewicza 1, 85-821 Bydgoszcz, Poland
| |
Collapse
|
8
|
Kim DY, Liu Y, Kim G, An SB, Han I. Innovative Strategies in 3D Bioprinting for Spinal Cord Injury Repair. Int J Mol Sci 2024; 25:9592. [PMID: 39273538 PMCID: PMC11395085 DOI: 10.3390/ijms25179592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
Spinal cord injury (SCI) is a catastrophic condition that disrupts neurons within the spinal cord, leading to severe motor and sensory deficits. While current treatments can alleviate pain, they do not promote neural regeneration or functional recovery. Three-dimensional (3D) bioprinting offers promising solutions for SCI repair by enabling the creation of complex neural tissue constructs. This review provides a comprehensive overview of 3D bioprinting techniques, bioinks, and stem cell applications in SCI repair. Additionally, it highlights recent advancements in 3D bioprinted scaffolds, including the integration of conductive materials, the incorporation of bioactive molecules like neurotrophic factors, drugs, and exosomes, and the design of innovative structures such as multi-channel and axial scaffolds. These innovative strategies in 3D bioprinting can offer a comprehensive approach to optimizing the spinal cord microenvironment, advancing SCI repair. This review highlights a comprehensive understanding of the current state of 3D bioprinting in SCI repair, offering insights into future directions in the field of regenerative medicine.
Collapse
Affiliation(s)
- Daniel Youngsuk Kim
- Research Competency Milestones Program (RECOMP), School of Medicine, CHA University, Seongnam-si 13488, Republic of Korea
- Department of Medicine, School of Medicine, CHA University, Seongnam-si 13496, Republic of Korea
| | - Yanting Liu
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea
| | - Gyubin Kim
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea
| | - Seong Bae An
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea
| | - Inbo Han
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea
| |
Collapse
|
9
|
Yarali E, Mirzaali MJ, Ghalayaniesfahani A, Accardo A, Diaz-Payno PJ, Zadpoor AA. 4D Printing for Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2402301. [PMID: 38580291 DOI: 10.1002/adma.202402301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Indexed: 04/07/2024]
Abstract
4D (bio-)printing endows 3D printed (bio-)materials with multiple functionalities and dynamic properties. 4D printed materials have been recently used in biomedical engineering for the design and fabrication of biomedical devices, such as stents, occluders, microneedles, smart 3D-cell engineered microenvironments, drug delivery systems, wound closures, and implantable medical devices. However, the success of 4D printing relies on the rational design of 4D printed objects, the selection of smart materials, and the availability of appropriate types of external (multi-)stimuli. Here, this work first highlights the different types of smart materials, external stimuli, and design strategies used in 4D (bio-)printing. Then, it presents a critical review of the biomedical applications of 4D printing and discusses the future directions of biomedical research in this exciting area, including in vivo tissue regeneration studies, the implementation of multiple materials with reversible shape memory behaviors, the creation of fast shape-transformation responses, the ability to operate at the microscale, untethered activation and control, and the application of (machine learning-based) modeling approaches to predict the structure-property and design-shape transformation relationships of 4D (bio)printed constructs.
Collapse
Affiliation(s)
- Ebrahim Yarali
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft, 2628 CD, The Netherlands
- Department of Precision and Microsystems Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft, 2628 CD, The Netherlands
| | - Mohammad J Mirzaali
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft, 2628 CD, The Netherlands
| | - Ava Ghalayaniesfahani
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft, 2628 CD, The Netherlands
- Department of Chemistry, Materials and Chemical Engineering, Giulio Natta, Politecnico di Milano, Piazza Leonardo da Vinci, 32, Milano, 20133, Italy
| | - Angelo Accardo
- Department of Precision and Microsystems Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft, 2628 CD, The Netherlands
| | - Pedro J Diaz-Payno
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft, 2628 CD, The Netherlands
- Department of Orthopedics and Sports Medicine, Erasmus MC University Medical Center, Rotterdam, 3015 CN, The Netherlands
| | - Amir A Zadpoor
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft, 2628 CD, The Netherlands
| |
Collapse
|
10
|
Lipari S, Sacco P, Marsich E, Donati I. Silk Fibroin-Enriched Bioink Promotes Cell Proliferation in 3D-Bioprinted Constructs. Gels 2024; 10:469. [PMID: 39057492 PMCID: PMC11275288 DOI: 10.3390/gels10070469] [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: 07/03/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Three-dimensional (3D) bioprinting technology enables the controlled deposition of cells and biomaterials (i.e., bioink) to easily create complex 3D biological microenvironments. Silk fibroin (SF) has recently emerged as a compelling bioink component due to its advantageous mechanical and biological properties. This study reports on the development and optimization of a novel bioink for extrusion-based 3D bioprinting and compares different bioink formulations based on mixtures of alginate methacrylate (ALMA), gelatin and SF. The rheological parameters of the bioink were investigated to predict printability and stability, and the optimal concentration of SF was selected. The bioink containing a low amount of SF (0.002% w/V) was found to be the best formulation. Light-assisted gelation of ALMA was exploited to obtain the final hydrogel matrix. Rheological analyses showed that SF-enriched hydrogels exhibited greater elasticity than SF-free hydrogels and were more tolerant to temperature fluctuations. Finally, MG-63 cells were successfully bioprinted and their viability and proliferation over time were analyzed. The SF-enriched bioink represents an excellent biomaterial in terms of printability and allows high cell proliferation over a period of up to 3 weeks. These data confirm the possibility of using the selected formulation for the successful bioprinting of cells into extracellular matrix-like microenvironments.
Collapse
Affiliation(s)
- Sara Lipari
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri, n.5, I-34127 Trieste, Italy; (S.L.); (P.S.)
| | - Pasquale Sacco
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri, n.5, I-34127 Trieste, Italy; (S.L.); (P.S.)
| | - Eleonora Marsich
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazza dell’Ospitale, n.1, I-34129 Trieste, Italy;
| | - Ivan Donati
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri, n.5, I-34127 Trieste, Italy; (S.L.); (P.S.)
| |
Collapse
|
11
|
Gezek M, Altunbek M, Torres Gouveia ME, Camci-Unal G. 3D Printed Eggshell Microparticle-Laden Thermoplastic Scaffolds for Bone Tissue Engineering. ACS APPLIED MATERIALS & INTERFACES 2024; 16:32957-32970. [PMID: 38885611 DOI: 10.1021/acsami.4c02800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Three-dimensional (3D) printing, an additive manufacturing technique, is increasingly used in the field of tissue engineering. The ability to create complex structures with high precision makes the 3D printing of this material a preferred method for constructing personalized and functional materials. However, the challenge lies in developing affordable and accessible materials with the desired physiochemical and biological properties. In this study, we used eggshell microparticles (ESPs), an example of bioceramic and unconventional biomaterials, to reinforce thermoplastic poly(ε-caprolactone) (PCL) scaffolds via extrusion-based 3D printing. The goal was to conceive a sustainable, affordable, and unique personalized medicine approach. The scaffolds were fabricated with varying concentrations of eggshells, ranging from 0 to 50% (w/w) in the PCL scaffolds. To assess the physicochemical properties, we employed scanning electron microscopy, Fourier-transform infrared spectroscopy, thermogravimetric analysis, differential scanning calorimetry, and X-ray diffraction analysis. Mechanical properties were evaluated through compression testing, and degradation kinetics were studied through accelerated degradation with the remaining mass ranging between 89.4 and 28.3%. In vitro, we evaluated the characteristics of the scaffolds using the MC3T3-E1 preosteoblasts over a 14 day period. In vitro characterization involved the use of the Alamar blue assay, confocal imaging, and real-time quantitative polymerase chain reaction. The results of this study demonstrate the potential of 3D printed biocomposite scaffolds, consisting of thermoplastic PCL reinforced with ESPs, as a promising alternative for bone-graft applications.
Collapse
Affiliation(s)
- Mert Gezek
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Mine Altunbek
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
| | | | - Gulden Camci-Unal
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
- Department of Surgery, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| |
Collapse
|
12
|
Arriola-Alvarez I, Jaunarena I, Izeta A, Lafuente H. Progenitor Cell Sources for 3D Bioprinting of Lymphatic Vessels and Potential Clinical Application. Tissue Eng Part A 2024; 30:353-366. [PMID: 37950710 DOI: 10.1089/ten.tea.2023.0204] [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: 11/13/2023] Open
Abstract
The lymphatic system maintains tissue fluid homeostasis and it is involved in the transport of nutrients and immunosurveillance. It also plays a pivotal role in both pathological and regenerative processes. Lymphatic development in the embryo occurs by polarization and proliferation of lymphatic endothelial cells from the lymph sacs, that is, lymphangiogenesis. Alternatively, lymphvasculogenesis further contributes to the formation of lymphatic vessels. In adult tissues, lymphatic formation rarely occurs under physiological conditions, being restricted to pathological processes. In lymphvasculogenesis, progenitor cells seem to be a source of lymphatic vessels. Indeed, mesenchymal stem cells, adipose stem cells, endothelial progenitor cells, and colony-forming endothelial cells are able to promote lymphatic regeneration by different mechanisms, such as direct differentiation and paracrine effects. In this review, we summarize what is known on the diverse stem/progenitor cell niches available for the lymphatic system, emphasizing the potential that these cells hold for lymphatic tissue engineering through 3D bioprinting and their translation to clinical application.
Collapse
Affiliation(s)
- Inazio Arriola-Alvarez
- Tissue Engineering Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain
| | - Ibon Jaunarena
- Gynecology Oncology Unit, Donostia University Hospital, Donostia-San Sebastián, Spain
- Obstetrics and Gynaecology Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain
- University of the Basque Country (UPV/EHU), Department of Medical Surgical Specialties, Leioa, Spain
| | - Ander Izeta
- Tissue Engineering Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain
- Department of Biomedical Engineering and Sciences, Tecnun-University of Navarra, Donostia-San Sebastián, Spain
| | - Héctor Lafuente
- Tissue Engineering Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain
| |
Collapse
|
13
|
Periz R, Geuß M, Mameka N, Markmann J, Steinhart M. High-Temperature Melt Stamping of Polymers Using Polymer/Nanoporous Gold Composite Stamps. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308478. [PMID: 38263727 DOI: 10.1002/smll.202308478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 01/05/2024] [Indexed: 01/25/2024]
Abstract
Parallel lithographic deposition of polymers onto counterpart substrates is a widely applied surface manufacturing operation. However, polymers may only be soluble in organic solvents or are insoluble at all. Solvent evaporation during stamping may trigger hardly controllable capillarity-driven flow processes or phase separation, and polymer solutions may spread on the counterpart substrates. Solvent-free stamping of melts prevents these drawbacks. Here, a stamp design for the deposition of melts is devised, which intrinsically circumvents ink depletion. The stamps' topographically patterned contact surfaces with protruding contact elements contacting the counterpart substrates consist of a nanoporous gold layer with a thickness of a few micrometers. The nanoporous gold layer is attached to a molten polymer layer, which is support for the nanoporous gold layer and ink reservoir at the same time. The nanoporous gold layer in turn stabilizes the topography of the stamps' contact surfaces. As examples, arrays of submicron microdots of polystyrene and poly(vinylidenefluoride-trifluoroethylene) (PVDF-TrFE) are manufactured. The P(VDF-TrFE) microdots are partially crystalline, ferroelectric, and can be locally poled. It is envisioned that the methodology reported here can be automatized and may be extended to functional low-molecular-mass compounds, such as active pharmaceutical ingredients.
Collapse
Affiliation(s)
- Ruža Periz
- School of Biology and Chemistry and CellNanOs, Universität Osnabrück, Barbarastr. 7, 49076, Osnabrück, Germany
| | - Markus Geuß
- School of Biology and Chemistry and CellNanOs, Universität Osnabrück, Barbarastr. 7, 49076, Osnabrück, Germany
| | - Nadiia Mameka
- Helmholtz-Zentrum Hereon, Institute of Materials Mechanics, 21502, Geesthacht, Germany
| | - Jürgen Markmann
- Helmholtz-Zentrum Hereon, Institute of Materials Mechanics, 21502, Geesthacht, Germany
- Institute of Materials Physics and Technology, Hamburg University of Technology, 21073, Hamburg, Germany
| | - Martin Steinhart
- School of Biology and Chemistry and CellNanOs, Universität Osnabrück, Barbarastr. 7, 49076, Osnabrück, Germany
| |
Collapse
|
14
|
Zhang Q, Yan K, Zheng X, Liu Q, Han Y, Liu Z. Research progress of photo-crosslink hydrogels in ophthalmology: A comprehensive review focus on the applications. Mater Today Bio 2024; 26:101082. [PMID: 38774449 PMCID: PMC11107262 DOI: 10.1016/j.mtbio.2024.101082] [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: 01/27/2024] [Revised: 04/19/2024] [Accepted: 05/03/2024] [Indexed: 05/24/2024] Open
Abstract
Hydrogel presents a three-dimensional polymer network with high water content. Over the past decade, hydrogel has developed from static material to intelligent material with controllable response. Various stimuli are involved in the formation of hydrogel network, among which photo-stimulation has attracted wide attention due to the advantages of controllable conditions, which has a good application prospect in the treatment of ophthalmic diseases. This paper reviews the application of photo-crosslink hydrogels in ophthalmology, focusing on the types of photo-crosslink hydrogels and their applications in ophthalmology, including drug delivery, tissue engineering and 3D printing. In addition, the limitations and future prospects of photo-crosslink hydrogels are also provided.
Collapse
Affiliation(s)
- Qinghe Zhang
- Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang Hunan 421001, China
| | - Ke Yan
- Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang Hunan 421001, China
| | - Xiaoqin Zheng
- Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang Hunan 421001, China
| | - Qiuping Liu
- Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang Hunan 421001, China
| | - Yi Han
- Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang Hunan 421001, China
| | - Zuguo Liu
- Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang Hunan 421001, China
- Xiamen University Affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen Fujian 361005, China
| |
Collapse
|
15
|
Wang F, Song P, Wang J, Wang S, Liu Y, Bai L, Su J. Organoid bioinks: construction and application. Biofabrication 2024; 16:032006. [PMID: 38697093 DOI: 10.1088/1758-5090/ad467c] [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: 11/23/2023] [Accepted: 05/02/2024] [Indexed: 05/04/2024]
Abstract
Organoids have emerged as crucial platforms in tissue engineering and regenerative medicine but confront challenges in faithfully mimicking native tissue structures and functions. Bioprinting technologies offer a significant advancement, especially when combined with organoid bioinks-engineered formulations designed to encapsulate both the architectural and functional elements of specific tissues. This review provides a rigorous, focused examination of the evolution and impact of organoid bioprinting. It emphasizes the role of organoid bioinks that integrate key cellular components and microenvironmental cues to more accurately replicate native tissue complexity. Furthermore, this review anticipates a transformative landscape invigorated by the integration of artificial intelligence with bioprinting techniques. Such fusion promises to refine organoid bioink formulations and optimize bioprinting parameters, thus catalyzing unprecedented advancements in regenerative medicine. In summary, this review accentuates the pivotal role and transformative potential of organoid bioinks and bioprinting in advancing regenerative therapies, deepening our understanding of organ development, and clarifying disease mechanisms.
Collapse
Affiliation(s)
- Fuxiao Wang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
- These authors contributed equally
| | - Peiran Song
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
- These authors contributed equally
| | - Jian Wang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
- These authors contributed equally
| | - Sicheng Wang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai 200444, People's Republic of China
| | - Yuanyuan Liu
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, People's Republic of China
| | - Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
- Wenzhou Institute of Shanghai University, Wenzhou 325000, People's Republic of China
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
| |
Collapse
|
16
|
Wei Q, An Y, Zhao X, Li M, Zhang J. Three-dimensional bioprinting of tissue-engineered skin: Biomaterials, fabrication techniques, challenging difficulties, and future directions: A review. Int J Biol Macromol 2024; 266:131281. [PMID: 38641503 DOI: 10.1016/j.ijbiomac.2024.131281] [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: 12/31/2023] [Revised: 03/17/2024] [Accepted: 03/29/2024] [Indexed: 04/21/2024]
Abstract
As an emerging new manufacturing technology, Three-dimensional (3D) bioprinting provides the potential for the biomimetic construction of multifaceted and intricate architectures of functional integument, particularly functional biomimetic dermal structures inclusive of cutaneous appendages. Although the tissue-engineered skin with complete biological activity and physiological functions is still cannot be manufactured, it is believed that with the advances in matrix materials, molding process, and biotechnology, a new generation of physiologically active skin will be born in the future. In pursuit of furnishing readers and researchers involved in relevant research to have a systematic and comprehensive understanding of 3D printed tissue-engineered skin, this paper furnishes an exegesis on the prevailing research landscape, formidable obstacles, and forthcoming trajectories within the sphere of tissue-engineered skin, including: (1) the prevalent biomaterials (collagen, chitosan, agarose, alginate, etc.) routinely employed in tissue-engineered skin, and a discerning analysis and comparison of their respective merits, demerits, and inherent characteristics; (2) the underlying principles and distinguishing attributes of various current printing methodologies utilized in tissue-engineered skin fabrication; (3) the present research status and progression in the realm of tissue-engineered biomimetic skin; (4) meticulous scrutiny and summation of the extant research underpinning tissue-engineered skin inform the identification of prevailing challenges and issues.
Collapse
Affiliation(s)
- Qinghua Wei
- School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, China; Innovation Center NPU Chongqing, Northwestern Polytechnical University, Chongqing 400000, China.
| | - Yalong An
- School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Xudong Zhao
- School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Mingyang Li
- School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Juan Zhang
- School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
17
|
Jiu J, Liu H, Li D, Li J, Liu L, Yang W, Yan L, Li S, Zhang J, Li X, Li JJ, Wang B. 3D bioprinting approaches for spinal cord injury repair. Biofabrication 2024; 16:032003. [PMID: 38569491 DOI: 10.1088/1758-5090/ad3a13] [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: 10/10/2023] [Accepted: 04/03/2024] [Indexed: 04/05/2024]
Abstract
Regenerative healing of spinal cord injury (SCI) poses an ongoing medical challenge by causing persistent neurological impairment and a significant socioeconomic burden. The complexity of spinal cord tissue presents hurdles to successful regeneration following injury, due to the difficulty of forming a biomimetic structure that faithfully replicates native tissue using conventional tissue engineering scaffolds. 3D bioprinting is a rapidly evolving technology with unmatched potential to create 3D biological tissues with complicated and hierarchical structure and composition. With the addition of biological additives such as cells and biomolecules, 3D bioprinting can fabricate preclinical implants, tissue or organ-like constructs, andin vitromodels through precise control over the deposition of biomaterials and other building blocks. This review highlights the characteristics and advantages of 3D bioprinting for scaffold fabrication to enable SCI repair, including bottom-up manufacturing, mechanical customization, and spatial heterogeneity. This review also critically discusses the impact of various fabrication parameters on the efficacy of spinal cord repair using 3D bioprinted scaffolds, including the choice of printing method, scaffold shape, biomaterials, and biological supplements such as cells and growth factors. High-quality preclinical studies are required to accelerate the translation of 3D bioprinting into clinical practice for spinal cord repair. Meanwhile, other technological advances will continue to improve the regenerative capability of bioprinted scaffolds, such as the incorporation of nanoscale biological particles and the development of 4D printing.
Collapse
Affiliation(s)
- Jingwei Jiu
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, People's Republic of China
| | - Haifeng Liu
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, People's Republic of China
| | - Dijun Li
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, People's Republic of China
| | - Jiarong Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Lu Liu
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Wenjie Yang
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Lei Yan
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, People's Republic of China
| | - Songyan Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jing Zhang
- Department of Emergency Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, People's Republic of China
| | - Xiaoke Li
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, People's Republic of China
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Bin Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| |
Collapse
|
18
|
Dadashzadeh A, Moghassemi S, Amorim CA. Bioprinting of a Liposomal Oxygen-Releasing Scaffold for Ovary Tissue Engineering. Tissue Eng Part A 2024. [PMID: 38534964 DOI: 10.1089/ten.tea.2024.0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024] Open
Abstract
This study addresses a critical challenge in bioprinting for regenerative medicine, specifically the issue of hypoxia compromising cell viability in engineered tissues. To overcome this hurdle, a novel approach using a microfluidic bioprinter is used to create a two-layer structure resembling the human ovary. This structure incorporates a liposomal oxygen-releasing system to enhance cell viability. The bioprinting technique enables the simultaneous extrusion of two distinct bioinks, namely, bioink A (comprising alginate 1% and 5 mg/mL PEGylated fibrinogen in a 20:1 molar ratio) and bioink B (containing alginate 0.5%). In addition, liposomal catalase and hydrogen peroxide (H2O2) are synthesized and incorporated into bioinks A and B, respectively. The liposomes are prepared using thin film hydration with a monodisperse size (140-160 nm) and high encapsulation efficiency. To assess construct functionality, isolated human ovarian cells are added to bioink A. The bioprinted constructs, with or without liposomal oxygen-releasing systems, are cultured under hypoxic and normoxic conditions for 3 days. Live/Dead assay results demonstrate that liposomal oxygen-releasing systems effectively preserve cell viability in hypoxic conditions, resembling viability under normoxic conditions without liposomes. PrestoBlue assay reveals significantly higher mitochondrial activity in constructs with liposomal oxygen delivery systems under both hypoxic and normoxic conditions. The evaluation of apoptosis status through annexin V immunostaining shows that liposomal oxygen-releasing scaffolds successfully protect cells from hypoxic stress, exhibiting a proportion of apoptotic cells similar to normoxic conditions. In contrast, constructs lacking liposomes in hypoxic conditions exhibit a higher incidence of cells in early-stage apoptosis. In conclusion, the study demonstrates the promising potential of bioprinted oxygen-releasing liposomal scaffolds to protect ovarian stromal cells in hypoxic environments. These innovative scaffolds not only offer protection but also recapitulate the mechanical differences between the medulla and the cortex in the normal ovary structure. This opens new avenues for advanced ovary tissue engineering and transplantation strategies.
Collapse
Affiliation(s)
- Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Christiani A Amorim
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
19
|
Shibru MG, Ali ZM, Almansoori AS, Paunovic J, Pantic IV, Corridon PR. Slaughterhouse waste: a unique and sustainable source for dECM-based bioinks. Regen Med 2024; 19:113-118. [PMID: 38356397 DOI: 10.2217/rme-2023-0194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024] Open
Affiliation(s)
- Meklit G Shibru
- Department of Biomedical Engineering & Biotechnology, College of Medicine & Health Sciences, Khalifa University of Science & Technology, Abu Dhabi, United Arab Emirates
| | - Zehara M Ali
- Department of Biomedical Engineering & Biotechnology, College of Medicine & Health Sciences, Khalifa University of Science & Technology, Abu Dhabi, United Arab Emirates
| | - Aliyaa S Almansoori
- Department of Biomedical Engineering & Biotechnology, College of Medicine & Health Sciences, Khalifa University of Science & Technology, Abu Dhabi, United Arab Emirates
| | - Jovana Paunovic
- University of Belgrade, Faculty of Medicine, Department of Pathophysiology, Dr. Subotica 9, RS-11129, Belgrade, Serbia
| | - Igor V Pantic
- University of Belgrade, Faculty of Medicine, Department of Medical Physiology, Laboratory for Cellular Physiology, Visegradska 26/II, RS-11129, Belgrade, Serbia
- University of Haifa, 199 Abba Hushi Blvd, Mount Carmel, Haifa, IL, 3498838, Israel
- Department of Pharmacology, College of Medicine & Health Sciences, Khalifa University of Science & Technology
| | - Peter R Corridon
- Department of Biomedical Engineering & Biotechnology, College of Medicine & Health Sciences, Khalifa University of Science & Technology, Abu Dhabi, United Arab Emirates
- Biomedical Engineering & Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi, United Arab Emirates
- Center for Biotechnology, Khalifa University of Science & Technology, Abu Dhabi, United Arab Emirates
| |
Collapse
|
20
|
Aazmi A, Zhang D, Mazzaglia C, Yu M, Wang Z, Yang H, Huang YYS, Ma L. Biofabrication methods for reconstructing extracellular matrix mimetics. Bioact Mater 2024; 31:475-496. [PMID: 37719085 PMCID: PMC10500422 DOI: 10.1016/j.bioactmat.2023.08.018] [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: 05/09/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/19/2023] Open
Abstract
In the human body, almost all cells interact with extracellular matrices (ECMs), which have tissue and organ-specific compositions and architectures. These ECMs not only function as cellular scaffolds, providing structural support, but also play a crucial role in dynamically regulating various cellular functions. This comprehensive review delves into the examination of biofabrication strategies used to develop bioactive materials that accurately mimic one or more biophysical and biochemical properties of ECMs. We discuss the potential integration of these ECM-mimics into a range of physiological and pathological in vitro models, enhancing our understanding of cellular behavior and tissue organization. Lastly, we propose future research directions for ECM-mimics in the context of tissue engineering and organ-on-a-chip applications, offering potential advancements in therapeutic approaches and improved patient outcomes.
Collapse
Affiliation(s)
- Abdellah Aazmi
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Duo Zhang
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 51817, China
| | - Corrado Mazzaglia
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Mengfei Yu
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Zhen Wang
- Center for Laboratory Medicine, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Huayong Yang
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Yan Yan Shery Huang
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Liang Ma
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
21
|
Zhang S, Xu G, Wu J, Liu X, Fan Y, Chen J, Wallace G, Gu Q. Microphysiological Constructs and Systems: Biofabrication Tactics, Biomimetic Evaluation Approaches, and Biomedical Applications. SMALL METHODS 2024; 8:e2300685. [PMID: 37798902 DOI: 10.1002/smtd.202300685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/23/2023] [Indexed: 10/07/2023]
Abstract
In recent decades, microphysiological constructs and systems (MPCs and MPSs) have undergone significant development, ranging from self-organized organoids to high-throughput organ-on-a-chip platforms. Advances in biomaterials, bioinks, 3D bioprinting, micro/nanofabrication, and sensor technologies have contributed to diverse and innovative biofabrication tactics. MPCs and MPSs, particularly tissue chips relevant to absorption, distribution, metabolism, excretion, and toxicity, have demonstrated potential as precise, efficient, and economical alternatives to animal models for drug discovery and personalized medicine. However, current approaches mainly focus on the in vitro recapitulation of the human anatomical structure and physiological-biochemical indices at a single or a few simple levels. This review highlights the recent remarkable progress in MPC and MPS models and their applications. The challenges that must be addressed to assess the reliability, quantify the techniques, and utilize the fidelity of the models are also discussed.
Collapse
Affiliation(s)
- Shuyu Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine/Department of Fetal Medicine and Prenatal Diagnosis/BioResource Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Guoshi Xu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 100049, China
| | - Juan Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 100049, China
| | - Xiao Liu
- Department of Gastroenterology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yong Fan
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine/Department of Fetal Medicine and Prenatal Diagnosis/BioResource Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Jun Chen
- Intelligent Polymer Research Institute, Australian Institute for Innovative Materials, Innovation Campus, University of Wollongong, North Wollongong, NSW, 2500, Australia
| | - Gordon Wallace
- Intelligent Polymer Research Institute, Australian Institute for Innovative Materials, Innovation Campus, University of Wollongong, North Wollongong, NSW, 2500, Australia
| | - Qi Gu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 100049, China
| |
Collapse
|
22
|
Wang XH, Liu N, Zhang H, Yin ZS, Zha ZG. From cells to organs: progress and potential in cartilaginous organoids research. J Transl Med 2023; 21:926. [PMID: 38129833 PMCID: PMC10740223 DOI: 10.1186/s12967-023-04591-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/04/2023] [Indexed: 12/23/2023] Open
Abstract
While cartilage tissue engineering has significantly improved the speed and quality of cartilage regeneration, the underlying metabolic mechanisms are complex, making research in this area lengthy and challenging. In the past decade, organoids have evolved rapidly as valuable research tools. Methods to create these advanced human cell models range from simple tissue culture techniques to complex bioengineering approaches. Cartilaginous organoids in part mimic the microphysiology of human cartilage and fill a gap in high-fidelity cartilage disease models to a certain extent. They hold great promise to elucidate the pathogenic mechanism of a diversity of cartilage diseases and prove crucial in the development of new drugs. This review will focus on the research progress of cartilaginous organoids and propose strategies for cartilaginous organoid construction, study directions, and future perspectives.
Collapse
Affiliation(s)
- Xiao-He Wang
- Department of Bone and Joint Surgery, the First Affliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Ning Liu
- Department of Bone and Joint Surgery, the First Affliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Hui Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Zong-Sheng Yin
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Zhen-Gang Zha
- Department of Bone and Joint Surgery, the First Affliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
23
|
Barrulas RV, Corvo MC. Rheology in Product Development: An Insight into 3D Printing of Hydrogels and Aerogels. Gels 2023; 9:986. [PMID: 38131974 PMCID: PMC10742728 DOI: 10.3390/gels9120986] [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: 11/25/2023] [Revised: 12/09/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023] Open
Abstract
Rheological characterisation plays a crucial role in developing and optimising advanced materials in the form of hydrogels and aerogels, especially if 3D printing technologies are involved. Applications ranging from tissue engineering to environmental remediation require the fine-tuning of such properties. Nonetheless, their complex rheological behaviour presents unique challenges in additive manufacturing. This review outlines the vital rheological parameters that influence the printability of hydrogel and aerogel inks, emphasising the importance of viscosity, yield stress, and viscoelasticity. Furthermore, the article discusses the latest developments in rheological modifiers and printing techniques that enable precise control over material deposition and resolution in 3D printing. By understanding and manipulating the rheological properties of these materials, researchers can explore new possibilities for applications such as biomedicine or nanotechnology. An optimal 3D printing ink requires strong shear-thinning behaviour for smooth extrusion, forming continuous filaments. Favourable thixotropic properties aid viscosity recovery post-printing, and adequate yield stress and G' are crucial for structural integrity, preventing deformation or collapse in printed objects, and ensuring high-fidelity preservation of shapes. This insight into rheology provides tools for the future of material design and manufacturing in the rapidly evolving field of 3D printing of hydrogels and aerogels.
Collapse
Affiliation(s)
| | - Marta C. Corvo
- i3N|Cenimat, Department of Materials Science (DCM), NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal;
| |
Collapse
|
24
|
Sabzevari A, Rayat Pisheh H, Ansari M, Salati A. Progress in bioprinting technology for tissue regeneration. J Artif Organs 2023; 26:255-274. [PMID: 37119315 DOI: 10.1007/s10047-023-01394-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 04/09/2023] [Indexed: 05/01/2023]
Abstract
In recent years, due to the increase in diseases that require organ/tissue transplantation and the limited donor, on the other hand, patients have lost hope of recovery and organ transplantation. Regenerative medicine is one of the new sciences that promises a bright future for these patients by providing solutions to repair, improve function, and replace tissue. One of the technologies used in regenerative medicine is three-dimensional (3D) bioprinters. Bioprinting is a new strategy that is the basis for starting a global revolution in the field of medical sciences and has attracted much attention. 3D bioprinters use a combination of advanced biology and cell science, computer science, and materials science to create complex bio-hybrid structures for various applications. The capacity to use this technology can be demonstrated in regenerative medicine to make various connective tissues, such as skin, cartilage, and bone. One of the essential parts of a 3D bioprinter is the bio-ink. Bio-ink is a combination of biologically active molecules, cells, and biomaterials that make the printed product. In this review, we examine the main bioprinting strategies, such as inkjet printing, laser, and extrusion-based bioprinting, as well as some of their applications.
Collapse
Affiliation(s)
- Alireza Sabzevari
- Department of Biomedical Engineering, Meybod University, Meybod, Iran
| | | | - Mojtaba Ansari
- Department of Biomedical Engineering, Meybod University, Meybod, Iran.
| | - Amir Salati
- Tissue Engineering and Applied Cell Sciences Group, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
25
|
Shah DD, Raghani NR, Chorawala MR, Singh S, Prajapati BG. Harnessing three-dimensional (3D) cell culture models for pulmonary infections: State of the art and future directions. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2861-2880. [PMID: 37266588 PMCID: PMC10235844 DOI: 10.1007/s00210-023-02541-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/17/2023] [Indexed: 06/03/2023]
Abstract
Pulmonary infections have been a leading etiology of morbidity and mortality worldwide. Upper and lower respiratory tract infections have multifactorial causes, which include bacterial, viral, and rarely, fungal infections. Moreover, the recent emergence of SARS-CoV-2 has created havoc and imposes a huge healthcare burden. Drug and vaccine development against these pulmonary pathogens like respiratory syncytial virus, SARS-CoV-2, Mycobacteria, etc., requires a systematic set of tools for research and investigation. Currently, in vitro 2D cell culture models are widely used to emulate the in vivo physiologic environment. Although this approach holds a reasonable promise over pre-clinical animal models, it lacks the much-needed correlation to the in vivo tissue architecture, cellular organization, cell-to-cell interactions, downstream processes, and the biomechanical milieu. In view of these inadequacies, 3D cell culture models have recently acquired interest. Mammalian embryonic and induced pluripotent stem cells may display their remarkable self-organizing abilities in 3D culture, and the resulting organoids replicate important structural and functional characteristics of organs such the kidney, lung, gut, brain, and retina. 3D models range from scaffold-free systems to scaffold-based and hybrid models as well. Upsurge in organs-on-chip models for pulmonary conditions has anticipated encouraging results. Complexity and dexterity of developing 3D culture models and the lack of standardized working procedures are a few of the setbacks, which are expected to be overcome in the coming times. Herein, we have elaborated the significance and types of 3D cell culture models for scrutinizing pulmonary infections, along with the in vitro techniques, their applications, and additional systems under investigation.
Collapse
Affiliation(s)
- Disha D Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Neha R Raghani
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Sudarshan Singh
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200, Thailand.
| | - Bhupendra G Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva, 384012, India.
| |
Collapse
|
26
|
Teli P, Kale V, Vaidya A. Beyond animal models: revolutionizing neurodegenerative disease modeling using 3D in vitro organoids, microfluidic chips, and bioprinting. Cell Tissue Res 2023; 394:75-91. [PMID: 37572163 DOI: 10.1007/s00441-023-03821-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 07/27/2023] [Indexed: 08/14/2023]
Abstract
Neurodegenerative diseases (NDs) are characterized by uncontrolled loss of neuronal cells leading to a progressive deterioration of brain functions. The transition rate of numerous neuroprotective drugs against Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and Huntington's disease, leading to FDA approval, is only 8-14% in the last two decades. Thus, in spite of encouraging preclinical results, these drugs have failed in human clinical trials, demonstrating that traditional cell cultures and animal models cannot accurately replicate human pathophysiology. Hence, in vitro three-dimensional (3D) models have been developed to bridge the gap between human and animal studies. Such technological advancements in 3D culture systems, such as human-induced pluripotent stem cell (iPSC)-derived cells/organoids, organ-on-a-chip technique, and 3D bioprinting, have aided our understanding of the pathophysiology and underlying mechanisms of human NDs. Despite these recent advances, we still lack a 3D model that recapitulates all the key aspects of NDs, thus making it difficult to study the ND's etiology in-depth. Hence in this review, we propose developing a combinatorial approach that allows the integration of patient-derived iPSCs/organoids with 3D bioprinting and organ-on-a-chip technique as it would encompass the neuronal cells along with their niche. Such a 3D combinatorial approach would characterize pathological processes thoroughly, making them better suited for high-throughput drug screening and developing effective novel therapeutics targeting NDs.
Collapse
Affiliation(s)
- Prajakta Teli
- Symbiosis International (Deemed University), Symbiosis School of Biological Sciences, Pune, 412115, India
- Symbiosis International (Deemed University), Symbiosis Center for Stem Cell Research, Pune, 412115, India
| | - Vaijayanti Kale
- Symbiosis International (Deemed University), Symbiosis School of Biological Sciences, Pune, 412115, India
- Symbiosis International (Deemed University), Symbiosis Center for Stem Cell Research, Pune, 412115, India
| | - Anuradha Vaidya
- Symbiosis International (Deemed University), Symbiosis School of Biological Sciences, Pune, 412115, India.
- Symbiosis International (Deemed University), Symbiosis Center for Stem Cell Research, Pune, 412115, India.
| |
Collapse
|
27
|
Hachimi Alaoui C, Réthoré G, Weiss P, Fatimi A. Sustainable Biomass Lignin-Based Hydrogels: A Review on Properties, Formulation, and Biomedical Applications. Int J Mol Sci 2023; 24:13493. [PMID: 37686299 PMCID: PMC10487582 DOI: 10.3390/ijms241713493] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/27/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Different techniques have been developed to overcome the recalcitrant nature of lignocellulosic biomass and extract lignin biopolymer. Lignin has gained considerable interest owing to its attractive properties. These properties may be more beneficial when including lignin in the preparation of highly desired value-added products, including hydrogels. Lignin biopolymer, as one of the three major components of lignocellulosic biomaterials, has attracted significant interest in the biomedical field due to its biocompatibility, biodegradability, and antioxidant and antimicrobial activities. Its valorization by developing new hydrogels has increased in recent years. Furthermore, lignin-based hydrogels have shown great potential for various biomedical applications, and their copolymerization with other polymers and biopolymers further expands their possibilities. In this regard, lignin-based hydrogels can be synthesized by a variety of methods, including but not limited to interpenetrating polymer networks and polymerization, crosslinking copolymerization, crosslinking grafted lignin and monomers, atom transfer radical polymerization, and reversible addition-fragmentation transfer polymerization. As an example, the crosslinking mechanism of lignin-chitosan-poly(vinyl alcohol) (PVA) hydrogel involves active groups of lignin such as hydroxyl, carboxyl, and sulfonic groups that can form hydrogen bonds (with groups in the chemical structures of chitosan and/or PVA) and ionic bonds (with groups in the chemical structures of chitosan and/or PVA). The aim of this review paper is to provide a comprehensive overview of lignin-based hydrogels and their applications, focusing on the preparation and properties of lignin-based hydrogels and the biomedical applications of these hydrogels. In addition, we explore their potential in wound healing, drug delivery systems, and 3D bioprinting, showcasing the unique properties of lignin-based hydrogels that enable their successful utilization in these areas. Finally, we discuss future trends in the field and draw conclusions based on the findings presented.
Collapse
Affiliation(s)
- Chaymaa Hachimi Alaoui
- Chemical Science and Engineering Research Team (ERSIC), FPBM, Sultan Moulay Slimane University, Mghila, P.O. Box 592, Beni Mellal 23000, Morocco;
- Nantes Université, Oniris, Univ Angers, INSERM, Regenerative Medicine and Skeleton, RmeS, UMR 1229, F-44000 Nantes, France
| | - Gildas Réthoré
- Nantes Université, Oniris, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RmeS, UMR 1229, F-44000 Nantes, France; (G.R.); (P.W.)
| | - Pierre Weiss
- Nantes Université, Oniris, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RmeS, UMR 1229, F-44000 Nantes, France; (G.R.); (P.W.)
| | - Ahmed Fatimi
- Chemical Science and Engineering Research Team (ERSIC), FPBM, Sultan Moulay Slimane University, Mghila, P.O. Box 592, Beni Mellal 23000, Morocco;
| |
Collapse
|
28
|
Doganay MT, Chelliah CJ, Tozluyurt A, Hujer AM, Obaro SK, Gurkan U, Patel R, Bonomo RA, Draz M. 3D Printed Materials for Combating Antimicrobial Resistance. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2023; 67:371-398. [PMID: 37790286 PMCID: PMC10545363 DOI: 10.1016/j.mattod.2023.05.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Three-dimensional (3D) printing is a rapidly growing technology with a significant capacity for translational applications in both biology and medicine. 3D-printed living and non-living materials are being widely tested as a potential replacement for conventional solutions for testing and combating antimicrobial resistance (AMR). The precise control of cells and their microenvironment, while simulating the complexity and dynamics of an in vivo environment, provides an excellent opportunity to advance the modeling and treatment of challenging infections and other health conditions. 3D-printing models the complicated niches of microbes and host-pathogen interactions, and most importantly, how microbes develop resistance to antibiotics. In addition, 3D-printed materials can be applied to testing and delivering antibiotics. Here, we provide an overview of 3D printed materials and biosystems and their biomedical applications, focusing on ever increasing AMR. Recent applications of 3D printing to alleviate the impact of AMR, including developed bioprinted systems, targeted bacterial infections, and tested antibiotics are presented.
Collapse
Affiliation(s)
- Mert Tunca Doganay
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Cyril John Chelliah
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Abdullah Tozluyurt
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Andrea M Hujer
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Research Service, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | | | - Umut Gurkan
- Mechanical and Aerospace Engineering Department, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology and Division of Public Health, Infectious Diseases, and Occupational medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Robert A Bonomo
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Research Service, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
- Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- CWRU-Cleveland VAMC Center for Antimicrobial Resistance and Epidemiology (Case VA CARES) Cleveland, OH, USA
| | - Mohamed Draz
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
- Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH 44106, USA
| |
Collapse
|
29
|
Alizadeh Sardroud H, Chen X, Eames BF. Reinforcement of Hydrogels with a 3D-Printed Polycaprolactone (PCL) Structure Enhances Cell Numbers and Cartilage ECM Production under Compression. J Funct Biomater 2023; 14:313. [PMID: 37367278 DOI: 10.3390/jfb14060313] [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: 04/30/2023] [Revised: 05/30/2023] [Accepted: 06/04/2023] [Indexed: 06/28/2023] Open
Abstract
Hydrogels show promise in cartilage tissue engineering (CTE) by supporting chondrocytes and maintaining their phenotype and extracellular matrix (ECM) production. Under prolonged mechanical forces, however, hydrogels can be structurally unstable, leading to cell and ECM loss. Furthermore, long periods of mechanical loading might alter the production of cartilage ECM molecules, including glycosaminoglycans (GAGs) and collagen type 2 (Col2), specifically with the negative effect of stimulating fibrocartilage, typified by collagen type 1 (Col1) secretion. Reinforcing hydrogels with 3D-printed Polycaprolactone (PCL) structures offer a solution to enhance the structural integrity and mechanical response of impregnated chondrocytes. This study aimed to assess the impact of compression duration and PCL reinforcement on the performance of chondrocytes impregnated with hydrogel. Results showed that shorter loading periods did not significantly affect cell numbers and ECM production in 3D-bioprinted hydrogels, but longer periods tended to reduce cell numbers and ECM compared to unloaded conditions. PCL reinforcement enhanced cell numbers under mechanical compression compared to unreinforced hydrogels. However, the reinforced constructs seemed to produce more fibrocartilage-like, Col1-positive ECM. These findings suggest that reinforced hydrogel constructs hold potential for in vivo cartilage regeneration and defect treatment by retaining higher cell numbers and ECM content. To further enhance hyaline cartilage ECM formation, future studies should focus on adjusting the mechanical properties of reinforced constructs and exploring mechanotransduction pathways.
Collapse
Affiliation(s)
- Hamed Alizadeh Sardroud
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| | - B Frank Eames
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| |
Collapse
|
30
|
Kim J. Characterization of Biocompatibility of Functional Bioinks for 3D Bioprinting. Bioengineering (Basel) 2023; 10:bioengineering10040457. [PMID: 37106644 PMCID: PMC10135811 DOI: 10.3390/bioengineering10040457] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/02/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Three-dimensional (3D) bioprinting with suitable bioinks has become a critical tool for fabricating 3D biomimetic complex structures mimicking physiological functions. While enormous efforts have been devoted to developing functional bioinks for 3D bioprinting, widely accepted bioinks have not yet been developed because they have to fulfill stringent requirements such as biocompatibility and printability simultaneously. To further advance our knowledge of the biocompatibility of bioinks, this review presents the evolving concept of the biocompatibility of bioinks and standardization efforts for biocompatibility characterization. This work also briefly reviews recent methodological advances in image analyses to characterize the biocompatibility of bioinks with regard to cell viability and cell-material interactions within 3D constructs. Finally, this review highlights a number of updated contemporary characterization technologies and future perspectives to further advance our understanding of the biocompatibility of functional bioinks for successful 3D bioprinting.
Collapse
Affiliation(s)
- Jinku Kim
- Department of Biological and Chemical Engineering, Hongik University, Sejong 30016, Republic of Korea
| |
Collapse
|
31
|
Borden LK, Gargava A, Kokilepersaud UJ, Raghavan SR. Universal Way to "Glue" Capsules and Gels into 3D Structures by Electroadhesion. ACS APPLIED MATERIALS & INTERFACES 2023; 15:17070-17077. [PMID: 36961991 DOI: 10.1021/acsami.2c20793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
We demonstrate the use of electroadhesion (EA), i.e., adhesion induced by an electric field, to connect a variety of soft materials into 3D structures. EA requires a cationic and an anionic material, but these can be of diverse origin, including covalently cross-linked hydrogels made by polymerizing charged monomers or physical gels/capsules formed by the ionic cross-linking of biopolymers (e.g., alginate and chitosan). Between each cationic/anionic pair, EA is induced rapidly (in ∼10 s) by low voltages (∼10 V DC)─and the adhesion is permanent after the field is turned off. The adhesion is strong enough to allow millimeter-scale capsules/gels to be assembled in 3D into robust structures such as capsule-capsule chains, capsule arrays on a base gel, and a 3D cube of capsules. EA-based assembly of spherical building blocks can be done more precisely, rapidly, and easily than by any alternative techniques. Moreover, the adhesion can be reversed (by switching the polarity of the field)─hence any errors during assembly can be undone and fixed. EA can also be used for selective sorting of charged soft matter─for example, a 'finger robot' can selectively 'pick up' capsules of the opposite charge by EA and subsequently 'drop off' these structures by reversing the polarity. Overall, our work shows how electric fields can be used to connect soft matter without the need for an adhesive or glue.
Collapse
Affiliation(s)
- Leah K Borden
- Department of Chemical & Biomolecular Engineering, University of Maryland, College Park, Maryland 20742, United States
| | - Ankit Gargava
- Department of Chemical & Biomolecular Engineering, University of Maryland, College Park, Maryland 20742, United States
| | - Uma J Kokilepersaud
- Department of Chemical & Biomolecular Engineering, University of Maryland, College Park, Maryland 20742, United States
| | - Srinivasa R Raghavan
- Department of Chemical & Biomolecular Engineering, University of Maryland, College Park, Maryland 20742, United States
| |
Collapse
|
32
|
Abuarqoub D, Theeb LS, Omari MB, Hamadneh YI, Alrawabdeh JA, Aslam N, Jafar H, Awidi A. The Osteogenic Role of Biomaterials Combined with Human-Derived Dental Stem Cells in Bone Tissue Regeneration. Tissue Eng Regen Med 2023; 20:251-270. [PMID: 36808303 PMCID: PMC10070593 DOI: 10.1007/s13770-022-00514-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 02/23/2023] Open
Abstract
The use of stem cells in regenerative medicine had great potential for clinical applications. However, cell delivery strategies have critical importance in stimulating the differentiation of stem cells and enhancing their potential to regenerate damaged tissues. Different strategies have been used to investigate the osteogenic potential of dental stem cells in conjunction with biomaterials through in vitro and in vivo studies. Osteogenesis has a broad implication in regenerative medicine, particularly for maxillofacial defects. This review summarizes some of the most recent developments in the field of tissue engineering using dental stem cells.
Collapse
Affiliation(s)
- Duaa Abuarqoub
- Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan.
- Cell Therapy Center, The University of Jordan, Amman, Jordan.
| | - Laith S Theeb
- School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Mohammad B Omari
- School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Yazan I Hamadneh
- School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | | | - Nazneen Aslam
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Hanan Jafar
- Cell Therapy Center, The University of Jordan, Amman, Jordan
- School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Abdalla Awidi
- Cell Therapy Center, The University of Jordan, Amman, Jordan.
- School of Medicine, The University of Jordan, Amman, 11942, Jordan.
| |
Collapse
|
33
|
D’Amora U, Ronca A, Scialla S, Soriente A, Manini P, Phua JW, Ottenheim C, Pezzella A, Calabrese G, Raucci MG, Ambrosio L. Bioactive Composite Methacrylated Gellan Gum for 3D-Printed Bone Tissue-Engineered Scaffolds. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:772. [PMID: 36839140 PMCID: PMC9963483 DOI: 10.3390/nano13040772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 06/18/2023]
Abstract
Gellan gum (GG) was chemically modified with methacrylic moieties to produce a photocrosslinkable biomaterial ink, hereinafter called methacrylated GG (GGMA), with improved physico-chemical properties, mechanical behavior and stability under physiological conditions. Afterwards, GGMA was functionalized by incorporating two different bioactive compounds, a naturally derived eumelanin extracted from the black soldier fly (BSF-Eumel), or hydroxyapatite nanoparticles (HAp), synthesized by the sol-gel method. Different ink formulations based on GGMA (2 and 4% (w/v)), BSF-Eumel, at a selected concentration (0.3125 mg/mL), or HAp (10 and 30% wHAp/wGGMA) were developed and processed by three-dimensional (3D) printing. All the functionalized GGMA-based ink formulations allowed obtaining 3D-printed GGMA-based scaffolds with a well-organized structure. For both bioactive signals, the scaffolds with the highest GGMA concentration (4% (w/v)) and the highest percentage of infill (45%) showed the best performances in terms of morphological and mechanical properties. Indeed, these scaffolds showed a good structural integrity over 28 days. Given the presence of negatively charged groups along the eumelanin backbone, scaffolds consisting of GGMA/BSF-Eumel demonstrated a higher stability. From a mechanical point of view, GGMA/BSF-Eumel scaffolds exhibited values of storage modulus similar to those of GGMA ones, while the inclusion of HAp at 30% (wHAp/wGGMA) led to a storage modulus of 32.5 kPa, 3.5-fold greater than neat GGMA. In vitro studies proved the capability of the bioactivated 3D-printed scaffolds to support 7F2 osteoblast cell growth and differentiation. BSF-Eumel and HAp triggered a different time-dependent physiological response in the osteoblasts. Specifically, while the ink with BSF-Eumel acted as a stimulus towards cell proliferation, reaching the highest value at 14 days, a higher expression of alkaline phosphatase activity was detected for scaffolds consisting of GGMA and HAp. The overall findings demonstrated the possible use of these biomaterial inks for 3D-printed bone tissue-engineered scaffolds.
Collapse
Affiliation(s)
- Ugo D’Amora
- Institute of Polymers, Composites and Biomaterials, National Research Council, 80125 Naples, Italy
| | - Alfredo Ronca
- Institute of Polymers, Composites and Biomaterials, National Research Council, 80125 Naples, Italy
| | - Stefania Scialla
- Institute of Polymers, Composites and Biomaterials, National Research Council, 80125 Naples, Italy
| | - Alessandra Soriente
- Institute of Polymers, Composites and Biomaterials, National Research Council, 80125 Naples, Italy
| | - Paola Manini
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
- Bioelectronics Task Force, University of Naples Federico II, 80126 Naples, Italy
| | - Jun Wei Phua
- Insectta, 60 Jalan Penjara, Singapore 149375, Singapore
| | | | - Alessandro Pezzella
- Institute of Polymers, Composites and Biomaterials, National Research Council, 80125 Naples, Italy
- Bioelectronics Task Force, University of Naples Federico II, 80126 Naples, Italy
- Department of Physics “E. Pancini”, University of Naples Federico II, 80126 Naples, Italy
| | - Giovanna Calabrese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy
| | - Maria Grazia Raucci
- Institute of Polymers, Composites and Biomaterials, National Research Council, 80125 Naples, Italy
- Bioelectronics Task Force, University of Naples Federico II, 80126 Naples, Italy
| | - Luigi Ambrosio
- Institute of Polymers, Composites and Biomaterials, National Research Council, 80125 Naples, Italy
| |
Collapse
|
34
|
Altunbek M, Afghah F, Caliskan OS, Yoo JJ, Koc B. Design and bioprinting for tissue interfaces. Biofabrication 2023; 15. [PMID: 36716498 DOI: 10.1088/1758-5090/acb73d] [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: 08/04/2022] [Accepted: 01/30/2023] [Indexed: 02/01/2023]
Abstract
Tissue interfaces include complex gradient structures formed by transitioning of biochemical and mechanical properties in micro-scale. This characteristic allows the communication and synchronistic functioning of two adjacent but distinct tissues. It is particularly challenging to restore the function of these complex structures by transplantation of scaffolds exclusively produced by conventional tissue engineering methods. Three-dimensional (3D) bioprinting technology has opened an unprecedented approach for precise and graded patterning of chemical, biological and mechanical cues in a single construct mimicking natural tissue interfaces. This paper reviews and highlights biochemical and biomechanical design for 3D bioprinting of various tissue interfaces, including cartilage-bone, muscle-tendon, tendon/ligament-bone, skin, and neuro-vascular/muscular interfaces. Future directions and translational challenges are also provided at the end of the paper.
Collapse
Affiliation(s)
- Mine Altunbek
- Sabanci Nanotechnology Research and Application Center, Istanbul 34956, Turkey.,Sabanci University Faculty of Engineering and Natural Sciences, Istanbul 34956, Turkey
| | - Ferdows Afghah
- Sabanci Nanotechnology Research and Application Center, Istanbul 34956, Turkey.,Sabanci University Faculty of Engineering and Natural Sciences, Istanbul 34956, Turkey
| | - Ozum Sehnaz Caliskan
- Sabanci Nanotechnology Research and Application Center, Istanbul 34956, Turkey.,Sabanci University Faculty of Engineering and Natural Sciences, Istanbul 34956, Turkey
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, NC 27157, United States of America
| | - Bahattin Koc
- Sabanci Nanotechnology Research and Application Center, Istanbul 34956, Turkey.,Sabanci University Faculty of Engineering and Natural Sciences, Istanbul 34956, Turkey
| |
Collapse
|
35
|
Brancato V. 3D Bioprinting for Cancer Models. Cancer Nanotechnol 2023. [DOI: 10.1007/978-3-031-17831-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
36
|
Sithole MN, Mndlovu H, du Toit LC, Choonara YE. Advances in Stimuli-responsive Hydrogels for Tissue Engineering and Regenerative Medicine Applications: A Review Towards Improving Structural Design for 3D Printing. Curr Pharm Des 2023; 29:3187-3205. [PMID: 37779402 DOI: 10.2174/0113816128246888230920060802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/17/2023] [Accepted: 07/14/2023] [Indexed: 10/03/2023]
Abstract
The physicochemical properties of polymeric hydrogels render them attractive for the development of 3D printed prototypes for tissue engineering in regenerative medicine. Significant effort has been made to design hydrogels with desirable attributes that facilitate 3D printability. In addition, there is significant interest in exploring stimuli-responsive hydrogels to support automated 3D printing into more structurally organised prototypes such as customizable bio-scaffolds for regenerative medicine applications. Synthesizing stimuli-responsive hydrogels is dependent on the type of design and modulation of various polymeric materials to open novel opportunities for applications in biomedicine and bio-engineering. In this review, the salient advances made in the design of stimuli-responsive polymeric hydrogels for 3D printing in tissue engineering are discussed with a specific focus on the different methods of manipulation to develop 3D printed stimuli-responsive polymeric hydrogels. Polymeric functionalisation, nano-enabling and crosslinking are amongst the most common manipulative attributes that affect the assembly and structure of 3D printed bio-scaffolds and their stimuli- responsiveness. The review also provides a concise incursion into the various applications of stimuli to enhance the automated production of structurally organized 3D printed medical prototypes.
Collapse
Affiliation(s)
- Mduduzi Nkosinathi Sithole
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, Gauteng, 2193, South Africa
| | - Hillary Mndlovu
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, Gauteng, 2193, South Africa
| | - Lisa C du Toit
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, Gauteng, 2193, South Africa
| | - Yahya Essop Choonara
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, Gauteng, 2193, South Africa
| |
Collapse
|
37
|
Augustine R, Gezek M, Seray Bostanci N, Nguyen A, Camci-Unal G. Oxygen-Generating Scaffolds: One Step Closer to the Clinical Translation of Tissue Engineered Products. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2023; 455:140783. [PMID: 36644784 PMCID: PMC9835968 DOI: 10.1016/j.cej.2022.140783] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The lack of oxygen supply in engineered constructs has been an ongoing challenge for tissue engineering and regenerative medicine. Upon implantation of an engineered tissue, spontaneous blood vessel formation does not happen rapidly, therefore, there is typically a limited availability of oxygen in engineered biomaterials. Providing oxygen in large tissue-engineered constructs is a major challenge that hinders the development of clinically relevant engineered tissues. Similarly, maintaining adequate oxygen levels in cell-laden tissue engineered products during transportation and storage is another hurdle. There is an unmet demand for functional scaffolds that could actively produce and deliver oxygen, attainable by incorporating oxygen-generating materials. Recent approaches include encapsulation of oxygen-generating agents such as solid peroxides, liquid peroxides, and fluorinated substances in the scaffolds. Recent approaches to mitigate the adverse effects, as well as achieving a sustained and controlled release of oxygen, are discussed. Importance of oxygen-generating materials in various tissue engineering approaches such as ex vivo tissue engineering, in situ tissue engineering, and bioprinting are highlighted in detail. In addition, the existing challenges, possible solutions, and future strategies that aim to design clinically relevant multifunctional oxygen-generating biomaterials are provided in this review paper.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Mert Gezek
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Nazli Seray Bostanci
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Angelina Nguyen
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Gulden Camci-Unal
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, United States
- Department of Surgery, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| |
Collapse
|
38
|
Bhushan S, Singh S, Maiti TK, Sharma C, Dutt D, Sharma S, Li C, Tag Eldin EM. Scaffold Fabrication Techniques of Biomaterials for Bone Tissue Engineering: A Critical Review. Bioengineering (Basel) 2022; 9:728. [PMID: 36550933 PMCID: PMC9774188 DOI: 10.3390/bioengineering9120728] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 11/27/2022] Open
Abstract
Bone tissue engineering (BTE) is a promising alternative to repair bone defects using biomaterial scaffolds, cells, and growth factors to attain satisfactory outcomes. This review targets the fabrication of bone scaffolds, such as the conventional and electrohydrodynamic techniques, for the treatment of bone defects as an alternative to autograft, allograft, and xenograft sources. Additionally, the modern approaches to fabricating bone constructs by additive manufacturing, injection molding, microsphere-based sintering, and 4D printing techniques, providing a favorable environment for bone regeneration, function, and viability, are thoroughly discussed. The polymers used, fabrication methods, advantages, and limitations in bone tissue engineering application are also emphasized. This review also provides a future outlook regarding the potential of BTE as well as its possibilities in clinical trials.
Collapse
Affiliation(s)
- Sakchi Bhushan
- Department of Paper Technology, IIT Roorkee, Saharanpur 247001, India
| | - Sandhya Singh
- Department of Paper Technology, IIT Roorkee, Saharanpur 247001, India
| | - Tushar Kanti Maiti
- Department of Polymer and Process Engineering, IIT Roorkee, Saharanpur 247001, India
| | - Chhavi Sharma
- Department of Polymer and Process Engineering, IIT Roorkee, Saharanpur 247001, India
| | - Dharm Dutt
- Department of Paper Technology, IIT Roorkee, Saharanpur 247001, India
| | - Shubham Sharma
- Mechanical Engineering Department, University Center for Research & Development, Chandigarh University, Mohali 140413, India
- School of Mechanical and Automotive Engineering, Qingdao University of Technology, Qingdao 266520, China
| | - Changhe Li
- School of Mechanical and Automotive Engineering, Qingdao University of Technology, Qingdao 266520, China
| | | |
Collapse
|
39
|
Expanding Quality by Design Principles to Support 3D Printed Medical Device Development Following the Renewed Regulatory Framework in Europe. Biomedicines 2022; 10:biomedicines10112947. [PMID: 36428514 PMCID: PMC9687721 DOI: 10.3390/biomedicines10112947] [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: 10/20/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022] Open
Abstract
The vast scope of 3D printing has ignited the production of tailored medical device (MD) development and catalyzed a paradigm shift in the health-care industry, particularly following the COVID pandemic. This review aims to provide an update on the current progress and emerging opportunities for additive manufacturing following the introduction of the new medical device regulation (MDR) within the EU. The advent of early-phase implementation of the Quality by Design (QbD) quality management framework in MD development is a focal point. The application of a regulatory supported QbD concept will ensure successful MD development, as well as pointing out the current challenges of 3D bioprinting. Utilizing a QbD scientific and risk-management approach ensures the acceleration of MD development in a more targeted way by building in all stakeholders' expectations, namely those of the patients, the biomedical industry, and regulatory bodies.
Collapse
|
40
|
Cruz-Maya I, Guarino V. 3D Scaffolds Fabrication via Bicomponent Microgels Assembly: Process Optimization and In Vitro Characterization. MICROMACHINES 2022; 13:1726. [PMID: 36296078 PMCID: PMC9607065 DOI: 10.3390/mi13101726] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 06/16/2023]
Abstract
In the last decade, different technological approaches have been proposed for the fabrication of 3D models suitable to evaluate in vitro cell response. Among them, electro fluid dynamic atomization (EFDA) belonging to the family of electro-assisted technologies allows for the dropping of polysaccharides and/or proteins solutions to produce micro-scaled hydrogels or microgels with the peculiar features of hydrogel-like materials (i.e., biocompatibility, wettability, swelling). In this work, a method to fabricate 3D scaffolds by the assembly of bicomponent microgels made of sodium alginate and gelatin was proposed. As first step, optical and scanning electron microscopy with the support of image analysis enabled to explore the basic properties of single blocks in terms of correlation between particle morphology and process parameters (i.e., voltage, flow rate, electrode gap, and needle diameter). Chemical analysis via ninhydrin essays and FTIR analysis confirmed the presence of gelatin, mostly retained by physical interactions into the alginate network mediated by electrostatic forces. In vitro tests confirmed the effect of biochemical signals exerted by the protein on the biological response of hMSCs cultured onto the microgels surface. Hence, it is concluded that alginate/gelatin microgels assemblies can efficiently work as 3D scaffolds able to support in vitro cells functions, thus providing a friendly microenvironment to investigate in vitro cell interactions.
Collapse
|
41
|
Singh G, Singh S, Kumar R, Parkash C, Pruncu C, Ramakrishna S. Tissues and organ printing: An evolution of technology and materials. Proc Inst Mech Eng H 2022; 236:1695-1710. [DOI: 10.1177/09544119221125084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Since its beginnings, three-dimensional printing (3DP) technology has been successful because of ongoing advances in operating principles, the range of materials and cost-saving measures. However, the 3DP technological progressions in the biomedical sector have majorly taken place in the last decade after the evolution of novel 3DP systems, generally categorised as bioprinters and biomaterials to provide a replacement, transplantation or regeneration of the damaged organs and tissue constructs of the human body. There is now substantial scientific literature accessible to support the benefits of digital healthcare procedures with the help of bioprinters. It is of the highest significance to know the fundamental principles of the available printers and the compatibility of biomaterials as their feedstock, notwithstanding the huge potential of bioprinting systems to manufacture organs and other human body components. This paper provides a precise and helpful reading of the different categories of bioprinters, suitable biomaterials, numerical simulations and modelling and examples of much acknowledged clinical practices. The paper will also cite the prominent issues that still have not received desired solutions. Overall, the article will be of great use for all the professionals, scholars and engineers concerned with the 3DP, bioprinting and biomaterials.
Collapse
Affiliation(s)
- Gurminder Singh
- Department of Mechanical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Sunpreet Singh
- Department of Mechanical Engineering, National University of Singapore, Singapore, Singapore
- Mechanical Engineering Department, Chandigarh University, Punjab
| | - Raman Kumar
- Mechanical Engineering, Guru Nanak Dev Engineering College, Ludhiana, Punjab, India
| | - Chander Parkash
- School of Mechanical Engineering, Lovely Professional University, Phagwara, Punjab, India
| | - Catalin Pruncu
- Departimento di Meccanica, Matematica e Management, Politecnico di Bari, 70125 Bari, Italy
| | - Seeram Ramakrishna
- Department of Mechanical Engineering, National University of Singapore, Singapore, Singapore
| |
Collapse
|
42
|
Mirzaali MJ, Moosabeiki V, Rajaai SM, Zhou J, Zadpoor AA. Additive Manufacturing of Biomaterials-Design Principles and Their Implementation. MATERIALS (BASEL, SWITZERLAND) 2022; 15:5457. [PMID: 35955393 PMCID: PMC9369548 DOI: 10.3390/ma15155457] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 05/04/2023]
Abstract
Additive manufacturing (AM, also known as 3D printing) is an advanced manufacturing technique that has enabled progress in the design and fabrication of customised or patient-specific (meta-)biomaterials and biomedical devices (e.g., implants, prosthetics, and orthotics) with complex internal microstructures and tuneable properties. In the past few decades, several design guidelines have been proposed for creating porous lattice structures, particularly for biomedical applications. Meanwhile, the capabilities of AM to fabricate a wide range of biomaterials, including metals and their alloys, polymers, and ceramics, have been exploited, offering unprecedented benefits to medical professionals and patients alike. In this review article, we provide an overview of the design principles that have been developed and used for the AM of biomaterials as well as those dealing with three major categories of biomaterials, i.e., metals (and their alloys), polymers, and ceramics. The design strategies can be categorised as: library-based design, topology optimisation, bio-inspired design, and meta-biomaterials. Recent developments related to the biomedical applications and fabrication methods of AM aimed at enhancing the quality of final 3D-printed biomaterials and improving their physical, mechanical, and biological characteristics are also highlighted. Finally, examples of 3D-printed biomaterials with tuned properties and functionalities are presented.
Collapse
Affiliation(s)
- Mohammad J. Mirzaali
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Mekelweg 2, 2628 CD Delft, The Netherlands
| | | | | | | | | |
Collapse
|
43
|
Moya-Garcia CR, Okuyama H, Sadeghi N, Li J, Tabrizian M, Li-Jessen NYK. In vitro models for head and neck cancer: Current status and future perspective. Front Oncol 2022; 12:960340. [PMID: 35992863 PMCID: PMC9381731 DOI: 10.3389/fonc.2022.960340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 06/29/2022] [Indexed: 12/12/2022] Open
Abstract
The 5-year overall survival rate remains approximately 50% for head and neck (H&N) cancer patients, even though new cancer drugs have been approved for clinical use since 2016. Cancer drug studies are now moving toward the use of three-dimensional culture models for better emulating the unique tumor microenvironment (TME) and better predicting in vivo response to cancer treatments. Distinctive TME features, such as tumor geometry, heterogenous cellularity, and hypoxic cues, notably affect tissue aggressiveness and drug resistance. However, these features have not been fully incorporated into in vitro H&N cancer models. This review paper aims to provide a scholarly assessment of the designs, contributions, and limitations of in vitro models in H&N cancer drug research. We first review the TME features of H&N cancer that are most relevant to in vitro drug evaluation. We then evaluate a selection of advanced culture models, namely, spheroids, organotypic models, and microfluidic chips, in their applications for H&N cancer drug research. Lastly, we propose future opportunities of in vitro H&N cancer research in the prospects of high-throughput drug screening and patient-specific drug evaluation.
Collapse
Affiliation(s)
| | - Hideaki Okuyama
- School of Communication Sciences and Disorders, McGill University, Montreal, QC, Canada
- Department of Otolaryngology – Head & Neck Surgery, Kyoto University, Kyoto, Japan
| | - Nader Sadeghi
- Department of Otolaryngology – Head and Neck Surgery, McGill University, Montreal, QC, Canada
- Research Institute of McGill University Health Center, McGill University, Montreal, QC, Canada
| | - Jianyu Li
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
- Department of Mechanical Engineering, McGill University, Montreal, QC, Canada
| | - Maryam Tabrizian
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
- *Correspondence: Maryam Tabrizian, ; Nicole Y. K. Li-Jessen,
| | - Nicole Y. K. Li-Jessen
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
- School of Communication Sciences and Disorders, McGill University, Montreal, QC, Canada
- Department of Otolaryngology – Head and Neck Surgery, McGill University, Montreal, QC, Canada
- Research Institute of McGill University Health Center, McGill University, Montreal, QC, Canada
- *Correspondence: Maryam Tabrizian, ; Nicole Y. K. Li-Jessen,
| |
Collapse
|
44
|
|
45
|
Lee YJ, Lee JS, Ajiteru O, Lee OJ, Lee JS, Lee H, Kim SW, Park JW, Kim KY, Choi KY, Hong H, Sultan T, Kim SH, Park CH. Biocompatible fluorescent silk fibroin bioink for digital light processing 3D printing. Int J Biol Macromol 2022; 213:317-327. [PMID: 35605719 DOI: 10.1016/j.ijbiomac.2022.05.123] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 11/28/2022]
Abstract
Chemically modified silk fibroin (SF) bioink has been used for three-dimensional (3D) bioprinting in tissue engineering because of its biocompatibility and printability. Also, fluorescent silk fibroin (FSF) from transgenic silkworms has been recently applied in biomedicine because of its fluorescence property. However, the fabrication of fluorescent hydrogel from FSF has not been elucidated. In this study, we showed the fabrication of a digital light processing (DLP) printable bioink from a chemically modified FSF. This bioink was fabricated by covalent conjugation of FSF and glycidyl methacrylate (GMA) and can be printed into various structures, such as the brain, ear, hand, lung, and internal organs. The physical properties of glycidyl methacrylated fluorescent silk fibroin (FSGMA) hydrogel was like the glycidyl methacrylated non-fluorescent silk fibroin (SGMA) hydrogel. The FSGMA hydrogel significantly retains its fluorescence property and has excellent biocompatibility. All these properties make FSGMA hydrogel a potent tool in encapsulated cell tracking and observing the scaffolds' degradation in vivo. This study suggested that our 3D DLP printable FSF bioink could play a promising role in the biomedical field.
Collapse
Affiliation(s)
- Young Jin Lee
- Nano-Bio Regenerative Medical Institute (NBRM), Hallym University, Chuncheon 24252, Republic of Korea
| | - Joong Seob Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea
| | - Olatunji Ajiteru
- Nano-Bio Regenerative Medical Institute (NBRM), Hallym University, Chuncheon 24252, Republic of Korea
| | - Ok Joo Lee
- Nano-Bio Regenerative Medical Institute (NBRM), Hallym University, Chuncheon 24252, Republic of Korea
| | - Ji Seung Lee
- Nano-Bio Regenerative Medical Institute (NBRM), Hallym University, Chuncheon 24252, Republic of Korea
| | - Hanna Lee
- Nano-Bio Regenerative Medical Institute (NBRM), Hallym University, Chuncheon 24252, Republic of Korea
| | - Seong Wan Kim
- Department of Agricultural Biology, National Institute of Agricultural Sciences, Rural Development Administration, Wanju, Jeollabuk-do 55365, Republic of Korea
| | - Jong Woo Park
- Department of Agricultural Biology, National Institute of Agricultural Sciences, Rural Development Administration, Wanju, Jeollabuk-do 55365, Republic of Korea
| | - Kee Young Kim
- Department of Agricultural Biology, National Institute of Agricultural Sciences, Rural Development Administration, Wanju, Jeollabuk-do 55365, Republic of Korea
| | - Kyu Young Choi
- Depratment of Otorhinolaryngology-Head and Neck Surgery, Kangnam Sacred Heart Hospital, Seoul 07441, Republic of Korea
| | - Heesun Hong
- Nano-Bio Regenerative Medical Institute (NBRM), Hallym University, Chuncheon 24252, Republic of Korea
| | - Tipu Sultan
- Nano-Bio Regenerative Medical Institute (NBRM), Hallym University, Chuncheon 24252, Republic of Korea
| | - Soon Hee Kim
- Nano-Bio Regenerative Medical Institute (NBRM), Hallym University, Chuncheon 24252, Republic of Korea
| | - Chan Hum Park
- Nano-Bio Regenerative Medical Institute (NBRM), Hallym University, Chuncheon 24252, Republic of Korea; Depratment of Otorhinolaryngology-Head and Neck Surgery, Chuncheon Sacred Heart Hospital, Chuncheon 24253, Republic of Korea.
| |
Collapse
|
46
|
Panda S, Hajra S, Mistewicz K, Nowacki B, In-Na P, Krushynska A, Mishra YK, Kim HJ. A focused review on three-dimensional bioprinting technology for artificial organ fabrication. Biomater Sci 2022; 10:5054-5080. [PMID: 35876134 DOI: 10.1039/d2bm00797e] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Three-dimensional (3D) bioprinting technology has attracted a great deal of interest because it can be easily adapted to many industries and research sectors, such as biomedical, manufacturing, education, and engineering. Specifically, 3D bioprinting has provided significant advances in the medical industry, since such technology has led to significant breakthroughs in the synthesis of biomaterials, cells, and accompanying elements to produce composite living tissues. 3D bioprinting technology could lead to the immense capability of replacing damaged or injured tissues or organs with newly dispensed cell biomaterials and functional tissues. Several types of bioprinting technology and different bio-inks can be used to replicate cells and generate supporting units as complex 3D living tissues. Bioprinting techniques have undergone great advancements in the field of regenerative medicine to provide 3D printed models for numerous artificial organs and transplantable tissues. This review paper aims to provide an overview of 3D-bioprinting technologies by elucidating the current advancements, recent progress, opportunities, and applications in this field. It highlights the most recent advancements in 3D-bioprinting technology, particularly in the area of artificial organ development and cancer research. Additionally, the paper speculates on the future progress in 3D-bioprinting as a versatile foundation for several biomedical applications.
Collapse
Affiliation(s)
- Swati Panda
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology, Daegu-42988, South Korea.
| | - Sugato Hajra
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology, Daegu-42988, South Korea.
| | - Krystian Mistewicz
- Institute of Physics - Center for Science and Education, Silesian University of Technology, Krasińskiego 8, Katowice, Poland
| | - Bartłomiej Nowacki
- Faculty of Materials Engineering, Silesian University of Technology, Krasińskiego 8, Katowice, Poland
| | - Pichaya In-Na
- Department of Chemical Technology, Faculty of Science, Chulalongkorn University, 254 Phyathai Road, Wangmai, Pathumwan, Bangkok-10330, Thailand
| | - Anastasiia Krushynska
- Engineering and Technology Institute Groningen (ENTEG), Faculty of Science and Engineering, University of Groningen, Nijenborgh 4, Groningen, 9747 AG, Netherlands
| | - Yogendra Kumar Mishra
- Mads Clausen Institute, NanoSYD, University of Southern Denmark, Alsion 2, 6400 Sønderborg, Denmark
| | - Hoe Joon Kim
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology, Daegu-42988, South Korea. .,Robotics and Mechatronics Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu-42988, South Korea
| |
Collapse
|
47
|
Shi E, Lou L, Warburton L, Rubinsky B. 3D Printing in Combined Cartesian and Curvilinear Coordinates. J Med Device 2022. [DOI: 10.1115/1.4055064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Abstract
A 3D printing technology that facilitates continuous printing along a combination of cartesian and curvilinear coordinates, designed for in vivo and in situ bioprinting is introduced. The combined cartesian/curvilinear printing head motion is accomplished by attaching a biomimetic, flexible, "tendon cable" soft robot arm to a conventional cartesian three axis 3D printing carousel. This allows printing along a combination of cartesian and curvilinear coordinates using five independent stepper motors controlled by an Arduino Uno with each motor requiring a microstep driver powered via a 12V power supply. Three of the independent motors control the printing head motion along conventional cartesian coordinates while two of the independent motors control the length of each pair of the four "tendon cables" which in turn controls the radius of curvature and the angle displacement of the soft printer head along two orthogonal planes. This combination imparts motion along six independent degrees of freedom in cartesian and curvilinear coordinates. The design of the system is described together with experimental results which demonstrate that this design can print continuously along curved and inclined surfaces while avoiding the "staircase" effect, which is typical of conventional three axis 3D printing along curvilinear surfaces.
Collapse
Affiliation(s)
- Edward Shi
- Department of Mechanical Engineering, University of California Berkeley; Department of Mechanical Engineering, University of Colorado , Boulder
| | - Leo Lou
- Department of Bioengineering, University of California Berkeley
| | - Linnea Warburton
- Department of Mechanical Engineering, University of California Berkeley
| | - Boris Rubinsky
- Department of Mechanical Engineering, University of California Berkeley; Department of Bioengineering, University of California Berkeley
| |
Collapse
|
48
|
Teixeira MC, Lameirinhas NS, Carvalho JPF, Silvestre AJD, Vilela C, Freire CSR. A Guide to Polysaccharide-Based Hydrogel Bioinks for 3D Bioprinting Applications. Int J Mol Sci 2022; 23:6564. [PMID: 35743006 PMCID: PMC9223682 DOI: 10.3390/ijms23126564] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 02/04/2023] Open
Abstract
Three-dimensional (3D) bioprinting is an innovative technology in the biomedical field, allowing the fabrication of living constructs through an approach of layer-by-layer deposition of cell-laden inks, the so-called bioinks. An ideal bioink should possess proper mechanical, rheological, chemical, and biological characteristics to ensure high cell viability and the production of tissue constructs with dimensional stability and shape fidelity. Among the several types of bioinks, hydrogels are extremely appealing as they have many similarities with the extracellular matrix, providing a highly hydrated environment for cell proliferation and tunability in terms of mechanical and rheological properties. Hydrogels derived from natural polymers, and polysaccharides, in particular, are an excellent platform to mimic the extracellular matrix, given their low cytotoxicity, high hydrophilicity, and diversity of structures. In fact, polysaccharide-based hydrogels are trendy materials for 3D bioprinting since they are abundant and combine adequate physicochemical and biomimetic features for the development of novel bioinks. Thus, this review portrays the most relevant advances in polysaccharide-based hydrogel bioinks for 3D bioprinting, focusing on the last five years, with emphasis on their properties, advantages, and limitations, considering polysaccharide families classified according to their source, namely from seaweed, higher plants, microbial, and animal (particularly crustaceans) origin.
Collapse
Affiliation(s)
| | | | | | | | | | - Carmen S. R. Freire
- CICECO—Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (M.C.T.); (N.S.L.); (J.P.F.C.); (A.J.D.S.); (C.V.)
| |
Collapse
|
49
|
Yang Z, Yi P, Liu Z, Zhang W, Mei L, Feng C, Tu C, Li Z. Stem Cell-Laden Hydrogel-Based 3D Bioprinting for Bone and Cartilage Tissue Engineering. Front Bioeng Biotechnol 2022; 10:865770. [PMID: 35656197 PMCID: PMC9152119 DOI: 10.3389/fbioe.2022.865770] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 04/18/2022] [Indexed: 12/30/2022] Open
Abstract
Tremendous advances in tissue engineering and regenerative medicine have revealed the potential of fabricating biomaterials to solve the dilemma of bone and articular defects by promoting osteochondral and cartilage regeneration. Three-dimensional (3D) bioprinting is an innovative fabrication technology to precisely distribute the cell-laden bioink for the construction of artificial tissues, demonstrating great prospect in bone and joint construction areas. With well controllable printability, biocompatibility, biodegradability, and mechanical properties, hydrogels have been emerging as an attractive 3D bioprinting material, which provides a favorable biomimetic microenvironment for cell adhesion, orientation, migration, proliferation, and differentiation. Stem cell-based therapy has been known as a promising approach in regenerative medicine; however, limitations arise from the uncontrollable proliferation, migration, and differentiation of the stem cells and fortunately could be improved after stem cells were encapsulated in the hydrogel. In this review, our focus was centered on the characterization and application of stem cell-laden hydrogel-based 3D bioprinting for bone and cartilage tissue engineering. We not only highlighted the effect of various kinds of hydrogels, stem cells, inorganic particles, and growth factors on chondrogenesis and osteogenesis but also outlined the relationship between biophysical properties like biocompatibility, biodegradability, osteoinductivity, and the regeneration of bone and cartilage. This study was invented to discuss the challenge we have been encountering, the recent progress we have achieved, and the future perspective we have proposed for in this field.
Collapse
Affiliation(s)
- Zhimin Yang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ping Yi
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
| | - Zhongyue Liu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wenchao Zhang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lin Mei
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chengyao Feng
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chao Tu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhihong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
50
|
Ronzoni FL, Aliberti F, Scocozza F, Benedetti L, Auricchio F, Sampaolesi M, Cusella G, Redwan IN, Ceccarelli G, Conti M. Myoblast 3D bioprinting to burst in vitro skeletal muscle differentiation. J Tissue Eng Regen Med 2022; 16:484-495. [PMID: 35246958 PMCID: PMC9311434 DOI: 10.1002/term.3293] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/28/2022] [Accepted: 02/17/2022] [Indexed: 12/22/2022]
Abstract
Skeletal muscle regeneration is one of the major areas of interest in sport medicine as well as trauma centers. Three-dimensional (3D) bioprinting (BioP) is nowadays widely adopted to manufacture 3D constructs for regenerative medicine but a comparison between the available biomaterial-based inks (bioinks) is missing. The present study aims to assess the impact of different hydrogels on the viability, proliferation, and differentiation of murine myoblasts (C2C12) encapsulated in 3D bioprinted constructs aided to muscle regeneration. We tested three different commercially available hydrogels bioinks based on: (1) gelatin methacrylate and alginate crosslinked by UV light; (2) gelatin methacrylate, xanthan gum, and alginate-fibrinogen; (3) nanofibrillated cellulose (NFC)/alginate-fibrinogen crosslinked with calcium chloride and thrombin. Constructs embedding the cells were manufactured by extrusion-based BioP and C2C12 viability, proliferation, and differentiation were assessed after 24 h, 7, 14, 21, and 28 days in culture. Although viability, proliferation, and differentiation were observed in all the constructs, among the investigated bioinks, the best results were obtained by using NFC/alginate-fibrinogen-based hydrogel from 7 to 14 days in culture, when the embedded myoblasts started fusing, forming at day 21 and day 28 multinucleated myotubes within the 3D bioprinted structures. The results revealed an extensive myotube alignment all over the linear structure of the hydrogel, demonstrating cell maturation, and enhanced myogenesis. The bioprinting strategies that we describe here denote a strong and endorsed approach for the creation of in vitro artificial muscle to improve skeletal muscle tissue engineering for future therapeutic applications.
Collapse
Affiliation(s)
- Flavio L. Ronzoni
- Department of Public Health, Experimental and Forensic MedicineHuman Anatomy UnitUniversity of PaviaPaviaItaly
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly
| | - Flaminia Aliberti
- Department of Public Health, Experimental and Forensic MedicineHuman Anatomy UnitUniversity of PaviaPaviaItaly
- Fondazione IRCCS Policlinico San MatteoCenter for Inherited Cardiovascular DiseasesTransplant Research AreaPaviaItaly
| | - Franca Scocozza
- Department of Civil EngineeringUniversity of PaviaPaviaItaly
| | - Laura Benedetti
- Department of Public Health, Experimental and Forensic MedicineHuman Anatomy UnitUniversity of PaviaPaviaItaly
| | | | - Maurilio Sampaolesi
- Department of Public Health, Experimental and Forensic MedicineHuman Anatomy UnitUniversity of PaviaPaviaItaly
- Department of Development and RegenerationTranslational CardiomyologyKU LeuvenLeuvenBelgium
| | - Gabriella Cusella
- Department of Public Health, Experimental and Forensic MedicineHuman Anatomy UnitUniversity of PaviaPaviaItaly
| | | | - Gabriele Ceccarelli
- Department of Public Health, Experimental and Forensic MedicineHuman Anatomy UnitUniversity of PaviaPaviaItaly
| | - Michele Conti
- Department of Civil EngineeringUniversity of PaviaPaviaItaly
| |
Collapse
|