Next Article in Journal
An Adaptive Neuro-Fuzzy Inference Model to Predict Punching Shear Strength of Flat Concrete Slabs
Next Article in Special Issue
Nano-Structured Demineralized Human Dentin Matrix to Enhance Bone and Dental Repair and Regeneration
Previous Article in Journal
A Novel CAD Tool for Electric Educational Diagrams
Previous Article in Special Issue
Nanomaterials in Craniofacial Tissue Regeneration: A Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Adoption of Three-Dimensional Additive Manufacturing from Biomedical Material Design to 3D Organ Printing

1
Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Strasse 8-10, 10589 Berlin, Germany
2
Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569 Stuttgart, Germany
3
Institut für Biomaterialien und biomolekulare Systeme Universität Stuttgart, Pfaffenwaldring 57, D-70550 Stuttgart, Germany
4
CSIR-Indian Institute of Toxicology Research, 31 Vishvigyan Bhawan, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001, India
5
Department of Biotechnology, Savitribai Phule Pune University, Pune 411007, India
*
Author to whom correspondence should be addressed.
Appl. Sci. 2019, 9(4), 811; https://doi.org/10.3390/app9040811
Submission received: 4 February 2019 / Revised: 19 February 2019 / Accepted: 20 February 2019 / Published: 25 February 2019
(This article belongs to the Special Issue Nanomaterials for Bone Tissue Engineering)

Abstract

:
Three-dimensional (3D) bioprinting promises to change future lifestyle and the way we think about aging, the field of medicine, and the way clinicians treat ailing patients. In this brief review, we attempt to give a glimpse into how recent developments in 3D bioprinting are going to impact vast research ranging from complex and functional organ transplant to future toxicology studies and printed organ-like 3D spheroids. The techniques were successfully applied to reconstructed complex 3D functional tissue for implantation, application-based high-throughput (HTP) platforms for absorption, distribution, metabolism, and excretion (ADME) profiling to understand the cellular basis of toxicity. We also provide an overview of merits/demerits of various bioprinting techniques and the physicochemical basis of bioink for tissue engineering. We briefly discuss the importance of universal bioink technology, and of time as the fourth dimension. Some examples of bioprinted tissue are shown, followed by a brief discussion on future biomedical applications.

1. Introduction

Human organs are highly specialized tissue structures performing particular distinctive functions. In the case of dysfunctional organs, clinical treatments are often limited by a scarcity of available donors and by immune rejection of donated tissue [1]. To overcome the lack of available transplantable organs, tissue-engineering approaches are used, which face some challenges [2]. Borrowing the concept of three-dimensional (3D) printing from additive manufacturing technologies, whereby a digital design for a 3D structure is fabricated layer by layer following the bottom-up approach, 3D bioprinting is now being pursued as a potential solution to some of the challenges faced in tissue-engineering methods [3,4,5,6,7,8]. Layer-by-layer precise positioning of biological materials, and biochemical and living cells, with spatial control of the placement of functional components, is used to fabricate 3D tissue structures [9].
A typical bioprinting process consists of three major steps, namely pre-processing, processing, and post-processing (Figure 1). Pre-processing involves imaging of the tissue or organ and the reconstruction of 3D models from the imaging (Figure 1a–d). Multidetector computed tomography (MDCT) is widely used for rapid prototyping because of its simpler image-processing requirements. Cone-beam computed tomography (CBCT), positron emission tomography (PET), single-photon emission computed tomography (SPECT), magnetic resonance imaging (MRI), and ultrasonography (US) are other non-invasive imaging modalities [10]. The processing step involves the bioprinting process using an appropriate bioink [11,12,13,14] (Figure 1e–g). The bioprinting process can be classified into four different categories, including laser-based bioprinting [15,16,17,18,19,20,21,22,23], droplet-based bioprinting [24,25,26,27,28,29,30,31], extrusion-based bioprinting [28,29,32,33,34,35,36,37,38,39,40,41,42], and stereolithography-based bioprinting [13,43,44,45,46,47,48,49,50]. Post-processing involves maturation of the bioprinted tissue before its intended use [9,51] (Figure 1h–i).

2. Laser-Based Bioprinting

The main components of a laser-based bioprinter are the laser source, a laser transparent print ribbon coated with a layer of cell-laden bioink, and a substrate or collector slide on a motorized stage. The energy from the laser is utilized to pattern cell-laden bioinks in a three-dimensional spatial arrangement with the aid of computer-aided design and manufacturing (CAD/CAM). The high resolution and reproducibility of this process makes it a viable option for use in biomedical applications [53]. Some of the variations of this method based on the type of laser source and laser transparent print ribbon are shown in Table 1. Stem-cell grafts, skin tissue, multicellular arrays, and biopapers were reported to be printed using this method [23,54]. The major advantage of laser printing is the non-contact process. This eliminates nozzle clogging and also results in high cell viabilities [55]. However, there are several disadvantages of laser-based bioprinting, which outweigh the advantages. Laser exposure on the cells is not without risk, and the use of metal to absorb the laser energy can induce cytotoxicity [53].

3. Droplet-Based Bioprinting

Such a process ejects cell-laden bioink out of the nozzle onto a substrate in the form of droplets [51]. Inkjet printers are one of the most commonly used type for both non-biological and biological applications [9]. Inkjet printers can use thermal [27] or acoustic [56] forces, among others, to eject drops of liquid onto a substrate [9], as seen in Table 2. One of the major advantages of droplet-based bioprinting is its compatibility with a wide variety of biological materials. Furthermore, such bioprinters provide high resolution (20–100 µm) and speed (1–10,000 droplets/s) while being a low-cost alternative [29]. For example, a high-throughput cell printing system was demonstrated for drug screening [30,57,58,59,60,61]. At the same time, a major drawback of this technique is the requirement for the biological material to be in a liquid and less viscous form [29], which may not always be the case.

4. Extrusion-Based Bioprinting

In extrusion-based bioprinting, the bioink is extruded out of the nozzle using pneumatic pressure or mechanical force. The biggest advantage of extrusion-based bioprinting is the scalability due to the continuous flow of bioink and large deposition rate (Table 3). At the same time, the resolution of this method is lower than other methods [62,63]. While the printability of high-viscosity bioinks and high cell concentrations is an advantage [37], the inherent nozzle-clogging problem is a disadvantage [64]. Due to their low cost and simple-to-use nature, extrusion-based bioprinters are the most widely used of all bioprinters [51] (Table 4). Cell-laden constructs with tunable 3D microenvironments were constructed by bioprinting gelatin methacryloyl (GelMA)/alginate core/sheath microfibers using extrusion-based bioprinting and subsequent ultraviolet (UV) cross-linking [40]. Further stabilization strategies in extrusion-based bioprinting were also reported, in order to successfully complete the printing of intact, accurate, and biologically relevant constructs with desirable properties [65].

5. Stereolithography-Based Bioprinting

In stereolithography-based bioprinters, UV light is used to cure layers of photopolymer, stacks of which form the 3D object (Table 3). The biggest advantage of stereolithography in general and stereolithography-based bioprinting in particular is its very high resolution. Other advantages include high cell concentrations and no problem of nozzle clogging. The preparation of three-dimensional biodegradable poly(ethylene glycol)/poly(d,l-lactide) hydrogel structures using stereolithography at high resolutions was shown [46]. Cell-encapsulated hydrogels were also shown to be 3D-printed using stereolithography [48]. Cell-attachable and visible-light cross-linkable bioinks, based on gelatin methacryloyl (GelMA) with eosin Y (EY) photoinitiation, for stereolithography three-dimensional (3D) bioprinting were developed and used to print cell-laden hydrogels [13]. However, there are many disadvantages of this method. The biggest disadvantage is that only photocurable bioinks can be used. Another disadvantage is that the cells will get exposed to harmful UV light, which affects the cell viability [44].

6. 3D-Printed Tissues and Organs

Bioprinting was used to generate two-dimensional (2D) and 3D structures for various purposes, including fabrication of scaffolds and tissue constructs for tissue regeneration (Table 5). Some examples of printed tissues are shown in Figure 2. Markstedt et al. [66] printed shapes resembling human ear and sheep mensci using a bioink containing alginate and nanofibrillated cellulose and an inkjet-based 3D bioprinter which is largely applied for fibrous nanomaterials packaging [67]. Duan et al. [62] printed aortic valve conduits using hydrogel-based bioinks laden with aortic root sinus smooth-muscle cells and aortic valve leaflet interstitial cells and an extrusion-based 3D bioprinter. Table 5 summarizes the applications of 3D bioprinting in tissue engineering.
Therefore, it can be said that bioprinting holds tremendous potential and is fast moving toward fully functional 3D-printed organs. For example, in the future, chronic toxicological diseases that are majorly due to industrial particulate pollutants such as pulmonary fibrosis could be cured by transplanting 3D-printed lungs from patients’ own programmed cells. Life expectancy can be increased because patients will not be left waiting until a suitable organ is available from an organ donor. Body cells taken from patient blood or from a skin biopsy will be transported to a laboratory [87]. Here, cells will be programmed into routine culture to be transformed into diseased organ cells (e.g., lung cells) and will be expanded in volume/number for the 3D bioprinting to resemble a lung after a few weeks. After maturation into sterile culture conditions, the artificial lung will be ready to be implanted inside the patient (Figure 3) to replace the dysfunctional organ [88]. This whole process will take just a few months and will also produce personalized organs for the patient from their own cells. This will reduce the possibility of rejection by the body, and the patient will not have to spend the rest of his/her life on anti-rejection drugs and having to deal with all of the associated side effects. Petersen et al. [89] (Figure 3) proposed using scaffolds of extracellular matrix from lungs of adult rat that retain the hierarchical branching with cellular components removed. A bioreactor was used to culture pulmonary epithelium and vascular endothelium on the acellular lung matrix, resulting in hierarchical organization within the matrix and efficient repopulation of the vascular compartment. When implanted into rats in vivo for short time intervals (45 to 120 minutes) the engineered lungs participated in gas exchange, although the inflation of engineered lung was found to be less than that of the native lung, and some bleeding and clotting was observed. While this represents a step toward developing a viable strategy for generating fully functional lungs in vitro, there remains the issue of extracting scaffolds from lungs, among others. This is where 3D bioprinting can help by developing patient-specific, on-demand biological scaffolds.
The entire process involves nano- and micro-to macroscale bottom-up engineering [90,91] using a simple desktop 3D printer. This opens up the possibility that, one day, we will be able to bioprint amputated sub-organs, missing organs, and digitally designed cosmetic body parts. Instead of using plastics to print structures, researchers will use living cells mixed with biocompatible scaffolds to build living tissue inside a sterile safety cabinet to keep the cells protected from harmful foreign substances. In this context, it is also important to discuss the relevance of four-dimensional (4D) bioprinting, the fourth dimension being time [92]. While 3D bioprinting is set to make our lives easier by printing required living tissue on demand, in some cases, it may lose relevance if it is too time-consuming. Therefore, time taken to create the end-product is an important parameter to consider when judging the effectiveness of bioprinting processes. Furthermore, it was suggested that a universal bioink would be a significant technological advancement that could standardize the bioprinting field and accelerate the realization of human tissue product biomanufacturing [93]. With advancement in 3D printing, parallel advancements in 3D bioprinting can also be expected in the future. For example, dip-pen nanolithography is being developed, combining advantages of electron beam lithography, inkjet printing, and microcontact printing [94]. Such methods also allow the parallel application of different inks, which may be useful for printing complex tissue structures when integrated with in silico modeling [95,96].

7. Future Outlook: 3D Bioprinting Air–Liquid Interface (ALI) as an Artificial Material for Nanotoxicity Assessment of Particulate Matter

With the huge potential that 3D bioprinting holds, other applications apart from tissue/organ regeneration can be realized [97], for example, printing a lattice-like membrane, which can act as a biological tape. Such a membrane when placed in a culture microincubator could be used to recreate the microenvironment of the human body. To mimic the air–liquid interface in vivo, 3D bioprinting can build living lung-like tissues via printing the inside of the incredibly intricate branching network of tubes [98,99]. Each of these tubes ends in a tiny structure of air sacs/pouches where oxygen and carbon dioxide are exchanged, which gives an idea of just how complex this structure is. There are 300 million of these tiny air sacs in each lung [100], which makes it a very challenging structure to bioprint. However, a part of it can be printed to be used as a very useful model for toxicological research [101]. Another example is an asthma attack, where patients breathe in certain particulate allergens (micro- and nano- to macroscale particles/airborne spores) which aggravate muscle contraction of reduced-diameter airways [102]. In the future, starting from those airway muscle cells, one can recreate them in the lab and print them into tubular structures [103]. Further incubating these constructs in a suitable microenvironment to mature into similar functional airway muscles is possible via adding a stimulatory compound like histamine. It is released in asthmatic patient airways during an asthma attack, causing muscle contraction. We will be able to 3D print an airway muscle tissue mimicking the biological lifelike contraction and, thus, test advance therapeutics to reverse the contraction, relaxing the air tube as an anti-asthma drug does in patients. The fact that drug tests can be performed in these tissues is a very important point, because the drug development industry faces a big challenge of human trials after testing in vitro cells grown in petri dishes and preclinical tests in in vivo rodents. Rodents such as mice and rats respond very differently to test therapeutic compounds than humans do [104]. There is a huge chasm between the preclinical tools that we test the drugs on and the humans for whom the drugs are designed to help. Therefore, 90% of drugs that show promise in animals actually fail to work in humans, usually because they are just not effective at fighting disease or, sometimes, they are downright toxic [105]. These 3D-printed tissues can help the drug development process by enabling pharmaceutical companies to test these compounds in tissues that reproduce the complexity of the human body [106]. This will save lives by providing better drugs to patients faster and for less expense. It also has an ethical and moral impact, because research can drastically reduce the number of animals that are used for drug development. By the year 2050, it is estimated that the meat and leather industry combined will need around one hundred billion farm animals to supply us with our animal-based needs such as meat, leather, milk, etc. [107]. To supplement those kinds of needs, animal cells can now be grown in the laboratory in just the same way as human cells; thus, there is potential here to replace a large proportion of these animals using bioprinted cells. We can differentiate them into muscle-like cells and then print those cells into meat products. The first bioprinted beef burger was revealed back in 2013, although incurring high costs (approximately $300,000) [108]. As technology moves forward rapidly, bioprinted leather is also a potential use for this technology. Skin cells can be grown, and the industry could generate customized leather products with specific thicknesses or textures or colors, making it feasible for the potential replacement of animal products by even better bioprinted animal products [109].

Funding

This research received no external funding.

Acknowledgments

A.V.S. thanks the Max Planck Society for the grassroot project grant 2017 (M10335) and 2018 (M10338).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Rustad, K.C.; Sorkin, M.; Levi, B.; Longaker, M.T.; Gurtner, G.C. Strategies for organ level tissue engineering. Organogenesis 2010, 6, 151–157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Atala, A.; Kasper, F.K.; Mikos, A.G. Engineering Complex Tissues. Sci. Transl. Med. 2012, 4, 160rv112. [Google Scholar] [CrossRef] [PubMed]
  3. Griffith, L.G.; Naughton, G. Tissue engineering--current challenges and expanding opportunities. Science 2002, 295, 1009. [Google Scholar] [CrossRef] [PubMed]
  4. Ikada, Y. Challenges in tissue engineering. J. R. Soc. Interface 2006, 3, 589–601. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Nigam, R.; Mahanta, B. An overview of various biomimetic scaffolds: Challenges and applications in tissue engineering. J. Tissue Sci. Eng. 2014, 5, 1. [Google Scholar]
  6. Khademhosseini, A.; Langer, R. A decade of progress in tissue engineering. Nat. Protoc. 2016, 11, 1775. [Google Scholar] [CrossRef] [PubMed]
  7. Mironov, V.; Reis, N.; Derby, B. Review: Bioprinting: A beginning. Tissue Eng. 2006, 12, 631–634. [Google Scholar] [CrossRef] [PubMed]
  8. Seol, Y.-J.; Kang, H.-W.; Lee, S.J.; Atala, A.; Yoo, J.J. Bioprinting technology and its applications. Eur. J. Cardiothorac. Surg. 2014, 46, 342–348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Murphy, S.V.; Atala, A. 3D bioprinting of tissues and organs. Nat. Biotechnol. 2014, 32, 773. [Google Scholar] [CrossRef] [PubMed]
  10. Rengier, F.; Mehndiratta, A.; von Tengg-Kobligk, H.; Zechmann, C.M.; Unterhinninghofen, R.; Kauczor, H.U.; Giesel, F.L. 3D printing based on imaging data: review of medical applications. Int. J. Comput. Assist. Radiol. Surg. 2010, 5, 335–341. [Google Scholar] [CrossRef] [PubMed]
  11. Gopinathan, J.; Noh, I. Recent trends in bioinks for 3D printing. Biomater. Res. 2018, 22, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Gungor-Ozkerim, P.S.; Inci, I.; Zhang, Y.S.; Khademhosseini, A.; Dokmeci, M.R. Bioinks for 3D bioprinting: an overview. Biomater. Sci. 2018, 6, 915–946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Wang, Z.; Kumar, H.; Tian, Z.; Jin, X.; Holzman, J.F.; Menard, F.; Kim, K. Visible light photoinitiation of cell-adhesive gelatin methacryloyl hydrogels for stereolithography 3D bioprinting. ACS Appl. Mater. Interfaces 2018, 10, 26859–26869. [Google Scholar] [CrossRef] [PubMed]
  14. Bieligmeyer, M.; Artukovic, F.; Nussberger, S.; Hirth, T.; Schiestel, T.; Müller, M. Reconstitution of the membrane protein OmpF into biomimetic block copolymer-phospholipid hybrid membranes. Beilstein J. Nanotechnol. 2016, 7, 881–892. [Google Scholar] [CrossRef] [PubMed]
  15. Guillotin, B.; Souquet, A.; Catros, S.; Duocastella, M.; Pippenger, B.; Bellance, S.; Bareille, R.; Rémy, M.; Bordenave, L.; Amédée, J.; et al. Laser assisted bioprinting of engineered tissue with high cell density and microscale organization. Biomaterials 2010, 31, 7250–7256. [Google Scholar] [CrossRef] [PubMed]
  16. Barron, J.A.; Wu, P.; Ladouceur, H.D.; Ringeisen, B.R. Biological laser printing: A novel technique for creating heterogeneous 3-dimensional cell patterns. BioMi 2004, 6, 139–147. [Google Scholar] [CrossRef]
  17. Guillemot, F.; Souquet, A.; Catros, S.; Guillotin, B.; Lopez, J.; Faucon, M.; Pippenger, B.; Bareille, R.; Rémy, M.; Bellance, S.; et al. High-throughput laser printing of cells and biomaterials for tissue engineering. Acta Biomater. 2010, 6, 2494–2500. [Google Scholar] [CrossRef] [PubMed]
  18. Zhang, X.; Zhang, Y. Tissue engineering applications of three-dimensional bioprinting. Cell Biochem. Biophys. 2015, 72, 777–782. [Google Scholar] [CrossRef] [PubMed]
  19. Ali, M.; Pages, E.; Ducom, A.; Fontaine, A.; Guillemot, F. Controlling laser-induced jet formation for bioprinting mesenchymal stem cells with high viability and high resolution. Biofabrication 2014, 6, 045001. [Google Scholar] [CrossRef] [PubMed]
  20. Koch, L.; Deiwick, A.; Franke, A.; Schwanke, K.; Haverich, A.; Zweigerdt, R.; Chichkov, B. Laser bioprinting of human induced pluripotent stem cells—the effect of printing and biomaterials on cell survival, pluripotency, and differentiation. Biofabrication 2018, 10, 035005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Wang, Z.; Jin, X.; Tian, Z.; Menard, F.; Holzman, J.F.; Kim, K. A Novel, Well-resolved direct laser bioprinting system for rapid cell encapsulation and microwell fabrication. Adv. Healthcare Mater. 2018, 7, 1701249. [Google Scholar] [CrossRef] [PubMed]
  22. Petit, S.; Kérourédan, O.; Devillard, R.; Cormier, E. Femtosecond versus picosecond laser pulses for film-free laser bioprinting. Appl. Opt. 2017, 56, 8648–8655. [Google Scholar] [CrossRef] [PubMed]
  23. Koch, L.; Deiwick, A.; Chichkov, B. Laser-Based Cell Printing. In 3D Printing and Biofabrication; Springer, Cham: Basel, Switzerland, 2018; pp. 303–329. [Google Scholar]
  24. Klebe, R.J. Cytoscribing: A method for micropositioning cells and the construction of two- and three-dimensional synthetic tissues. Exp. Cell Res. 1988, 179, 362–373. [Google Scholar] [CrossRef]
  25. Xu, T.; Zhao, W.; Zhu, J.-M.; Albanna, M.Z.; Yoo, J.J.; Atala, A. Complex heterogeneous tissue constructs containing multiple cell types prepared by inkjet printing technology. Biomaterials 2013, 34, 130–139. [Google Scholar] [CrossRef] [PubMed]
  26. Xu, T.; Jin, J.; Gregory, C.; Hickman, J.J.; Boland, T. Inkjet printing of viable mammalian cells. Biomaterials 2005, 26, 93–99. [Google Scholar] [CrossRef] [PubMed]
  27. Cui, X.; Bolond, T.; D’Lima, D.D.; Lotz, M.K. Thermal inkjet printing in tissue engineering and regenerative medicine. Recent Pat. Drug Deliv. Formul. 2012, 6, 149–155. [Google Scholar] [CrossRef] [PubMed]
  28. Sundaramurthi, D.; Rauf, S.; Hauser, C. 3D bioprinting technology for regenerative medicine applications. Int. J. Bioprinting 2016, 2, 9–26. [Google Scholar] [CrossRef]
  29. Dai, G.; Lee, V. Three-dimensional bioprinting and tissue fabrication: prospects for drug discovery and regenerative medicine. Adv. Health Care Technol. 2015, 1, 23–25. [Google Scholar] [CrossRef]
  30. Li, X.; Chen, J.; Liu, B.; Wang, X.; Ren, D.; Xu, T. Inkjet Printing for Biofabrication. In 3D Printing and Biofabrication. Reference Series in Biomedical Engineering; Ovsianikov, A., Yoo, J., Mironov, V., Eds.; Springer, Cham: Basel, Switzerland, 2018; pp. 1–19. [Google Scholar] [CrossRef]
  31. Hewes, S.; Wong, A.D.; Searson, P.C. Bioprinting microvessels using an inkjet printer. Bioprinting 2017, 7, 14–18. [Google Scholar] [CrossRef]
  32. Cohen, D.L.; Malone, E.; Lipson, H.; Bonassar, L.J. Direct freeform fabrication of seeded hydrogels in arbitrary geometries. Tissue Eng. 2006, 12, 1325–1335. [Google Scholar] [CrossRef] [PubMed]
  33. Iwami, K.; Noda, T.; Ishida, K.; Morishima, K.; Nakamura, M.; Umeda, N. Bio rapid prototyping by extruding/aspirating/refilling thermoreversible hydrogel. Biofabrication 2010, 2, 014108. [Google Scholar] [CrossRef] [PubMed]
  34. Shor, L.; Güçeri, S.; Chang, R.; Gordon, J.; Kang, Q.; Hartsock, L.; An, Y.; Sun, W. Precision extruding deposition (PED) fabrication of polycaprolactone (PCL) scaffolds for bone tissue engineering. Biofabrication 2009, 1, 015003. [Google Scholar] [CrossRef] [PubMed]
  35. Mannoor, M.S.; Jiang, Z.; James, T.; Kong, Y.L.; Malatesta, K.A.; Soboyejo, W.O.; Verma, N.; Gracias, D.H.; McAlpine, M.C. 3D printed bionic ears. Nano Lett. 2013, 13, 2634–2639. [Google Scholar] [CrossRef] [PubMed]
  36. Lee, V.K.; Dai, G. Printing of three-dimensional tissue analogs for regenerative medicine. Ann. Biomed. Eng. 2017, 45, 115–131. [Google Scholar] [CrossRef] [PubMed]
  37. Panwar, A.; Tan, P.L. Current status of bioinks for micro-extrusion-based 3D bioprinting. Molecules 2016, 21, 685. [Google Scholar] [CrossRef] [PubMed]
  38. Lee, K.; Jin, G.; Jang, C.H.; Jung, W.-K.; Kim, G. Preparation and characterization of multi-layered poly(ε-caprolactone)/chitosan scaffolds fabricated with a combination of melt-plotting/in situ plasma treatment and a coating method for hard tissue regeneration. J. Mater. Chem. B 2013, 1, 5831–5841. [Google Scholar] [CrossRef]
  39. Hong, J.M.; Kim, B.J.; Shim, J.-H.; Kang, K.S.; Kim, K.-J.; Rhie, J.W.; Cha, H.J.; Cho, D.-W. Enhancement of bone regeneration through facile surface functionalization of solid freeform fabrication-based three-dimensional scaffolds using mussel adhesive proteins. Acta Biomater. 2012, 8, 2578–2586. [Google Scholar] [CrossRef] [PubMed]
  40. Liu, W.; Zhong, Z.; Hu, N.; Zhou, Y.; Maggio, L.; Miri, A.K.; Fragasso, A.; Jin, X.; Khademhosseini, A.; Zhang, Y.S. Coaxial extrusion bioprinting of 3D microfibrous constructs with cell-favorable gelatin methacryloyl microenvironments. Biofabrication 2018, 10, 024102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Placone, J.K.; Engler, A.J. Recent advances in extrusion-based 3D printing for biomedical applications. Adv. Healthcare Mater. 2018, 7, 1701161. [Google Scholar] [CrossRef] [PubMed]
  42. Agarwala, S.; Lee, J.M.; Ng, W.L.; Layani, M.; Yeong, W.Y.; Magdassi, S. A novel 3D bioprinted flexible and biocompatible hydrogel bioelectronic platform. Biosens. Bioelectron. 2018, 102, 365–371. [Google Scholar] [CrossRef] [PubMed]
  43. Soman, P.; Chung, P.H.; Zhang, A.P.; Chen, S. Digital microfabrication of user-defined 3D microstructures in cell-laden hydrogels. Biotechnol. Bioeng. 2013, 110, 3038–3047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Arcaute, K.; Mann, B.K.; Wicker, R.B. Stereolithography of three-dimensional bioactive poly(ethylene glycol) constructs with encapsulated cells. Ann. Biomed. Eng. 2006, 34, 1429–1441. [Google Scholar] [CrossRef] [PubMed]
  45. Curley, J.L.; Jennings, S.R.; Moore, M.J. Fabrication of micropatterned hydrogels for neural culture systems using dynamic mask projection photolithography. J. Vis. Exp. 2011, 48, 2636. [Google Scholar] [CrossRef] [PubMed]
  46. Elomaa, L.; Pan, C.-C.; Shanjani, Y.; Malkovskiy, A.; Seppälä, J.V.; Yang, Y. Three-dimensional fabrication of cell-laden biodegradable poly(ethylene glycol-co-depsipeptide) hydrogels by visible light stereolithography. J. Mater. Chem. B 2015, 3, 8348–8358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Elomaa, L.; Teixeira, S.; Hakala, R.; Korhonen, H.; Grijpma, D.W.; Seppälä, J.V. Preparation of poly(ε-caprolactone)-based tissue engineering scaffolds by stereolithography. Acta Biomater. 2011, 7, 3850–3856. [Google Scholar] [CrossRef] [PubMed]
  48. Chan, V.; Zorlutuna, P.; Jeong, J.H.; Kong, H.; Bashir, R. Three-dimensional photopatterning of hydrogels using stereolithography for long-term cell encapsulation. Lab Chip. 2010, 10, 2062–2070. [Google Scholar] [CrossRef] [PubMed]
  49. Seck, T.M.; Melchels, F.P.W.; Feijen, J.; Grijpma, D.W. Designed biodegradable hydrogel structures prepared by stereolithography using poly(ethylene glycol)/poly(d,l-lactide)-based resins. J. Controlled Release 2010, 148, 34–41. [Google Scholar] [CrossRef] [PubMed]
  50. Miri, A.K.; Nieto, D.; Iglesias, L.; Goodarzi Hosseinabadi, H.; Maharjan, S.; Ruiz-Esparza, G.U.; Khoshakhlagh, P.; Manbachi, A.; Dokmeci, M.R.; Chen, S.; et al. Microfluidics-enabled multimaterial maskless stereolithographic bioprinting. Adv. Mater. 2018, 30, 1800242. [Google Scholar] [CrossRef] [PubMed]
  51. Vijayavenkataraman, S.; Yan, W.-C.; Lu, W.F.; Wang, C.-H.; Fuh, J.Y.H. 3D bioprinting of tissues and organs for regenerative medicine. Adv. Drug Del. Rev. 2018, 132, 296–332. [Google Scholar] [CrossRef] [PubMed]
  52. Norotte, C.; Marga, F.S.; Niklason, L.E.; Forgacs, G. Scaffold-free vascular tissue engineering using bioprinting. Biomaterials 2009, 30, 5910–5917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Schiele, N.R.; Corr, D.T.; Huang, Y.; Raof, N.A.; Xie, Y.; Chrisey, D.B. Laser-based direct-write techniques for cell printing. Biofabrication 2010, 2, 032001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Kawecki, F.; Clafshenkel, W.P.; Auger, F.A.; Bourget, J.M.; Fradette, J.; Devillard, R. Self-assembled human osseous cell sheets as living biopapers for the laser-assisted bioprinting of human endothelial cells. Biofabrication 2018, 10, 035006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Hopp, B.; Smausz, T.; Kresz, N.; Barna, N.; Bor, Z.; Kolozsvári, L.; Chrisey, D.B.; Szabó, A.; Nógrádi, A. Survival and proliferative ability of various living cell types after laser-induced forward transfer. Tissue Eng. 2005, 11, 1817–1823. [Google Scholar] [CrossRef] [PubMed]
  56. Okamoto, T.; Suzuki, T.; Yamamoto, N. Microarray fabrication with covalent attachment of DNA using Bubble Jet technology. Nat. Biotechnol. 2000, 18, 438. [Google Scholar] [CrossRef] [PubMed]
  57. Rodríguez-Dévora, J.I.; Zhang, B.; Reyna, D.; Shi, Z.-d.; Xu, T. High throughput miniature drug-screening platform using bioprinting technology. Biofabrication 2012, 4, 035001. [Google Scholar] [CrossRef] [PubMed]
  58. Matsusaki, M.; Sakaue, K.; Kadowaki, K.; Akashi, M. Three-Dimensional Human Tissue Chips Fabricated by Rapid and Automatic Inkjet Cell Printing. Adv. Healthc. Mater. 2013, 2, 534–539. [Google Scholar] [CrossRef] [PubMed]
  59. Xu, F.; Celli, J.; Rizvi, I.; Moon, S.; Hasan, T.; Demirci, U. A three-dimensional in vitro ovarian cancer coculture model using a high-throughput cell patterning platform. Biotechnol. J. 2011, 6, 204–212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Scoutaris, N.; Ross, S.; Douroumis, D. Current trends on medical and pharmaceutical applications of inkjet printing technology. Pharm. Res. 2016, 33, 1799–1816. [Google Scholar] [CrossRef] [PubMed]
  61. Gudapati, H.; Dey, M.; Ozbolat, I. A comprehensive review on droplet-based bioprinting: Past, present and future. Biomaterials 2016, 102, 20–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Duan, B.; Hockaday, L.A.; Kang, K.H.; Butcher, J.T. 3D Bioprinting of heterogeneous aortic valve conduits with alginate/gelatin hydrogels. J. Biomed. Mater. Res. Part A 2013, 101, 1255–1264. [Google Scholar] [CrossRef] [PubMed]
  63. Dababneh, A.B.; Ozbolat, I.T. Bioprinting technology: A current state-of-the-art review. J. Manuf. Sci. Eng. 2014, 136, 061016. [Google Scholar] [CrossRef]
  64. Malda, J.; Visser, J.; Melchels, F.P.; Jüngst, T.; Hennink, W.E.; Dhert, W.J.A.; Groll, J.; Hutmacher, D.W. 25th Anniversary article: Engineering hydrogels for biofabrication. Adv. Mater. 2013, 25, 5011–5028. [Google Scholar] [CrossRef] [PubMed]
  65. Shapira, A.; Noor, N.; Asulin, M.; Dvir, T. Stabilization strategies in extrusion-based 3D bioprinting for tissue engineering. Appl. Phys. Rev. 2018, 5, 041112. [Google Scholar] [CrossRef]
  66. Markstedt, K.; Mantas, A.; Tournier, I.; Martínez Ávila, H.; Hägg, D.; Gatenholm, P. 3D bioprinting human chondrocytes with nanocellulose–alginate bioink for cartilage tissue engineering applications. Biomacromolecules 2015, 16, 1489–1496. [Google Scholar] [CrossRef] [PubMed]
  67. Dwivedi, C.; Pandey, I.; Misra, V.; Giulbudagian, M.; Jungnickel, H.; Laux, P.; Luch, A.; Ramteke, P.W.; Vikram Singh, A. The prospective role of nanobiotechnology in food and food packaging products. Integr. Food Nutr. Metab. 2018, 5, 1–5. [Google Scholar] [CrossRef]
  68. Duan, B.; Kapetanovic, E.; Hockaday, L.A.; Butcher, J.T. Three-dimensional printed trileaflet valve conduits using biological hydrogels and human valve interstitial cells. Acta Biomater. 2014, 10, 1836–1846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Duan, B. State-of-the-art review of 3D bioprinting for cardiovascular tissue engineering. Ann. Biomed. Eng. 2017, 45, 195–209. [Google Scholar] [CrossRef] [PubMed]
  70. Gaetani, R.; Doevendans, P.A.; Metz, C.H.G.; Alblas, J.; Messina, E.; Giacomello, A.; Sluijter, J.P.G. Cardiac tissue engineering using tissue printing technology and human cardiac progenitor cells. Biomaterials 2012, 33, 1782–1790. [Google Scholar] [CrossRef] [PubMed]
  71. Gaebel, R.; Ma, N.; Liu, J.; Guan, J.; Koch, L.; Klopsch, C.; Gruene, M.; Toelk, A.; Wang, W.; Mark, P.; et al. Patterning human stem cells and endothelial cells with laser printing for cardiac regeneration. Biomaterials 2011, 32, 9218–9230. [Google Scholar] [CrossRef] [PubMed]
  72. Wu, P.K.; Ringeisen, B.R. Development of human umbilical vein endothelial cell (HUVEC) and human umbilical vein smooth muscle cell (HUVSMC) branch/stem structures on hydrogel layers via biological laser printing (BioLP). Biofabrication 2010, 2, 014111. [Google Scholar] [CrossRef] [PubMed]
  73. Cui, X.; Boland, T. Human microvasculature fabrication using thermal inkjet printing technology. Biomaterials 2009, 30, 6221–6227. [Google Scholar] [CrossRef] [PubMed]
  74. Jakab, K.; Norotte, C.; Damon, B.; Marga, F.; Neagu, A.; Besch-Williford, C.L.; Kachurin, A.; Church, K.H.; Park, H.; Mironov, V.; et al. Tissue engineering by self-assembly of cells printed into topologically defined structures. Tissue Eng. Part A 2008, 14, 413–421. [Google Scholar] [CrossRef] [PubMed]
  75. Phillippi, J.A.; Miller, E.; Weiss, L.; Huard, J.; Waggoner, A.; Campbell, P. Microenvironments Engineered by Inkjet Bioprinting Spatially Direct Adult Stem Cells Toward Muscle- and Bone-Like Subpopulations. Stem Cells 2008, 26, 127–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Ker, E.D.F.; Nain, A.S.; Weiss, L.E.; Wang, J.; Suhan, J.; Amon, C.H.; Campbell, P.G. Bioprinting of growth factors onto aligned sub-micron fibrous scaffolds for simultaneous control of cell differentiation and alignment. Biomaterials 2011, 32, 8097–8107. [Google Scholar] [CrossRef] [PubMed]
  77. Dwivedi, C.; Pandey, I.; Pandey, H.; Patil, S.; Mishra, S.B.; Pandey, A.C.; Zamboni, P.; Ramteke, P.W.; Singh, A.V. In vivo diabetic wound healing with nanofibrous scaffolds modified with gentamicin and recombinant human epidermal growth factor. J. Biomed. Mater. Res. Part A 2018, 106, 641–651. [Google Scholar] [CrossRef] [PubMed]
  78. Lee, C.H.; Cook, J.L.; Mendelson, A.; Moioli, E.K.; Yao, H.; Mao, J.J. Regeneration of the articular surface of the rabbit synovial joint by cell homing: a proof of concept study. Lancet 2010, 376, 440–448. [Google Scholar] [CrossRef] [Green Version]
  79. Fedorovich, N.E.; De Wijn, J.R.; Verbout, A.J.; Alblas, J.; Dhert, W.J.A. Three-dimensional fiber deposition of cell-laden, ciable, patterned constructs for bone tissue printing. Tissue Eng. Part A 2008, 14, 127–133. [Google Scholar] [CrossRef] [PubMed]
  80. Xu, T.; Gregory, C.A.; Molnar, P.; Cui, X.; Jalota, S.; Bhaduri, S.B.; Boland, T. Viability and electrophysiology of neural cell structures generated by the inkjet printing method. Biomaterials 2006, 27, 3580–3588. [Google Scholar] [CrossRef] [PubMed]
  81. Ilkhanizadeh, S.; Teixeira, A.I.; Hermanson, O. Inkjet printing of macromolecules on hydrogels to steer neural stem cell differentiation. Biomaterials 2007, 28, 3936–3943. [Google Scholar] [CrossRef] [PubMed]
  82. Marga, F.; Jakab, K.; Khatiwala, C.; Shepherd, B.; Dorfman, S.; Hubbard, B.; Colbert, S.; Gabor, F. Toward engineering functional organ modules by additive manufacturing. Biofabrication 2012, 4, 022001. [Google Scholar] [CrossRef] [PubMed]
  83. Hsu, S.-H. 3D bioprinting: A new insight into the therapeutic strategy of neural tissue regeneration AU - Hsieh, Fu-Yu. Organogenesis 2015, 11, 153–158. [Google Scholar] [CrossRef]
  84. Lee, W.; Debasitis, J.C.; Lee, V.K.; Lee, J.-H.; Fischer, K.; Edminster, K.; Park, J.-K.; Yoo, S.-S. Multi-layered culture of human skin fibroblasts and keratinocytes through three-dimensional freeform fabrication. Biomaterials 2009, 30, 1587–1595. [Google Scholar] [CrossRef] [PubMed]
  85. Huang, S.; Yao, B.; Xie, J.; Fu, X. 3D bioprinted extracellular matrix mimics facilitate directed differentiation of epithelial progenitors for sweat gland regeneration. Acta Biomater. 2016, 32, 170–177. [Google Scholar] [CrossRef] [PubMed]
  86. Byambaa, B.; Annabi, N.; Yue, K.; Trujillo-de Santiago, G.; Alvarez, M.M.; Jia, W.; Kazemzadeh-Narbat, M.; Shin, S.R.; Tamayol, A.; Khademhosseini, A. Bioprinted osteogenic and vasculogenic patterns for engineering 3D bone tissue. Adv. Healthcare Mater. 2017, 6, 1700015. [Google Scholar] [CrossRef] [PubMed]
  87. Singh, A.V.; Gailite, L.; Vyas, V.; Lenardi, C.; Forti, S.; Matteoli, M.; Milani, P. Rapid prototyping of nano- and micro-patterned substrates for the control of cell neuritogenesis by topographic and chemical cues. Mater. Sci. Eng. C 2011, 31, 892–899. [Google Scholar] [CrossRef]
  88. Singh, A.V.; Lenardi, C.; Gailite, L.; Gianfelice, A.; Milani, P. A simple lift-off-based patterning method for micro- and nanostructuring of functional substrates for cell culture. J. Micromech. Microeng. 2009, 19, 115028. [Google Scholar] [CrossRef]
  89. Petersen, T.H.; Calle, E.A.; Zhao, L.; Lee, E.J.; Gui, L.; Raredon, M.B.; Gavrilov, K.; Yi, T.; Zhuang, Z.W.; Breuer, C.; et al. Tissue-engineered lungs for in vivo implantation. Science 2010, 329, 538–541. [Google Scholar] [CrossRef] [PubMed]
  90. Singh, A.V.; Ferri, M.; Tamplenizza, M.; Borghi, F.; Divitini, G.; Ducati, C.; Lenardi, C.; Piazzoni, C.; Merlini, M.; Podestà, A.; et al. Bottom-up engineering of the surface roughness of nanostructured cubic zirconia to control cell adhesion. Nanotechnology 2012, 23, 475101. [Google Scholar] [CrossRef] [PubMed]
  91. Ajay, V.S.; Krunal, K.M. Top-down versus bottom-up nanoengineering routes to design advanced oropharmacological products. Curr. Pharm. Des. 2016, 22, 1534–1545. [Google Scholar] [CrossRef]
  92. Miao, S.; Castro, N.; Nowicki, M.; Xia, L.; Cui, H.; Zhou, X.; Zhu, W.; Lee, S.-j.; Sarkar, K.; Vozzi, G.; et al. 4D printing of polymeric materials for tissue and organ regeneration. Mater. Today 2017, 20, 577–591. [Google Scholar] [CrossRef] [PubMed]
  93. Skardal, A. Perspective: “Universal” bioink technology for advancing extrusion bioprinting-based biomanufacturing. Bioprinting 2018, 10, e00026. [Google Scholar] [CrossRef]
  94. Hirtz, M.; Oikonomou, A.; Georgiou, T.; Fuchs, H.; Vijayaraghavan, A. Multiplexed biomimetic lipid membranes on graphene by dip-pen nanolithography. Nat. Commun. 2013, 4, 2591. [Google Scholar] [CrossRef] [PubMed]
  95. Singh, A.V.; Jahnke, T.; Kishore, V.; Park, B.-W.; Batuwangala, M.; Bill, J.; Sitti, M. Cancer cells biomineralize ionic gold into nanoparticles-microplates via secreting defense proteins with specific gold-binding peptides. Acta Biomater. 2018, 71, 61–71. [Google Scholar] [CrossRef] [PubMed]
  96. Singh, A.V.; Jahnke, T.; Wang, S.; Xiao, Y.; Alapan, Y.; Kharratian, S.; Onbasli, M.C.; Kozielski, K.; David, H.; Richter, G.; et al. Anisotropic gold nanostructures: Optimization via in silico modeling for hyperthermia. ACS Appl. Nano Mater. 2018, 1, 6205–6216. [Google Scholar] [CrossRef]
  97. Ozbolat, I.T.; Peng, W.; Ozbolat, V. Application areas of 3D bioprinting. Drug Discov. Today 2016, 21, 1257–1271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Horváth, L.; Umehara, Y.; Jud, C.; Blank, F.; Petri-Fink, A.; Rothen-Rutishauser, B. Engineering an in vitro air-blood barrier by 3D bioprinting. Sci. Rep. 2015, 5, 7974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Meeta, G.; Ajay, V.S. Nanoparticle enabled drug delivery across the blood brain barrier: in vivo and in vitro models, opportunities and challenges. Curr. Pharm. Biotechnol. 2013, 14, 1201–1212. [Google Scholar] [CrossRef]
  100. Ochs, M.; Nyengaard, J.R.; Jung, A.; Knudsen, L.; Voigt, M.; Wahlers, T.; Richter, J.; Gundersen, H.J.G. The number of alveoli in the human lung. Am. J. Respir. Crit. Care Med. 2004, 169, 120–124. [Google Scholar] [CrossRef] [PubMed]
  101. Singh, A.V.; Laux, P.; Luch, A.; Sudrik, C.; Wiehr, S.; Wild, A.-M.; Santomauro, G.; Bill, J.; Sitti, M. Review of emerging concepts in nanotoxicology: opportunities and challenges for safer nanomaterial design. Toxicol. Mech. Methods 2019, 14, 1–10. [Google Scholar] [CrossRef] [PubMed]
  102. Holgate, S.T. The epidemic of allergy and asthma. Nature 1999, 402, 2. [Google Scholar] [CrossRef]
  103. Singh, A.V.; Patil, R.; Thombre, D.K.; Gade, W.N. Micro-nanopatterning as tool to study the role of physicochemical properties on cell–surface interactions. J. Biomed. Mater. Res. Part A 2013, 101, 3019–3032. [Google Scholar] [CrossRef] [PubMed]
  104. Ajay Vikram, S. Editorial (Thematic Issue: Recent Trends in Nano-Biotechnology Reinforcing Contemporary Pharmaceutical Design). Curr. Pharm. Des. 2016, 22, 1415–1417. [Google Scholar] [CrossRef]
  105. Bracken, M.B. Why animal studies are often poor predictors of human reactions to exposure. J. R. Soc. Med. 2009, 102, 120–122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Vikram Singh, A.; Gharat, T.; Batuwangala, M.; Park, B.-W.; Endlein, T.; Sitti, M. Three-dimensional patterning in biomedicine: Importance and applications in neuropharmacology. J. Biomed. Mater. Res. B Appl. Biomater. 2018, 106, 1369–1382. [Google Scholar] [CrossRef] [PubMed]
  107. Koneswaran, G.; Nierenberg, D. Global farm animal production and global warming: impacting and mitigating climate change. Environ. Health Perspect. 2008, 116, 578–582. [Google Scholar] [CrossRef] [PubMed]
  108. Building a $325,000 Burger. Available online: https://www.nytimes.com/2013/05/14/science/engineering-the-325000-in-vitro-burger.html (accessed on 30 January 2019).
  109. Singh, A.V.; Gemmati, D.; Kanase, A.; Pandey, I.; Misra, V.; Kishore, V.; Jahnke, T.; Bill, J. Nanobiomaterials for vascular biology and wound management: A review. Veins Lymphat. 2018, 7. [Google Scholar] [CrossRef]
Figure 1. A typical process for bioprinting 3D tissues. Imaging of the tissue or organ using (a) CT scanner (shown here is a Siemens SOMATOM Force © Siemens Healthcare GmbH, 2018), or (b) MRI machine (shown here is a Siemens MAGNETOM Sola © Siemens Healthcare GmbH, 2018) and (c,d) reconstruction of 3D models from the imaging (shown here are models of a near and a heart). (e) The composition of bioink depends on the intended tissue form and function. Using such inks, methods like (f) laser-based bioprinting, or (g) extrusion-based bioprinting can be employed to print the intended tissue. (h,i) Some form of post-processing or maturation may be needed before the 3-d bioprinted tissue can be used (shown here is the maturation of bioprinted tubes composed of porcine aortic smooth muscle cells in a perfusion bioreactor). 3D model of ear is reprinted from Mannoor et.al. [35], with permission from American Chemical Society; Bioink formulation schematic is taken from Gungor-Ozkerin et.al. [12], with permission from Royal Society of Chemistry; Schematic of laser-based bioprinting is taken from Guillemot et.al. [17], with permission from Elsevier; Schematic of extrusion-based bioprinting is taken from Mannoor et.al. [35], with permission from American Chemical Society; Post-processing images are taken from Norotte et.al. [52], with permission from Elsevier.
Figure 1. A typical process for bioprinting 3D tissues. Imaging of the tissue or organ using (a) CT scanner (shown here is a Siemens SOMATOM Force © Siemens Healthcare GmbH, 2018), or (b) MRI machine (shown here is a Siemens MAGNETOM Sola © Siemens Healthcare GmbH, 2018) and (c,d) reconstruction of 3D models from the imaging (shown here are models of a near and a heart). (e) The composition of bioink depends on the intended tissue form and function. Using such inks, methods like (f) laser-based bioprinting, or (g) extrusion-based bioprinting can be employed to print the intended tissue. (h,i) Some form of post-processing or maturation may be needed before the 3-d bioprinted tissue can be used (shown here is the maturation of bioprinted tubes composed of porcine aortic smooth muscle cells in a perfusion bioreactor). 3D model of ear is reprinted from Mannoor et.al. [35], with permission from American Chemical Society; Bioink formulation schematic is taken from Gungor-Ozkerin et.al. [12], with permission from Royal Society of Chemistry; Schematic of laser-based bioprinting is taken from Guillemot et.al. [17], with permission from Elsevier; Schematic of extrusion-based bioprinting is taken from Mannoor et.al. [35], with permission from American Chemical Society; Post-processing images are taken from Norotte et.al. [52], with permission from Elsevier.
Applsci 09 00811 g001
Figure 2. Examples of 3D-bioprinted tissue: (A) 3D-printed human ear and sheep menisci [66] (reprinted with permission from American Chemical Society Publications); (B) as-printed aortic valve conduit [62] (reprinted with permission from Wiley).
Figure 2. Examples of 3D-bioprinted tissue: (A) 3D-printed human ear and sheep menisci [66] (reprinted with permission from American Chemical Society Publications); (B) as-printed aortic valve conduit [62] (reprinted with permission from Wiley).
Applsci 09 00811 g002
Figure 3. Scheme for lung tissue engineering [89]. (A) Native adult rat lung is cannulated in the pulmonary artery and trachea for infusion of decellularization solutions. (B) Acellular lung matrix is devoid of cells after 2 to 3 h of treatment. (C) Acellular matrix is mounted inside a biomimetic bioreactor that allows seeding of vascular endothelium into the pulmonary artery and pulmonary epithelium into the trachea. (D) After four to eight days of culture, the engineered lung is removed from the bioreactor and is suitable for implantation into (E) the syngeneic rat recipient. (Reprinted with permission from the American Association for the Advancement of Science (AAAS)).
Figure 3. Scheme for lung tissue engineering [89]. (A) Native adult rat lung is cannulated in the pulmonary artery and trachea for infusion of decellularization solutions. (B) Acellular lung matrix is devoid of cells after 2 to 3 h of treatment. (C) Acellular matrix is mounted inside a biomimetic bioreactor that allows seeding of vascular endothelium into the pulmonary artery and pulmonary epithelium into the trachea. (D) After four to eight days of culture, the engineered lung is removed from the bioreactor and is suitable for implantation into (E) the syngeneic rat recipient. (Reprinted with permission from the American Association for the Advancement of Science (AAAS)).
Applsci 09 00811 g003
Table 1. The comparison among different laser-based bioprinting techniques. CCD—charge-coupled device.
Table 1. The comparison among different laser-based bioprinting techniques. CCD—charge-coupled device.
Laser-Based Bioprinting
CategoryLaser-Induced Forward Transfer (LFT)Absorbing Film-Assisted Laser-Induced Forward Transfer (AFA-LIFT)Biological Laser Processing (BioLP)Matrix-Assisted Pulsed Laser Evaporation Direct Writing (MAPLE-DW)Laser-Guided Direct Writing (LG DW)
DifferenceLaser transparent print ribbonWith thin metal layerWith thick metal layerWith biopolymer layer/
Laser pulsesHigh powerHigh powerHigh powerLow power/
CCD camera//Included//
Optical fiber////Included or not included
AdvantagesOverall
  • High cell viability
  • High resolution
  • High cell densities
  • Low-viscosity cell suspensions
Individual/Thick metal layer reducing the risk of laser energy on cells damageBiopolymer facilitating initial cell attachment/
Disadvantages
  • A risk of photonic cell damage
  • Scalability limitation
  • Fabrication of the laser print ribbon
  • High cost of laser system
  • Complexity of controlling the laser pulses
Table 2. The comparison among droplet-based bioprinting techniques.
Table 2. The comparison among droplet-based bioprinting techniques.
Droplet-based Bioprinting
CategoryInkjet BioprintingElectro-Hydrodynamic Jetting-Based BioprintingAcoustic BioprintingMicrovalve Bioprinting
Continuous InkjetDrop-on-Demand
Trigger DifferencePneumatic actuatorThermal, piezo-electric, electrostatic actuatorElectric fieldAcoustic actuatorPneumatic actuator
Advantages
  • High resolution
  • High printing speed
  • Affordability
  • Cell concentration gradient
  • High resolution
  • High-viscosity bioink
  • Without detrimental stressors
  • High resolution
  • High printing speed
  • Synchronized ejection from different print heads
Disadvantages
  • Low-viscosity bioink
  • Nozzle clogging
  • Droplets cannot be controlled precisely
  • Low-viscosity bioink
  • Nozzle clogging
  • Electric field might affect the long-term cell viability
  • Precise spatial placement of cells is onerous
  • Not too high a viscosity of bioink
  • Not too high a cell concentration
  • Nozzle clogging
  • Low resolution
  • Damage of cells
Table 3. The advantages and disadvantages of extrusion-based bioprinting and stereolithography-based bioprinting. UV—ultraviolet.
Table 3. The advantages and disadvantages of extrusion-based bioprinting and stereolithography-based bioprinting. UV—ultraviolet.
CategoryExtrusion-Based BioprintingStereolithography-Based Bioprinting
Trigger differencePneumatic pressure or mechanical forceLight (usually UV) irradiation
Advantages
  • Scalability
  • High-viscosity bioink
  • High cell concentration
  • Highest resolution
  • Reduced printing time
Disadvantages
  • Lowest resolution
  • Nozzle clogging
  • Shear-thinning bioink
  • Nozzle clogging
  • Photopolymerizable bioinks or bioinks containing UV-activated photo initiated damage of cells
  • UV irradiation damage of DNA and promotion of cell lysis
Table 4. A comparison of various bioprinting techniques as tabulated by Vijayvenkataraman et al. [51].
Table 4. A comparison of various bioprinting techniques as tabulated by Vijayvenkataraman et al. [51].
PropertiesLaser-Based BioprintingInkjet BioprintingEHD Jetting-Based BioprintingAcoustic BioprintingMicrovalve BioprintingExtrusion-Based BioprintingStereolithography-Based Bioprinting
Bioink viscosity1–300 mPa·s3–12 mPa·s1–1000 mPa·sNA1–200 mPa·s~600 kPa·s~5 Pa·s
Cell density108 cells/mL106 cells/mL106 cells/mL106 cells/mL106 cells/mL108 cells/mL>106 cells/mL
Speed200–1600 mm/s10,000 droplets per second10–500 mm/s10,000 droplets per second1000 droplets per second10–50 μm/sHigh
Resolution50 μm50 μm100 nm37 μm100 μm200 nm–6 μm
AccuracyHighMediumLowMediumMediumLowHigh
Cell viability>95%>80%>80%>90%>80%40–95%25–85%
Structural integrityLowLowHighLowLow–mediumHighMedium–high
ScalabilityLowHighHighMediumHighHighMedium–high
CostHighLowHighMedium–highMediumLow–mediumMedium
Table 5. Tissue-engineering applications using 3D bioprinting, adapted from Seol et al. [8] (with permission from Oxford University Press). BMP-2—; FGF-2—; TGF-β—; CNTF—; VEGF—; EGF—; GelMA—gelatin methacryloyl.
Table 5. Tissue-engineering applications using 3D bioprinting, adapted from Seol et al. [8] (with permission from Oxford University Press). BMP-2—; FGF-2—; TGF-β—; CNTF—; VEGF—; EGF—; GelMA—gelatin methacryloyl.
TissueTechniquesCell TypesGrowth FactorsMaterialsReferences
Heart valveExtrusion-based bioprintingAortic valve interstitial cell
Aortic root sinus smooth-muscle cell
Hyaluronic acid
Gelatin
Alginate
[62,68]
Myocardial tissueExtrusion-based bioprintingCardiomyocyte progenitor cellAlginate[69,70]
Blood vesselJetting-based bioprintingEndothelial cell
Smooth-muscle cell
Mesenchymal stem cell
Fibrin[71,72]
Extrusion-based bioprintingEndothelial cell
Cardiac cell
Smooth-muscle cell
Fibroblast
Collagen
Agarose
Alginate
[52,73,74]
Musculo-skeletal tissueJetting-based bioprintingMuscle-derived stem cells
Myoblast
Mesenchymal fibroblast
BMP-2
FGF-2
Fibrin[75,76,77]
Extrusion-based bioprintingBone marrow stromal cell
Endothelial progenitor cell
Endogenous stem cell
TGF-βAgarose
Alginate
Hydroxyapatite
Polycaprolactone
[78,79]
NerveJetting-based bioprintingEmbryonic motor neuron cell
Hippocampal cell
Cortical cell
Neuronal precursor cell
Neural stem cells
CNTF
VEGF
Soy agar
Collagen
Fibrin
[80,81]
Extrusion-based bioprintingBone-marrow stem cell
Schwann cells
Agarose[82,83]
SkinJetting-based bioprintingDermal fibroblast
Epidermal keratinocyte
Collagen[84]
Extrusion-based bioprintingEpitheleal progenitorsEGF BMP-4Gelatin[85]
BoneExtrusion-based bioprintingHuman mesenchymal stem cellsGelMA[86]

Share and Cite

MDPI and ACS Style

Vikram Singh, A.; Hasan Dad Ansari, M.; Wang, S.; Laux, P.; Luch, A.; Kumar, A.; Patil, R.; Nussberger, S. The Adoption of Three-Dimensional Additive Manufacturing from Biomedical Material Design to 3D Organ Printing. Appl. Sci. 2019, 9, 811. https://doi.org/10.3390/app9040811

AMA Style

Vikram Singh A, Hasan Dad Ansari M, Wang S, Laux P, Luch A, Kumar A, Patil R, Nussberger S. The Adoption of Three-Dimensional Additive Manufacturing from Biomedical Material Design to 3D Organ Printing. Applied Sciences. 2019; 9(4):811. https://doi.org/10.3390/app9040811

Chicago/Turabian Style

Vikram Singh, Ajay, Mohammad Hasan Dad Ansari, Shuo Wang, Peter Laux, Andreas Luch, Amit Kumar, Rajendra Patil, and Stephan Nussberger. 2019. "The Adoption of Three-Dimensional Additive Manufacturing from Biomedical Material Design to 3D Organ Printing" Applied Sciences 9, no. 4: 811. https://doi.org/10.3390/app9040811

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop