The Biofabrication of Diseased Artery In Vitro Models
Abstract
:1. Introduction
2. Critical Elements in the Vascular Tissue Microenvironment
2.1. Cells and ECMs in the Artery
2.2. Cell–Cell Crosstalk
2.3. Cell–ECM Interaction
2.4. Physiological Dynamics
3. Conventional Diseased Vascular In Vitro Models
3.1. Designed Apparatus
3.2. Transwell System
3.3. Needle-Templating Microchannels
4. Emerging Biofabrication Techniques
4.1. Tissue-Engineered Blood Vessels
4.2. Organ-on-a-Chip
4.3. Three-Dimensional Bioprinted Organotypic Constructs
4.3.1. Inkjet Bioprinting
4.3.2. Laser-Assisted Bioprinting
4.3.3. Extrusion-Based Bioprinting
4.3.4. Photopolymerization-Based Bioprinting
5. Advanced Arterial Disease In Vitro Models
5.1. Atherosclerosis
5.2. Thrombosis
5.3. Aneurysm
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Angyalossy, V.; Angeles, G.; Pace, M.R.; Lima, A.C.; Dias-Leme, C.L.; Lohmann, L.G.; Madero-Vega, C. An overview of the anatomy, development and evolution of the vascular system of lianas. Plant Ecol. Divers. 2012, 5, 167–182. [Google Scholar] [CrossRef]
- Hosseini, V.; Mallone, A.; Nasrollahi, F.; Ostrovidov, S.; Nasiri, R.; Mahmoodi, M.; Haghniaz, R.; Baidya, A.; Salek, M.M.; Darabi, M.A.; et al. Healthy and diseasedin vitromodels of vascular systems. Lab Chip 2021, 21, 641–659. [Google Scholar] [CrossRef] [PubMed]
- Rajendran, P.; Rengarajan, T.; Thangavel, J.; Nishigaki, Y.; Sakthisekaran, D.; Sethi, G.; Nishigaki, I. The vascular endothelium and human diseases. Int. J. Biol. Sci. 2013, 9, 1057–1069. [Google Scholar] [CrossRef] [Green Version]
- Geovanini, G.R.; Libby, P. Atherosclerosis and inflammation: Overview and updates. Clin. Sci. 2018, 132, 1243–1252. [Google Scholar] [CrossRef] [PubMed]
- Hodgson, J. The pandemic pipeline. Nat. Biotechnol. 2020, 38, 523–532. [Google Scholar] [CrossRef]
- Sartori, S. Tissue engineering approaches in the design of healthy and pathological in vitro tissue models. Front. Bioeng. Biotechnol. 2017, 5, 1–22. [Google Scholar] [CrossRef] [Green Version]
- Sénémaud, J.; Caligiuri, G.; Etienne, H.; Delbosc, S.; Michel, J.B.; Coscas, R. Translational relevance and recent advances of animal models of abdominal aortic aneurysm. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 401–410. [Google Scholar] [CrossRef] [Green Version]
- Patelis, N.; Moris, D.; Schizas, D.; Damaskos, C.; Perrea, D.; Bakoyiannis, C.; Liakakos, T.; Georgopoulos, S. Animal models in the research of Abdominal aortic aneurysms development. Physiol. Res. 2017, 66, 899–915. [Google Scholar] [CrossRef] [PubMed]
- Doherty, E.L.; Aw, W.Y.; Hickey, A.J.; Polacheck, W.J. Microfluidic and organ-on-a-chip approaches to investigate cellular and microenvironmental contributions to cardiovascular function and pathology. Front. Bioeng. Biotechnol. 2021, 9, 1–14. [Google Scholar] [CrossRef] [PubMed]
- Charwat, V.; Egger, D. The third dimension in cell culture: From 2D to 3D culture formats. In Cell Culture Technology; Kasper, C., Charwat, V., Lavrentieva, A., Eds.; Learning Materials in Biosciences; Springer: Cham, Switzerland, 2018; pp. 75–90. [Google Scholar] [CrossRef]
- Hernandez-Gea, V.; Toffanin, S.; Friedman, S.L.; Llovet, J.M. Role of the microenvironment in the pathogenesis and treatment of hepatocellular carcinoma. Gastroenterology 2013, 144, 512–527. [Google Scholar] [CrossRef] [Green Version]
- Urbanczyk, M.; Layland, S.L.; Schenke-Layland, K. The role of extracellular matrix in biomechanics and its impact on bioengineering of cells and 3D tissues. Matrix Biol. 2020, 85–86, 1–14. [Google Scholar] [CrossRef]
- Jayadev, R.; Sherwood, D.R. Basement membranes. Curr. Biol. 2017, 27, R207–R211. [Google Scholar] [CrossRef] [Green Version]
- Mazurek, R.; Dave, J.M.; Chandran, R.R.; Misra, A.; Sheikh, A.Q.; Greif, D.M. Vascular Cells in Blood Vessel Wall Development and Disease, 1st ed.; Elsevier Inc.: Amsterdam, The Netherlands, 2017; Volume 78. [Google Scholar]
- Chistiakov, D.A.; Orekhov, A.N.; Bobryshev, Y.V. Effects of shear stress on endothelial cells: Go with the flow. Acta Physiol. 2017, 219, 382–408. [Google Scholar] [CrossRef] [PubMed]
- Bevan, I.L. Pressure and flow-dependent vascular tone. FASEB J. 2019, 5, 2267–2273. [Google Scholar] [CrossRef] [PubMed]
- Liu, Z.; Khalil, R.A. Evolving mechanisms of vascular smooth muscle contraction highlight key targets in vascular disease. Biochem. Pharmacol. 2018, 153, 91–122. [Google Scholar] [CrossRef]
- Rensen, S.S.M.; Doevendans, P.A.F.M.; Van Eys, G.J.J.M. Regulation and characteristics of vascular smooth muscle cell phenotypic diversity. Neth. Hear. J. 2007, 15, 100–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Majesky, M.W. Adventitia and perivascular cells. Arterioscler. Thromb. Vasc. Biol. 2015, 35, e31–e35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pagano, P.J.; Gutterman, D.D. The adventitia: The outs and ins of vascular disease. Cardiovasc. Res. 2007, 75, 636–639. [Google Scholar] [CrossRef] [Green Version]
- Triggle, C.R.; Samuel, S.M.; Ravishankar, S.; Marei, I.; Arunachalam, G.; Ding, H. The endothelium: Influencing vascular smooth muscle in many ways. Can. J. Physiol. Pharmacol. 2012, 90, 713–718. [Google Scholar] [CrossRef]
- Zeng, Y. Endothelial glycocalyx as a critical signalling platform integrating the extracellular haemodynamic forces and chemical signalling. J. Cell. Mol. Med. 2017, 21, 1457–1462. [Google Scholar] [CrossRef]
- Baker, A.B.; Ettenson, D.S.; Jonas, M.; Nugent, M.A.; Iozzo, R.V.; Edelman, E.R. Endothelial cells provide feedback control for vascular remodeling through a mechanosensitive autocrine TGF-β signaling pathway. Circ. Res. 2008, 103, 289–297. [Google Scholar] [CrossRef] [PubMed]
- Ettenson, D.S.; Koo, E.W.Y.; Januzzi, J.L.; Edelman, E.R. Endothelial heparan sulfate is necessary but not sufficient for control of vascular smooth muscle cell growth. J. Cell. Physiol. 2000, 184, 93–100. [Google Scholar] [CrossRef]
- Nugent, M.A.; Nugent, H.M.; Iozzo, R.V.; Sanchack, K.; Edelman, E.R. Perlecan is required to inhibit thrombosis after deep vascular injury and contributes to endothelial cell-mediated inhibition of intimal hyperplasia. Proc. Natl. Acad. Sci. USA 2000, 97, 6722–6727. [Google Scholar] [CrossRef] [Green Version]
- Yang, K.; Proweller, A. Vascular smooth muscle notch signals regulate endothelial cell sensitivity to angiogenic stimulation. J. Biol. Chem. 2011, 286, 13741–13753. [Google Scholar] [CrossRef] [Green Version]
- Balcells, M.; Martorell, J.; Olivé, C.; Santacana, M.; Chitalia, V.; Cardoso, A.A.; Edelman, E.R. Smooth muscle cells orchestrate the endothelial cell response to flow and injury. Circulation 2010, 121, 2192–2199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chistiakov, D.A.; Orekhov, A.N.; Bobryshev, Y.V. Extracellular vesicles and atherosclerotic disease. Cell. Mol. Life Sci. 2015, 72, 2697–2708. [Google Scholar] [CrossRef] [PubMed]
- Hergenreider, E.; Heydt, S.; Tréguer, K.; Boettger, T.; Horrevoets, A.J.G.; Zeiher, A.M.; Scheffer, M.P.; Frangakis, A.S.; Yin, X.; Mayr, M.; et al. Atheroprotective communication between endothelial cells and smooth muscle cells through miRNAs. Nat. Cell Biol. 2012, 14, 249–256. [Google Scholar] [CrossRef] [PubMed]
- Zheng, B.; Yin, W.N.; Suzuki, T.; Zhang, X.H.; Zhang, Y.; Song, L.L.; Jin, L.S.; Zhan, H.; Zhang, H.; Li, J.S.; et al. Exosome-mediated miR-155 transfer from smooth muscle cells to endothelial cells induces endothelial injury and promotes atherosclerosis. Mol. Ther. 2017, 25, 1279–1294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Theodorou, K.; Boon, R.A. Endothelial cell metabolism in atherosclerosis. Front. Cell Dev. Biol. 2018, 6, 1–9. [Google Scholar] [CrossRef] [Green Version]
- Liao, J.K. Linking endothelial dysfunction with endothelial cell activation. J. Clin. Investig. 2013, 123, 540–541. [Google Scholar] [CrossRef]
- Post, A.; Wang, E.; Cosgriff-Hernandez, E. A Review of integrin-mediated endothelial cell phenotype in the design of cardiovascular devices. Ann. Biomed. Eng. 2019, 47, 366–380. [Google Scholar] [CrossRef] [PubMed]
- Basatemur, G.L.; Jørgensen, H.F.; Clarke, M.C.H.; Bennett, M.R.; Mallat, Z. Vascular smooth muscle cells in atherosclerosis. Nat. Rev. Cardiol. 2019, 16, 727–744. [Google Scholar] [CrossRef] [PubMed]
- Sorokin, V.; Vickneson, K.; Kofidis, T.; Woo, C.C.; Lin, X.Y.; Foo, R.; Shanahan, C.M. Role of vascular smooth muscle cell plasticity and interactions in vessel wall inflammation. Front. Immunol. 2020, 11, 1–16. [Google Scholar] [CrossRef]
- Sforza, D.M.; Putman, C.M.; Cebral, J.R. Hemodynamics of cerebral aneurysms. Annu. Rev. Fluid Mech. 2009, 41, 91–107. [Google Scholar] [CrossRef] [Green Version]
- Chiu, J.J.; Chien, S. Effects of disturbed flow on vascular endothelium: Pathophysiological basis and clinical perspectives. Physiol. Rev. 2011, 91, 327–387. [Google Scholar] [CrossRef] [Green Version]
- Song, H.H.G.; Rumma, R.T.; Ozaki, C.K.; Edelman, E.R.; Chen, C.S. Vascular tissue engineering: Progress, challenges, and clinical promise. Cell Stem Cell 2018, 22, 340–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, J.; Zhou, Y.; Liu, S.; Li, C. Biomechanical signal communication in vascular smooth muscle cells. J. Cell Commun. Signal. 2020, 14, 357–376. [Google Scholar] [CrossRef]
- Remuzzi, A.; Forbes Dewey, C.; Davies, P.F.; Gimbrone, M.A. Orientation of endothelial cells in shear fields in vitro. Biorheology 1984, 21, 617–630. [Google Scholar] [CrossRef]
- Dewey, C.F.; Bussolari, S.R.; Gimbrone, M.A.; Davies, P.F. The dynamic response of vascular endothelial cells to fluid shear stress. J. Biomech. Eng. 1981, 103, 177–185. [Google Scholar] [CrossRef]
- Malek, A.M.; Ahlquist, R.; Gibbons, G.H.; Dzau, V.J.; Izumo, S. A cone-plate apparatus for the in vitro biochemical and molecular analysis of the effect of shear stress on adherent cells. Methods Cell Sci. 1995, 17, 165–176. [Google Scholar] [CrossRef] [Green Version]
- Orr, A.W.; Stockton, R.; Simmers, M.B.; Sanders, J.M.; Sarembock, I.J.; Blackman, B.R.; Schwartz, M.A. Matrix-specific p21-activated kinase activation regulates vascular permeability in atherogenesis. J. Cell Biol. 2007, 176, 719–727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Paschos, N.K.; Brown, W.E.; Eswaramoorthy, R.; Hu, J.C.; Athanasiou, K.A. Advances in tissue engineering through stem cell-based co-culture. Ann. Am. Thorac. Soc. 2015, 9, 488–503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wong, K.H.K.; Truslow, J.G.; Khankhel, A.H.; Chan, K.L.S.; Tien, J. Artificial lymphatic drainage systems for vascularized microfluidic scaffolds. J. Biomed. Mater. Res. Part A 2013, 101, 2181–2190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Noonan, J.; Grassia, G.; MacRitchie, N.; Garside, P.; Guzik, T.J.; Bradshaw, A.C.; Maffia, P. A novel triple-cell two-dimensional model to study immune vascular interplay in atherosclerosis. Front. Immunol. 2019, 10, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Banerjee, J.; Shi, Y.; Azevedo, H.S. In vitro blood–brain barrier models for drug research: State-of-the-art and new perspectives on reconstituting these models on artificial basement membrane platforms. Drug Discov. Today 2016, 21, 1367–1386. [Google Scholar] [CrossRef]
- Hudecz, D.; Rocks, L.; Fitzpatrick, L.W.; Herda, L.M.; Dawson, K.A. Reproducibility in biological models of the blood-brain barrier. Eur. J. Nanomed. 2014, 6, 185–193. [Google Scholar] [CrossRef]
- Islam, K.; Timraz, S.B.; Nasser, R.; Gater, D.L.; Teo, J.C. Co-culture methods used to model atherosclerosis in vitro using endothelial, smooth muscle and monocyte cells. SM J. Biomed. Eng. 2016, 2, 1008. [Google Scholar]
- Wilhelm, I.; Krizbai, I.A. In vitro models of the blood-brain barrier for the study of drug delivery to the brain. Mol. Pharm. 2014, 11, 1949–1963. [Google Scholar] [CrossRef]
- Fomby, P.; Cherlin, A.J.; Hadjizadeh, A.; Doillon, C.J.; Sueblinvong, V.; Weiss, D.J.; Bates, J.H.T.; Gilbert, T.; Liles, W.C.; Lutzko, C.; et al. Stem cells and cell therapies in lung biology and diseases: Conference report. Ann. Am. Thorac. Soc. 2010, 12, 181–204. [Google Scholar] [CrossRef]
- Fang, X.; Neyrinck, A.P.; Matthay, M.A.; Lee, J.W. Allogeneic human mesenchymal stem cells restore epithelial protein permeability in cultured human alveolar type II cells by secretion of angiopoietin-1. J. Biol. Chem. 2010, 285, 26211–26222. [Google Scholar] [CrossRef] [Green Version]
- Stone, N.L.; England, T.J.; O’Sullivan, S.E. A novel transwell blood brain barrier model using primary human cells. Front. Cell. Neurosci. 2019, 13, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bicker, J.; Alves, G.; Fortuna, A.; Falcão, A. Blood-brain barrier models and their relevance for a successful development of CNS drug delivery systems: A review. Eur. J. Pharm. Biopharm. 2014, 87, 409–432. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Fan, B.; Wei, Y.; Suo, X.; Ding, Y. A simple multi-well stretching device to induce inflammatory responses of vascular endothelial cells. Lab Chip 2016, 16, 360–367. [Google Scholar] [CrossRef] [PubMed]
- Gu, X.; Xie, S.; Hong, D.; Ding, Y. An in vitro model of foam cell formation induced by a stretchable microfluidic device. Sci. Rep. 2019, 9, 7461. [Google Scholar] [CrossRef] [PubMed]
- Tien, J.; Wong, K.H.K.; Truslow, J.G. Vascularization of microfluidic hydrogels. Microfluid. Cell Cult. Syst. 2012, 205–221. [Google Scholar] [CrossRef]
- Chrobak, K.M.; Potter, D.R.; Tien, J. Formation of perfused, functional microvascular tubes in vitro. Microvasc. Res. 2006, 71, 185–196. [Google Scholar] [CrossRef]
- Price, G.M.; Tien, J. Subtractive methods for forming microfluidic gels of extracellular matrix proteins. Methods Bioeng. Microdevices Biol. Med. 2009, 235–248. [Google Scholar]
- Hunziker, P.R.; Wolf, M.P.; Wang, X.; Zhang, B.; Marsch, S.; Salieb-Beugelaar, G.B. Construction of programmable interconnected 3D microfluidic networks. J. Micromech. Microeng. 2015, 25, 025018. [Google Scholar] [CrossRef]
- Nichol, J.W.; Koshy, S.T.; Bae, H.; Hwang, C.M.; Yamanlar, S.; Khademhosseini, A. Cell-laden microengineered gelatin methacrylate hydrogels. Biomaterials 2010, 31, 5536–5544. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, D.H.T.; Stapleton, S.C.; Yang, M.T.; Cha, S.S.; Choi, C.K.; Galie, P.A.; Chen, C.S. Biomimetic model to reconstitute angiogenic sprouting morphogenesis in vitro. Proc. Natl. Acad. Sci. USA 2013, 110, 6712–6717. [Google Scholar] [CrossRef] [Green Version]
- Paek, J.; Park, S.E.; Lu, Q.; Park, K.T.; Cho, M.; Oh, J.M.; Kwon, K.W.; Yi, Y.S.; Song, J.W.; Edelstein, H.I.; et al. Microphysiological engineering of self-assembled and perfusable microvascular beds for the production of vascularized three-dimensional human microtissues. ACS Nano 2019, 13, 7627–7643. [Google Scholar] [CrossRef] [PubMed]
- Park, J.H.; Chung, B.G.; Lee, W.G.; Kim, J.; Brigham, M.D.; Shim, J.; Lee, S.; Hwang, C.M.; Durmus, N.G.; Demirci, U.; et al. Microporous cell-laden hydrogels for engineered tissue constructs. Biotechnol. Bioeng. 2010, 106, 138–148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Buchanan, C.F.; Voigt, E.E.; Szot, C.S.; Freeman, J.W.; Vlachos, P.P.; Rylander, M.N. Three-dimensional microfluidic collagen hydrogels for investigating flow-mediated tumor-endothelial signaling and vascular organization. Tissue Eng. Part C Methods 2014, 20, 64–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Polacheck, W.J.; Kutys, M.L.; Yang, J.; Eyckmans, J.; Wu, Y.; Vasavada, H.; Hirschi, K.K.; Chen, C.S. A non-canonical Notch complex regulates adherens junctions and vascular barrier function. Nature 2017, 552, 258–262. [Google Scholar] [CrossRef] [Green Version]
- Pradhan, S.; Banda, O.A.; Farino, C.J.; Sperduto, J.L.; Keller, K.A.; Taitano, R.; Slater, J.H. Biofabrication strategies and engineered in vitro systems for vascular mechanobiology. Adv. Healthc. Mater. 2020, 9, 22–25. [Google Scholar] [CrossRef]
- Papenburg, B.J.; Liu, J.; Higuera, G.A.; Barradas, A.M.C.; de Boer, J.; van Blitterswijk, C.A.; Wessling, M.; Stamatialis, D. Development and analysis of multi-layer scaffolds for tissue engineering. Biomaterials 2009, 30, 6228–6239. [Google Scholar] [CrossRef]
- L’Heureux, N.; Pâquet, S.; Labbé, R.; Germain, L.; Auger, F.A. A completely biological tissue-engineered human blood vessel. FASEB J. 1998, 12, 47–56. [Google Scholar] [CrossRef] [Green Version]
- Sakaguchi, K.; Shimizu, T.; Okano, T. Construction of three-dimensional vascularized cardiac tissue with cell sheet engineering. J. Control. Release 2015, 205, 83–88. [Google Scholar] [CrossRef]
- Goins, A.; Webb, A.R.; Allen, J.B. Multi-layer approaches to scaffold-based small diameter vessel engineering: A review. Mater. Sci. Eng. C 2019, 97, 896–912. [Google Scholar] [CrossRef]
- Yuan, B.; Jin, Y.; Sun, Y.; Wang, D.; Sun, J.; Wang, Z.; Zhang, W.; Jiang, X. A strategy for depositing different types of cells in three dimensions to mimic tubular structures in tissues. Adv. Mater. 2012, 24, 890–896. [Google Scholar] [CrossRef]
- Strobel, H.A.; Hookway, T.A.; Piola, M.; Fiore, G.B.; Soncini, M.; Alsberg, E.; Rolle, M.W. Assembly of tissue-engineered blood vessels with spatially controlled heterogeneities. Tissue Eng. Part A 2018, 24, 1492–1503. [Google Scholar] [CrossRef] [PubMed]
- Gao, G.; Park, W.; Kim, B.S.; Ahn, M.; Chae, S.; Cho, W.W.; Kim, J.; Lee, J.Y.; Jang, J.; Cho, D.W. Construction of a novel in vitro atherosclerotic model from geometry-tunable artery equivalents engineered via in-bath coaxial cell printing. Adv. Funct. Mater. 2021, 31, 1–15. [Google Scholar] [CrossRef]
- Mandenius, C. Conceptual design of micro-bioreactors and organ-on-chips for studies of cell cultures. Bioengineering 2018, 5, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, Q.; Liu, J.; Wang, X.; Feng, L.; Wu, J.; Zhu, X.; Wen, W.; Gong, X. Organ-on-a-chip: Recent breakthroughs and future prospects. Biomed. Eng. Online 2020, 19, 1–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Low, L.A.; Mummery, C.; Berridge, B.R.; Austin, C.P.; Tagle, D.A. Organs-on-chips: Into the next decade. Nat. Rev. Drug Discov. 2021, 20, 345–361. [Google Scholar] [CrossRef]
- Dellaquila, A.; Le Bao, C.; Letourneur, D.; Simon-Yarza, T. In vitro strategies to vascularize 3d physiologically relevant models. Adv. Sci. 2021, 8, 2100798. [Google Scholar] [CrossRef]
- Tsai, M.; Kita, A.; Leach, J.; Rounsevell, R.; Huang, J.N.; Moake, J.; Ware, R.E.; Fletcher, D.A.; Lam, W.A. In vitro modeling of the microvascular occlusion and thrombosis that occur in hematologic diseases using microfluidic technology. J. Clin. Investig. 2012, 122, 408–418. [Google Scholar] [CrossRef] [Green Version]
- Chen, Z.; Lu, J.; Zhang, C.; Hsia, I.; Yu, X.; Marecki, L.; Marecki, E.; Asmani, M.; Jain, S.; Neelamegham, S.; et al. Microclot array elastometry for integrated measurement of thrombus formation and clot biomechanics under fluid shear. Nat. Commun. 2019, 10, 2051. [Google Scholar] [CrossRef] [Green Version]
- Westein, E.; Van Der Meer, A.D.; Kuijpers, M.J.E.; Frimat, J.P.; Van Den Berg, A.; Heemskerk, J.W.M. Atherosclerotic geometries exacerbate pathological thrombus formation poststenosis in a von Willebrand factor-dependent manner. Proc. Natl. Acad. Sci. USA 2013, 110, 1357–1362. [Google Scholar] [CrossRef] [Green Version]
- Günther, A.; Yasotharan, S.; Vagaon, A.; Lochovsky, C.; Pinto, S.; Yang, J.; Lau, C.; Voigtlaender-Bolz, J.; Bolz, S.S. A microfluidic platform for probing small artery structure and function. Lab Chip 2010, 10, 2341–2349. [Google Scholar] [CrossRef]
- Zhang, C.; Neelamegham, S. Application of microfluidic devices in studies of thrombosis and hemostasis. Platelets 2017, 28, 434–440. [Google Scholar] [CrossRef] [PubMed]
- Sarkar, S.; Dadhania, M.; Rourke, P.; Desai, T.A.; Wong, J.Y. Vascular tissue engineering: Microtextured scaffold templates to control organization of vascular smooth muscle cells and extracellular matrix. Acta Biomater. 2005, 1, 93–100. [Google Scholar] [CrossRef]
- Shen, J.Y.; Chan-Park, M.B.; He, B.; Zhu, A.P.; Zhu, X.; Beuerman, R.W.; Yang, E.B.; Chen, W.; Chan, V. Three-dimensional microchannels in biodegradable polymeric films for control orientation and phenotype of vascular smooth muscle cells. Tissue Eng. 2006, 12, 2229–2240. [Google Scholar] [CrossRef] [PubMed]
- Choi, J.S.; Piao, Y.; Seo, T.S. Circumferential alignment of vascular smooth muscle cells in a circular microfluidic channel. Biomaterials 2014, 35, 63–70. [Google Scholar] [CrossRef]
- Benam, K.H.; Dauth, S.; Hassell, B.; Herland, A.; Jain, A.; Jang, K.J.; Karalis, K.; Kim, H.J.; MacQueen, L.; Mahmoodian, R.; et al. Engineered in vitro disease models. Annu. Rev. Pathol. Mech. Dis. 2015, 10, 195–262. [Google Scholar] [CrossRef] [Green Version]
- Zheng, F.; Fu, F.; Cheng, Y.; Wang, C.; Zhao, Y.; Gu, Z. Organ-on-a-chip systems: Microengineering to biomimic living systems. Small 2016, 12, 2253–2282. [Google Scholar] [CrossRef] [PubMed]
- Gold, K.; Gaharwar, A.K.; Jain, A. Emerging trends in multiscale modeling of vascular pathophysiology: Organ-on-a-chip and 3D printing. Biomaterials 2019, 196, 2–17. [Google Scholar] [CrossRef] [PubMed]
- Whitesides, G.M. The origins and the future of microfluidics. Nature 2006, 442, 368–373. [Google Scholar] [CrossRef]
- Zhang, Q.; Bosch-Rué, È.; Pérez, R.A.; Truskey, G.A. Biofabrication of tissue engineering vascular systems. APL Bioeng. 2021, 5, 21507. [Google Scholar] [CrossRef] [PubMed]
- Murphy, S.V.; Atala, A. 3D bioprinting of tissues and organs. Nat. Biotechnol. 2014, 32, 773–785. [Google Scholar] [CrossRef]
- Gao, Q.; Kim, B.; Gao, G. Advanced strategies for 3D bioprinting of tissue and organ analogs using alginate hydrogel bioinks. Marine Drugs 2021, 19, 708. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Derakhshanfar, S.; Mbeleck, R.; Xu, K.; Zhang, X.; Zhong, W.; Xing, M. 3D bioprinting for biomedical devices and tissue engineering: A review of recent trends and advances. Bioact. Mater. 2018, 3, 144–156. [Google Scholar] [CrossRef]
- 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]
- Zhao, L.; Lee, V.K.; Yoo, S.S.; Dai, G.; Intes, X. The integration of 3-D cell printing and mesoscopic fluorescence molecular tomography of vascular constructs within thick hydrogel scaffolds. Biomaterials 2012, 33, 5325–5332. [Google Scholar] [CrossRef] [Green Version]
- Lee, V.K.; Lanzi, A.M.; Ngo, H.; Yoo, S.S.; Vincent, P.A.; Dai, G. Generation of multi-scale vascular network system within 3D hydrogel using 3D bio-printing technology. Cell. Mol. Bioeng. 2014, 7, 460–472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saunders, R.E.; Gough, J.E.; Derby, B. Delivery of human fibroblast cells by piezoelectric drop-on-demand inkjet printing. Biomaterials 2008, 29, 193–203. [Google Scholar] [CrossRef] [PubMed]
- Nakamura, M.; Kobayashi, A.; Takagi, F.; Watanabe, A.; Hiruma, Y.; Ohuchi, K.; Iwasaki, Y.; Horie, M.; Morita, I.; Takatani, S. Biocompatible inkjet printing technique for designed seeding of individual living cells. Tissue Eng. 2005, 11, 1658–1666. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.D.; Choi, J.S.; Kim, B.S.; Chan Choi, Y.; Cho, Y.W. Piezoelectric inkjet printing of polymers: Stem cell patterning on polymer substrates. Polymer 2010, 51, 2147–2154. [Google Scholar] [CrossRef]
- 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]
- Guillotin, B.; Souquet, A.; Catros, S.; Duocastella, M.; Pippenger, B.; Bellance, S.; Bareille, R.; Rémy, M.; Bordenave, L.; Amédéej, 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]
- Arnold, C.B.; Serra, P.; Piqué, A. Laser direct-write techniques for printing of complex materials. MRS Bull. 2007, 32, 23–31. [Google Scholar] [CrossRef] [Green Version]
- Koch, L.; Kuhn, S.; Sorg, H.; Gruene, M.; Schlie, S.; Gaebel, R.; Polchow, B.; Reimers, K.; Stoelting, S.; Ma, N.; et al. Laser printing of skin cells and human stem cells. Tissue Eng. Part C Methods 2010, 16, 847–854. [Google Scholar] [CrossRef] [PubMed]
- Gruene, M.; Deiwick, A.; Koch, L.; Schlie, S.; Unger, C.; Hofmann, N.; Bernemann, I.; Glasmacher, B.; Chichkov, B. Laser printing of stem cells for biofabrication of scaffold-free autologous grafts. Tissue Eng. Part C Methods 2010, 17, 79–87. [Google Scholar] [CrossRef] [PubMed]
- Koch, L.; Deiwick, A.; Schlie, S.; Michael, S.; Gruene, M.; Coger, V.; Zychlinski, D.; Schambach, A.; Reimers, K.; Vogt, P.M.; et al. Skin tissue generation by laser cell printing. Biotechnol. Bioeng. 2012, 109, 1855–1863. [Google Scholar] [CrossRef] [PubMed]
- Guillotin, B.; Guillemot, F. Cell patterning technologies for organotypic tissue fabrication. Trends Biotechnol. 2011, 29, 183–190. [Google Scholar] [CrossRef] [PubMed]
- Memic, A.; Navaei, A.; Mirani, B.; Cordova, J.A.V.; Aldhahri, M.; Dolatshahi-Pirouz, A.; Akbari, M.; Nikkhah, M. Bioprinting technologies for disease modeling. Biotechnol. Lett. 2017, 39, 1279–1290. [Google Scholar] [CrossRef] [Green Version]
- Datta, P.; Dey, M.; Ataie, Z.; Unutmaz, D.; Ozbolat, I.T. 3D bioprinting for reconstituting the cancer microenvironment. NPJ Precis. Oncol. 2020, 4, 1–18. [Google Scholar] [CrossRef]
- Hinton, T.J.; Lee, A.; Feinberg, A.W. 3D bioprinting from the micrometer to millimeter length scales: Size does matter. Curr. Opin. Biomed. Eng. 2017, 1, 31–37. [Google Scholar] [CrossRef]
- Peltola, S.M.; Melchels, F.P.W.; Grijpma, D.W.; Kellomäki, M. A review of rapid prototyping techniques for tissue engineering purposes. Ann. Med. 2008, 40, 268–280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guvendiren, M.; Lu, H.D.; Burdick, J.A. Shear-thinning hydrogels for biomedical applications. Soft Matter. 2012, 8, 260–272. [Google Scholar] [CrossRef]
- 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]
- Chang, R.; Nam, J.; Sun, W. Direct cell writing of 3D microorgan for in vitro pharmacokinetic model. Tissue Eng. Part C Methods 2008, 14, 157–166. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Zhao, X.; Irvine, S.A.; Agrawal, A.; Cao, Y.; Lim, P.Q.; Tan, S.Y.; Venkatraman, S.S. 3D patterned substrates for bioartificial blood vessels—The effect of hydrogels on aligned cells on a biomaterial surface. Acta Biomater. 2015, 26, 159–168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Jang, J.; Park, H.J.; Kim, S.W.; Kim, H.; Park, J.Y.; Na, S.J.; Kim, H.J.; Park, M.N.; Choi, S.H.; Park, S.H.; et al. 3D printed complex tissue construct using stem cell-laden decellularized extracellular matrix bioinks for cardiac repair. Biomaterials 2017, 112, 264–274. [Google Scholar] [CrossRef]
- Pati, F.; Jang, J.; Ha, D.H.; Won Kim, S.; Rhie, J.W.; Shim, J.H.; Kim, D.H.; Cho, D.W. Printing three-dimensional tissue analogues with decellularized extracellular matrix bioink. Nat. Commun. 2014, 5, 3935. [Google Scholar] [CrossRef] [Green Version]
- Jia, W.; Gungor-Ozkerim, P.S.; Zhang, Y.S.; Yue, K.; Zhu, K.; Liu, W.; Pi, Q.; Byambaa, B.; Dokmeci, M.R.; Shin, S.R.; et al. Direct 3D bioprinting of perfusable vascular constructs using a blend bioink. Biomaterials 2016, 106, 58–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Radhakrishnan, J.; Varadaraj, S.; Dash, S.K.; Sharma, A.; Verma, R.S. Organotypic cancer tissue models for drug screening: 3D constructs, bioprinting and microfluidic chips. Drug Discov. Today 2020, 25, 879–890. [Google Scholar] [CrossRef] [PubMed]
- Augustine, R.; Kalva, S.N.; Ahmad, R.; Zahid, A.A.; Hasan, S.; Nayeem, A.; McClements, L.; Hasan, A. 3D Bioprinted cancer models: Revolutionizing personalized cancer therapy. Transl. Oncol. 2021, 14, 101015. [Google Scholar] [CrossRef] [PubMed]
- Knowlton, S.; Onal, S.; Yu, C.H.; Zhao, J.J.; Tasoglu, S. Bioprinting for cancer research. Trends Biotechnol. 2015, 33, 504–513. [Google Scholar] [CrossRef] [PubMed]
- Mosadegh, B.; Xiong, G.; Dunham, S.; Min, J.K. Current progress in 3D printing for cardiovascular tissue engineering. Biomed. Mater. 2015, 10, 34002. [Google Scholar] [CrossRef]
- Vukicevic, M.; Mosadegh, B.; Min, J.K.; Little, S.H. Cardiac 3D Printing and its Future Directions. JACC Cardiovasc. Imaging 2017, 10, 171–184. [Google Scholar] [CrossRef] [PubMed]
- Kang, H.W.; Lee, S.J.; Ko, I.K.; Kengla, C.; Yoo, J.J.; Atala, A. A 3D bioprinting system to produce human-scale tissue constructs with structural integrity. Nat. Biotechnol. 2016, 34, 312–319. [Google Scholar] [CrossRef]
- Massa, S.; Sakr, M.A.; Seo, J.; Bandaru, P.; Arneri, A.; Bersini, S.; Zare-Eelanjegh, E.; Jalilian, E.; Cha, B.H.; Antona, S.; et al. Bioprinted 3D vascularized tissue model for drug toxicity analysis. Biomicrofluidics 2017, 11, 44109. [Google Scholar] [CrossRef] [PubMed]
- Hribar, K.C.; Soman, P.; Warner, J.; Chung, P.; Chen, S. Light-assisted direct-write of 3D functional biomaterials. Lab Chip 2014, 14, 268–275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Morris, V.B.; Nimbalkar, S.; Younesi, M.; McClellan, P.; Akkus, O. Mechanical Properties, Cytocompatibility and manufacturability of chitosan: PEGDA hybrid-gel scaffolds by stereolithography. Ann. Biomed. Eng. 2017, 45, 286–296. [Google Scholar] [CrossRef] [PubMed]
- Pagac, M.; Hajnys, J.; Ma, Q.P.; Jancar, L.; Jansa, J.; Stefek, P.; Mesicek, J. A review of vat photopolymerization technology: Materials, applications, challenges, and future trends of 3d printing. Polymers 2021, 13, 598. [Google Scholar] [CrossRef] [PubMed]
- Wei, L.N.G.; Jia, M.L.; Zhou, M.M.; Chen, Y.W.; Lee, K.X.A.; Yeong, W.Y.; Shen, Y.F. Vat polymerization-based bioprinting—Process, materials, applications and regulatory challenges. Biofabrication 2020, 12, 22001. [Google Scholar] [CrossRef]
- Tang, M.; Rich, J.N.; Chen, S. Biomaterials and 3D bioprinting strategies to model glioblastoma and the blood–brain barrier. Adv. Mater. 2021, 33, 1–25. [Google Scholar] [CrossRef] [PubMed]
- Ma, X.; Qu, X.; Zhu, W.; Li, Y.S.; Yuan, S.; Zhang, H.; Liu, J.; Wang, P.; Lai, C.S.E.; Zanella, F.; et al. Deterministically patterned biomimetic human iPSC-derived hepatic model via rapid 3D bioprinting. Proc. Natl. Acad. Sci. USA 2016, 113, 2206–2211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tiwari, A.P.; Thorat, N.D.; Pricl, S.; Patil, R.M.; Rohiwal, S.; Townley, H. Bioink: A 3D-bioprinting tool for anticancer drug discovery and cancer management. Drug Discov. Today 2021, 26, 1574–1590. [Google Scholar] [CrossRef] [PubMed]
- Hribar, K.C.; Finlay, D.; Ma, X.; Qu, X.; Ondeck, M.G.; Chung, P.H.; Zanella, F.; Engler, A.J.; Sheikh, F.; Vuori, K.; et al. Nonlinear 3D projection printing of concave hydrogel microstructures for long-term multicellular spheroid and embryoid body culture. Lab Chip 2015, 15, 2412–2418. [Google Scholar] [CrossRef] [Green Version]
- Ye, W.; Li, H.; Yu, K.; Xie, C.; Wang, P.; Zheng, Y.; Zhang, P.; Xiu, J.; Yang, Y.; Zhang, F.; et al. 3D printing of gelatin methacrylate-based nerve guidance conduits with multiple channels. Mater. Des. 2020, 192, 108757. [Google Scholar] [CrossRef]
- Grigoryan, B.; Paulsen, S.J.; Corbett, D.C.; Sazer, D.W.; Fortin, C.L.; Zaita, A.J.; Greenfield, P.T.; Calafat, N.J.; Gounley, J.P.; Ta, A.H.; et al. Multivascular networks and functional intravascular topologies within biocompatible hydrogels. Science 2019, 364, 458–464. [Google Scholar] [CrossRef]
- Park, J.H.; Jang, J.; Lee, J.S.; Cho, D.W. Current advances in three-dimensional tissue/organ printing. Tissue Eng. Regen. Med. 2016, 13, 612–621. [Google Scholar] [CrossRef]
- Shami, A.; Gonçalves, I.; Hultgardh-Nilsson, A. Collagen and related extracellular matrix proteins in atherosclerotic plaque development. Curr. Opin. Lipidol. 2014, 25, 394–399. [Google Scholar] [CrossRef]
- Ozawa, N.; Sato, Y.; Mori, Y.; Masuda, H.; Yamane, M.; Yamamoto, Y.; Shirai, R.; Watanabe, R.; Sato, K.; Mori, Y.; et al. Legumain promotes atherosclerotic vascular remodeling. Int. J. Mol. Sci. 2019, 20, 2195. [Google Scholar] [CrossRef] [Green Version]
- Grover, S.P.; Mackman, N. Tissue factor in atherosclerosis and atherothrombosis. Atherosclerosis 2020, 307, 80–86. [Google Scholar] [CrossRef]
- Förstermann, U.; Xia, N.; Li, H. Roles of vascular oxidative stress and nitric oxide in the pathogenesis of atherosclerosis. Circ. Res. 2017, 120, 713–735. [Google Scholar] [CrossRef] [PubMed]
- Benjamin, E.J.; Blaha, M.J.; Chiuve, S.E.; Cushman, M.; Das, S.R.; Deo, R.; De Ferranti, S.D.; Floyd, J.; Fornage, M.; Gillespie, C.; et al. Heart Disease and Stroke Statistics’2017 Update: A Report from the American Heart Association. Circulation 2017, 135, e146–e603. [Google Scholar] [CrossRef] [PubMed]
- Libby, P.; Buring, J.E.; Badimon, L.; Hansson, G.K.; Deanfield, J.; Bittencourt, M.S.; Tokgözoğlu, L.; Lewis, E.F. Atherosclerosis. Nat. Rev. Dis. Prim. 2019, 5, 1–18. [Google Scholar] [CrossRef]
- Savoji, H.; Mohammadi, M.H.; Rafatian, N.; Toroghi, M.K.; Wang, E.Y.; Zhao, Y.; Korolj, A.; Ahadian, S.; Radisic, M. Cardiovascular disease models: A game changing paradigm in drug discovery and screening. Biomaterials 2019, 198, 3–26. [Google Scholar] [CrossRef] [PubMed]
- Zheng, W.; Huang, R.; Jiang, B.; Zhao, Y.; Zhang, W.; Jiang, X. An early-stage atherosclerosis research model based on microfluidics. Small 2016, 12, 2022–2034. [Google Scholar] [CrossRef]
- Truskey, G.A. Endothelial vascular smooth muscle cell coculture assay for high throughput screening assays to identify antiangiogenic and other therapeutic molecules. Int. J. High Throughput Screen. 2010, 2010, 171. [Google Scholar] [CrossRef] [Green Version]
- Robert, J.; Weber, B.; Frese, L.; Emmert, M.Y.; Schmidt, D.; Von Eckardstein, A.; Rohrer, L.; Hoerstrup, S.P. A three-dimensional engineered artery model for in vitro atherosclerosis research. PLoS ONE 2013, 8, e79821. [Google Scholar] [CrossRef] [Green Version]
- Mallone, A.; Gericke, C.; Hosseini, V.; Chahbi, K.; Haenseler, W.; Emmert, M.Y.; Eckardstein, A.; Walther, J.H.; Vogel, V. Human induced pluripotent stem cell-derived vessels as dynamic atherosclerosis model on a chip. bioRxiv 2021, 2020-11. [Google Scholar] [CrossRef]
- Su, C.; Menon, N.V.; Xu, X.; Teo, Y.R.; Cao, H.; Dalan, R.; Tay, C.Y.; Hou, H.W. A novel human arterial wall-on-a-chip to study endothelial inflammation and vascular smooth muscle cell migration in early atherosclerosis. Lab Chip 2021, 21, 2359–2371. [Google Scholar] [CrossRef]
- Furie, B.; Furie, B.C. Mechanisms of thrombus formation. Mechanisms of Disease. N. Engl. J. Med. 2008, 359, 938–949. [Google Scholar] [CrossRef]
- Byrnes, J.R.; Wolberg, A.S. Red blood cells in thrombosis. Blood 2017, 130, 1795–1799. [Google Scholar] [CrossRef] [PubMed]
- Turpie, A.G.G.; Esmon, C. Venous and arterial thrombosis—Pathogenesis and the rationale for anticoagulation. Thromb. Haemost. 2011, 105, 586–596. [Google Scholar] [CrossRef] [PubMed]
- Pandian, N.K.R.; Mannino, R.G.; Lam, W.A.; Jain, A. Thrombosis-on-a-chip: Prospective impact of microphysiological models of vascular thrombosis. Curr. Opin. Biomed. Eng. 2018, 5, 29–34. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.S.; Davoudi, F.; Walch, P.; Manbachi, A.; Luo, X.; Dell’Erba, V.; Miri, A.K.; Albadawi, H.; Arneri, A.; Li, X.; et al. Bioprinted thrombosis-on-a-chip. Lab Chip 2016, 16, 4097–4105. [Google Scholar] [CrossRef] [Green Version]
- Jain, A.; van der Meer, A.D.; Papa, A.L.; Barrile, R.; Lai, A.; Schlechter, B.L.; Otieno, M.A.; Louden, C.S.; Hamilton, G.A.; Michelson, A.D.; et al. Assessment of whole blood thrombosis in a microfluidic device lined by fixed human endothelium. Biomed. Microdev. 2016, 18, 1–7. [Google Scholar] [CrossRef] [Green Version]
- Zheng, Y.; Chen, J.; Craven, M.; Choi, N.W.; Totorica, S.; Diaz-Santana, A.; Kermani, P.; Hempstead, B.; Fischbach-Teschl, C.; López, J.A.; et al. In vitro microvessels for the study of angiogenesis and thrombosis. Proc. Natl. Acad. Sci. USA 2012, 109, 9342–9347. [Google Scholar] [CrossRef] [Green Version]
- Costa, P.F.; Albers, H.J.; Linssen, J.E.A.; Middelkamp, H.H.T.; Van Der Hout, L.; Passier, R.; Van Den Berg, A.; Malda, J.; Van Der Meer, A.D. Mimicking arterial thrombosis in a 3D-printed microfluidic: In vitro vascular model based on computed tomography angiography data. Lab Chip 2017, 17, 2785–2792. [Google Scholar] [CrossRef] [Green Version]
- Pahlavan, P.S.; Niroomand, F. Coronary artery aneurysm: A review. Clin. Cardiol. 2006, 29, 439–443. [Google Scholar] [CrossRef]
- Matta, A.G.; Yaacoub, N.; Nader, V.; Moussallem, N.; Carrie, D.; Roncalli, J. Coronary artery aneurysm: A review. World J. Cardiol. 2021, 13, 446–455. [Google Scholar] [CrossRef]
- Shelton, K.R.; Ali, A.T.; Moursi, M.M.; Eidt, J.F. Pseudoaneurysm: A review. Curr. Treat. Options Cardiovasc. Med. 2008, 10, 173–183. [Google Scholar] [CrossRef]
- Chalouhi, N.; Hoh, B.L.; Hasan, D. Review of cerebral aneurysm formation, growth, and rupture. Stroke 2013, 44, 3613–3622. [Google Scholar] [CrossRef] [PubMed]
- Lopez-Candales, A.; Kleiger, R.E.; Aleman-Gomez, J.; Kouchoukos, N.T.; Botney, M.D. Pulmonary artery aneurysm: Review and case report. Clin. Cardiol. 1995, 18, 738–740. [Google Scholar] [CrossRef] [PubMed]
- Aggarwal, S.; Qamar, A.; Sharma, V.; Sharma, A. Abdominal aortic aneurysm: A comprehensive review. Exp. Clin. Cardiol. 2011, 16, 11–15. [Google Scholar] [CrossRef]
- Meng, H.; Wang, Z.; Hoi, Y.; Gao, L.; Metaxa, E.; Swartz, D.D.; Kolega, J. Complex hemodynamics at the apex of an arterial bifurcation induces vascular remodeling resembling cerebral aneurysm initiation. Stroke 2007, 38, 1924–1931. [Google Scholar] [CrossRef] [PubMed]
- Cebral, J.; Ollikainen, E.; Chung, B.J.; Mut, F.; Sippola, V.; Jahromi, B.R.; Tulamo, R.; Hernesniemi, J.; Niemelä, M.; Robertson, A.; et al. Flow conditions in the intracranial aneurysm lumen are associated with inflammation and degenerative changes of the aneurysm wall. Am. J. Neuroradiol. 2017, 38, 119–126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Epshtein, M.; Levi, M.; Kraitem, A.M.; Zidan, H.; King, R.M.; Gawaz, M.; Gounis, M.J.; Korin, N. Biophysical targeting of high-risk cerebral aneurysms. Bioeng. Transl. Med. 2021, 7, e10251. [Google Scholar] [CrossRef]
- Osgood, M.L. Aneurysmal subarachnoid hemorrhage: Review of the pathophysiology and management strategies. Curr. Neurol. Neurosci. Rep. 2021, 21, 50. [Google Scholar] [CrossRef]
- Xiang, J.; Natarajan, S.K.; Tremmel, M.; Ma, D.; Mocco, J.; Hopkins, L.N.; Siddiqui, A.H.; Levy, E.I.; Meng, H. Hemodynamic-morphologic discriminants for intracranial aneurysm rupture. Stroke 2011, 42, 144–152. [Google Scholar] [CrossRef] [Green Version]
- Kaneko, N.; Mashiko, T.; Namba, K.; Tateshima, S.; Watanabe, E.; Kawai, K. A patient-specific intracranial aneurysm model with endothelial lining: A novel in vitro approach to bridge the gap between biology and flow dynamics. J. Neurointerv. Surg. 2018, 10, 306–309. [Google Scholar] [CrossRef]
- Mannino, R.G.; Myers, D.R.; Ahn, B.; Wang, Y.; Margo, R.; Gole, H.; Lin, A.S.; Guldberg, R.E.; Giddens, D.P.; Timmins, L.H.; et al. Do-it-yourself in vitro vasculature that recapitulates in vivo geometries for investigating endothelial-blood cell interactions. Sci. Rep. 2015, 5, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Hosseini, V.; Mallone, A.; Mirkhani, N.; Noir, J.; Salek, M.; Pasqualini, F.S.; Schuerle, S.; Khademhosseini, A.; Hoerstrup, S.P.; Vogel, V. A pulsatile flow system to engineer aneurysm and atherosclerosis mimetic extracellular matrix. Adv. Sci. 2020, 7, 1–14. [Google Scholar] [CrossRef] [PubMed]
- Medero, R.; Ruedinger, K.; Rutkowski, D.; Johnson, K.; Roldán-Alzate, A. In vitro assessment of flow variability in an intracranial aneurysm model using 4D flow MRI and tomographic PIV. Ann. Biomed. Eng. 2020, 48, 2484–2493. [Google Scholar] [CrossRef] [PubMed]
- Jang, L.K.; Alvarado, J.A.; Pepona, M.; Wasson, E.M.; Nash, L.D.; Ortega, J.M.; Randles, A.; Maitland, D.J.; Moya, M.L.; Hynes, W.F. Three-dimensional bioprinting of aneurysm-bearing tissue structure for endovascular deployment of embolization coils. Biofabrication 2020, 13, 15006. [Google Scholar] [CrossRef] [PubMed]
- Gao, R.; Tian, X.; Li, Q.; Song, X.; Shao, B.; Zeng, J.; Liu, Z.; Zhi, D.; Zhao, G.; Xia, H.; et al. Artificial blood vessel frameworks from 3d printing-based super-assembly as in vitro models for early diagnosis of intracranial aneurysms. Chem. Mater. 2020, 32, 3188–3198. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Pan, C.; Gao, Q.; Kim, B.-S.; Han, Y.; Gao, G. The Biofabrication of Diseased Artery In Vitro Models. Micromachines 2022, 13, 326. https://doi.org/10.3390/mi13020326
Pan C, Gao Q, Kim B-S, Han Y, Gao G. The Biofabrication of Diseased Artery In Vitro Models. Micromachines. 2022; 13(2):326. https://doi.org/10.3390/mi13020326
Chicago/Turabian StylePan, Chen, Qiqi Gao, Byoung-Soo Kim, Yafeng Han, and Ge Gao. 2022. "The Biofabrication of Diseased Artery In Vitro Models" Micromachines 13, no. 2: 326. https://doi.org/10.3390/mi13020326
APA StylePan, C., Gao, Q., Kim, B. -S., Han, Y., & Gao, G. (2022). The Biofabrication of Diseased Artery In Vitro Models. Micromachines, 13(2), 326. https://doi.org/10.3390/mi13020326