Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2497 2022-11-21 05:39:37 |
2 layout + 1 word(s) 2498 2022-11-21 07:46:16 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Wang, X.;  Chan, V.;  Corridon, P.R. Acellular Tissue-Engineered Vascular Grafts from Polymers. Encyclopedia. Available online: https://encyclopedia.pub/entry/35418 (accessed on 28 April 2024).
Wang X,  Chan V,  Corridon PR. Acellular Tissue-Engineered Vascular Grafts from Polymers. Encyclopedia. Available at: https://encyclopedia.pub/entry/35418. Accessed April 28, 2024.
Wang, Xinyu, Vincent Chan, Peter R. Corridon. "Acellular Tissue-Engineered Vascular Grafts from Polymers" Encyclopedia, https://encyclopedia.pub/entry/35418 (accessed April 28, 2024).
Wang, X.,  Chan, V., & Corridon, P.R. (2022, November 21). Acellular Tissue-Engineered Vascular Grafts from Polymers. In Encyclopedia. https://encyclopedia.pub/entry/35418
Wang, Xinyu, et al. "Acellular Tissue-Engineered Vascular Grafts from Polymers." Encyclopedia. Web. 21 November, 2022.
Acellular Tissue-Engineered Vascular Grafts from Polymers
Edit

Vascular tissue engineering (VTE) lies at the intersection of several emerging disciplines including material science, polymers, stem cell biology, and fabrication technologies to support the development of micro/macroscopic artificial and bioartificial vessels.

acellular multiscale tissue-engineered vascular grafts

1. Introduction

Vascular malfunctions contribute to various maladies and have emerged as a leading cause of global mortality. Specifically, as much as 32% of deaths worldwide were attributed to cardiovascular diseases (CVD) in 2019 alone [1]. At the same time, CVD and associated risk factors are causing substantial morbidity in patients worldwide [2]. Current treatments for CVD include medications, surgery, medical implants, mechanical devices, and rehabilitation [3][4][5]. Moreover, conventional surgical procedures such as grafting bypass, improve vessel patency, and vascular repair treats more severe conditions of CVDs, e.g., stroke and heart attack [6]. Specifically, innovative approaches like vascular tissue engineering (VTE) have been intensively explored to address the pathophysiology underlying CVD progression and improve the overall life quality of CVD patients through the direct replacement of damaged vessels [7][8].
Currently, VTE lies at the intersection of several emerging disciplines including material science, polymers, stem cell biology, and fabrication technologies to support the development of micro/macroscopic artificial and bioartificial vessels [9][10][11][12]. Different classes of vascular tissue equivalents have been successfully developed as potential replacements for damaged or malfunctioning blood vessels through advancements in human cell biology and cardiovascular physiology [13]. To engineer such artificial blood vessels for in vivo transplantation in patients or in vitro models of vascular pathophysiology, appropriate polymeric materials, cell culture technology, controlled microenvironment, and additive manufacturing are required to develop vascular scaffolds with varied complexity [11][13][14][15][16][17].
Several natural and synthetic polymers have been applied to fabricate biodegradable and biocompatible vascular scaffolds through combinations of chemical processing and manufacturing technologies such as hydrogelation [18], 3D bio-printing, electrospinning, casting/molding, laser degradation, phase inversion, sheet-based fabrication, medical textile (braiding/weaving/knitting), and gas foaming [19][20][21][22][23][24]. For instance, vascular scaffolds composed of poly(ε-caprolactone) (PCL) and collagen fabricated by electrospinning has shown higher durability than sole-PCL/poly (lactide-co-glycolide (PLGA) scaffolds [25]. The PCL/collagen composite scaffolds could bear long-term high pressure caused by loaded-volume blood flow and provide a favorable environment for vascular cell growth [26].
Scientists have also used poly-L-lactic acid (PLLA)/PCL added with heparin to create small-scale vessel substitutes through electrospinning and extrusion [27][28]. Apart from these examples, other polymers such as PU [29], gelatin [30], chitosan [31], PVA [32], PEG [33][34], PLCL [35][36][37], PGA [38], and PET [39] have been utilized to create multi-scale blood vessel replacements. The general strategy combines two or more of those polymers and fabricates them via the techniques above [14]. Moreover, these polymer-based tissue-engineered vascular grafts (TEVG) have enhanced biomechanical properties, which can better withstand in vivo blood pressures and establish sustainable cellular environments over long periods. Nevertheless, TEVGs must mechanically match the region of interest for transplantation purposes to provide the necessary degree of structural integrity, biocompatibility, biodegradability, and physiology functions [21]. Based on the numerous conditions that must be satisfied, research is still required to optimize TEVG technologies.

2. Characterization of Synthetic TEVGs in Clinical Use

Polyethylene terephthalate (PET, Dacron), expanded polytetrafluoroethylene (ePTFE), and polyurethane (PU) are the three major TVEGs that are invested in clinical use [40][41][42][43]. Clinically available Dacron grafts are fabricated via either weaving or knitting in an over-and-under pattern, leading to minimal porosity and creep [44]. Dacron is stable and can persist for more than 10 years after implantation without significant deterioration when applied as macro-scale vascular replacements. They have poor clinical performance and cause thrombus, inflammation, and compliance mismatches when used as small-diameter vascular grafts [45][46]. The compliance of current commercial Dacron TEVGs is 2.0 × 10−2% mmHg−1 with 42% of two-year patency [47].
Polytetrafluoroethylene (PTFE) was patented first in 1937 as Teflon. Expanded ePTFE (Gore-Tex) is the material employed on vascular grafts and manufactured using heating, stretching, and extruding processes, creating a microporous scaffold for firm cell adhesion [41][48]. An ePTEE vascular graft is non-woven, with a node–fibril structure, and performs well as aortic replacements having a 5-year primary patency rate of 91% to 95% but a lower patency rate for being analogs of substitutes with small ID [49][50]. The compliance of ePTEE is 1.5 × 10−2% mmHg−1 with 42% of two-year patency [51]. Specifically, both Dacron and ePTEE can be bonded to heparin [52]. Heparin-bonded ePTFE aortic grafts presented decreased thrombogenicity and enhanced patency rates at 8 weeks [53]. Heparin-bonded Dacron grafts are commercially available in Europe [44]. Significantly, the heparin-bonded Dacron showed promising wide application of SDVGs such as femoropopliteal bypass grafting, with eye-catching patency rates at 1, 2, and 3 years of 70%, 63%, and 55%, respectively [54].
Researchers prefer using PU for microcapillary scaffolds due to their microstructure [55]. Polyurethanes can be divided into fibrillar or foamy structures, and both tend to lack communicating spaces for potential capillary ingrowth [56][57]. In microporous foamy PU with a 15 μm pore size, relatively little capillary ingrowth can be achieved. Whereas once the pore size increased up to 157 μm, capillary sprouting occurred [58][59]. Although PU grafts possess many exciting features, such as EC growth under inferior hemodynamic conditions, excellent healing, subtle surgical handling, and low suture bleeding, sufficient evidence of the spread use of PU vascular grafts as human peripheral bypasses remains in scarcity because of lacking investigations [60].

3. Key Challenges Limiting the Translation of Polymer-Based TEVGs

Ideally, bioartificial blood vessels should possess the structural and functional capacities of native structures [61]. Therefore, identifying the conditions that may lead to deviation from these ideal characteristics is vital for reducing the potential of device failure. It is also essential that these structures be rendered with bio-inertness for supporting somatic growth post-transplantation [62]. To this end, pinpointing the key challenges the current polymeric TEVGs face in clinical translation is extremely necessary.
As we all know, the endothelium is essential in restricting the movement of water, cells, and protein between intravascular and interstitial compartments [63][64]. Based on the characterization demonstrated in below table, TEVGs solely composed of natural polymers have better performance regarding biological aspects [61]. These microscale vascular conduits are free of considerations regarding biocompatibility, degradability, and cytotoxicity. They are highly supportive of cell repopulation and nutrition exchange. Besides, different natural polymers will create vascular substitutes with specific physical performance. For instance, collagen type I exhibited a vital barrier function after cell seeding.
Moreover, the endothelium has to align on the basement membrane, where collagen type I is the essential component and regulator [18][65]. This characteristic explains why vascular replacements consisting of collagen type I have a vital barrier function and indicates the potential for endothelium regeneration [66]. However, the mechanical properties of these natural polymer scaffolds require significant improvement. Going back to collagen type I, the stiffness of collagen type I is 0.1–18 kPa when the concentration is 3–20 mg/mL [67]. Based on the fact that compliance is the inverse of stiffness [68], the compliance of collagen type I is around 10−2 cm/s. The compliance of vascular conduits made by collagen type I conducted with microfluid/hydrogelation is close to 10−6 cm/s. The compliance of native micro-vessels with the same dimension is unknown, but the compliance of this polymer has been highly reduced when formed into microvascular constructs.
However, the mechanical properties of polymers are flexible and changeable by distinct ways of fabrication, physical/chemical reactions, and incorporation with other materials [69]. The HA vascular micro-tubes in Table have a stiffness from 19 to 32 kPa, while, when it combines with PVA as a composite hydrogel, the stiffness can be extended to 200 kPa [70]. Other similar examples provide future research directions on amplifying the mechanical properties of natural polymer-based vascular homologs but also bring new challenges of choosing to fabricate techniques, a combination of polymers, and methods of modifications [69]. These problems and confusion can only be solved with arduous academic work. Besides that, the mechanical properties of these natural polymeric vascular substitutes still need to be discovered, which implies a shortage of small- and macro-scale vessel analogs generated by natural polymers.
For vascular scaffolds created by synthetic polymers, their dimensions become multiple at the micro-scale level, and the small ID vascular structures have been formed through the braiding of PET/PLGA and the casting/electrospinning of PLGA/ P(CL/LA). Except for this, the morphology of blood vessel conduits is not limited only by straight but also by branched tubes [69]. Most scaffolds’ mechanical features are available and are highly hopeful of reaching that of native blood vessels, as listed in Table 2. For example, in Table 2, the saphenous vein’s longitudinal elastic modulus (stiffness) can be 130 or 23.7 MPa. The mean diameter of the usual great saphenous vein (GSV) is 5.0 ± 2.4 mm. The mean diameter of a typical small saphenous vein (SSV) is 3.1 ± 1.3 mm [71].
Regarding the dimensions, various polymers and corresponding fabrication skills presented in Table 1 can meet the requirement, such as silicone, PU, heparin-releasing PLLA/PCL, and PEG/collagen/PU. For the stiffness, 10% (w/v) P(CL/LA)/PGLA (sealed) and 15% (w/v) P(CL/LA)/PGLA (sealed) are capable of matching the stiffness of native small saphenous with 23.7 MPa [69]. However, most synthetic polymers’ stiffness lies in the range of kPa. Apart from that, polymers’ suture retention strength and burst pressure are still predominantly lower than native vessels. Compared to single synthetic-polymer-made scaffolds, a mixture of polymers with or without biological molecules/natural polymers demonstrated potential neovascularization ability [72]. Therefore, new challenges arise in this field, and these issues are becoming more specific and detailed. How do we control the components’ percentage of composite polymers to optimize biomechanical properties? How can we choose suitable polymer partners among hundreds of polymer families? The methods and choices are increasing, but at the same time, the complexity of studies and characterization of those synthetic polymer-based scaffolds are also being augmented. Similar to vascular replacements created by natural polymers, more exploration and studies should be conducted and established to develop acellular vessel prostheses with small- and macro-dimensions.
Table 1. Polymer-Based TEVGs and Characterizations.
Polymers Applied Technology Characterization References
Natural Polymers Collagen type I Hydrogelation/microfluid; Strong barrier function after being seeded with human vascular cells; compliance coefficient of BSA: 5.5 × 10−6 ± 3.5 × 10−6 cm/s (n = 3) at days 3–4 and 7.9 × 10−6 ± 3.5 × 10−6 cm/s at days 6–7;
ID = 116 μm
[18][63]
Hydrogelation/laser degradation D = 50 μm [73][74]
Gelatin Hydrolyzation/microfluid Good fluidic access and cytocompatibility to murine mammary epithelial cells; microscale [30]
Silk Braiding Implanted as a rodent abdominal aorta with ECs/SMCs migration and alignment observed;
ID = 1.5 mm
[75]
Polysaccharides: HA Molding/microfluid/hydrogelation Efficient delivery of nutrients
Stiffness: 19–32 kPa; microscale
[76]
Polysaccharides: alginate/Cacl2 (addition) Extrusion/injection 3D printing Stiffness < 500 kPa; short maturation of SMCs;
D = 1–3 mm BT; D = 2 μm ST
L = 2 μm
T = 2 μm
[77]
Fibrin 3D-quasi microfluid Strong ADSCs attachment, regrowth, and differentiation; microscale [78]
Synthetic Polymers PCL/PVA Extrusion3D printing Porosity: 61% with strand space 0.7 mm; 74% with strand space 1 mm;
D = 2–4 mm BT
[32]
PCL/chitosan Electrospinning/extrusion 3D printing ST [31]
PCL/GelMA-gellan/alginate Extrusion 3D printing D = 4 mm ST [32]
PDMS/fibrin A tissue ring of SMCs after being seeded with HASMCs;
D = 5 mm ST
[79]
Silicone Stiffness: 20–244.78 kPa;
Support culturing of HUVECs, HA-VSMCs, HDF-n;
D = 0.5–2 mm ST
[80]
PU DLP 3D printing EM = 1.1 MPa No cytotoxicity at highest concentration 26 mgL−1;
ID = 1.5 mm OD = 4 mm ST
[29]
PPF P = 0.35 nm for ID = 2.5 mm support cell culturing of HUVECs, hMSCs, HUSMCs;
ID = 2.5 or 1 mm t = 0.25 or 0.15 mm ST
[81] [82]
PTHD-DA SLA 3D printing ID = 18 μm T = 3 μm L = 160 μm BT
ID = 2 μm T = 2 mm L = 2 mm ST
[83]
2PP 3D printing [83]
Heparin-releasing PLLA/PCL Electrospinning/extrusion 3D printing D = 5 mm L = 6 cm ST [27][28]
PGS/PCL/salt Casting/molding SRS = 0.45 ± 0.031 N, EM = 536 ± 119 kPa UTS = 3790 ± 1450 kPa BP = 2360 ± 673 mmHg C = 11% ± 2.2%, transplanted as rat abdominal aorta with progressive vascular remolding in 3 months;
ID = 720 μm T = 290 μm
[84]
10% (w/v) P(CL/LA)/PGLA (sealed) Casting/electrospinning SRS = 2.16 ± 0.037 N EM = 17.73 ± 3.09 MPa UTS = 2.93 ± 0.26 MPa BP = 1002.17 ± 181.98 mmHg, support HUVECs’ attachment and proliferation;
ID = 1.02 ± 0.5 cm
T = 0.21 ± 0.02 cm
[85]
15% (w/v) P(CL/LA)/PGLA (sealed) SRS = 3.20 ± 0.577 N EM = 26.90 ± 6.66 MPa UTS = 4.75 ± 0.97 MPa BP = 1321.66 ± 214.67 mmHg support HUVECs’ attachment and proliferation;
ID = 1.01 ± 0.08 cm
T = 0.19 ± 0.09 cm
PLCL (inner layer)/PGA/PLA (outer layer) Casting/electrospinning Cell infiltration in scaffold observed, transplanted as infrarenal aortic graft in mice, maintaining 8-month survival;
Outer layer ID = 600 μm, inner layer ID = 200 μm
T = 3 mm
[38]
PEGDA LD Elongated microchannels and molecule transportation between unconnected microchannels observed; support HUVECs’ seeding;
Microcapillary
[73][74]
PU/gelatin PI P = 2 μm PE = 1.2 ± 0.4 mLmin−1 UTS = 2700 ± 400 kPa
Support hMSCs’ adhesion and growth
[86]
PLLA/inner MSCs Sheet-based fabrication Patency of 100% in 8.6 weeks; vascular remolding observed, SMCs alignment in 60 days;
ID = 0.7 mm
[35][36][37]
PLCL/FB/collagen 4-week transplantation, patency unknown;
ID = 4.1 mm
PET/PLGA Braiding Small ID [39]
Polyester/PTT weaving EM = 1056 MPa under pressure 200 mmHg [87]
Spandex (over 80% PU)/polyester knitting Transplanted as dog abdominal aorta;
D = 8–10 mm
[88]
PLA/PCL CO2 gas foaming Recellularized with HUVECs exhibiting high viability and migration;
Small ID
[89]
PEG/collagen/PU Electrospinning/hydrogelation Mean pressure = 50 mmHg, peak to through pressure = 20 mmHg, circumferential modulus = 190 kPa, SRS = 406 ± 124 gf, BP = 1440 ± 40 mmHg, C = 5.9 ± 1.4%, support rapid endothelialization;
ID = 3.7–4.7 mm
[90]
PLGA/collagen/elastin Stiffness: 2–137 kPa, 2–901 kPa, support ECs, SMCs growth, dry pore area = 1.92 ± 0.23 μm2 wet pore area = 4.74 ± 0.43 μm2;
dry D = 384 ± 22 nm–1196 ± 79 nm, wet D = 446 ± 69 nm–1735 ± 103
[91][92]
PA/PEG Hydrogelation/molding P = 35 nm, stiffness:0.1–0.3 kPa, 1–4 kPa, 6–8 kPa, cell adhesion observed [93]
PGS molding Supported the seeding of hSkMDCs and HUVECs [94]
PDMS/peptides microfluid Enhanced blood biocompatibility and cell adhesion [95]
PLLA/gelatin Electrospinning Supported SMCs and HUVECs alignment and proliferation and improved cell proliferation;
ID = 2–6 mm
[96]
PCL/collagen UTS = 4.0 MPa, EM = 2.5 MPa [97]
PCL/PEO/GCC hydrogel sleeve C = 4.5%, water permeability = 528 mL/cm2/min, BP = 695 mmHg, SRS = 2.38 N, supported the seeding and culturing of vascular ECs and SMCs in vitro, quick cell growth, and stable flow perfusion;
Small ID
[98]
Elastin/PDO SRS = 375 gf, C = 3.8%, EM = 9.64 MPa [99]
collagen/elastin/PLGA/PLCL Substantial interactions between SMCs;
D = 200–800 nm T = 0.5 mm
[91]
collagen/elastin/PLLA UTS = 0.83 MPa, EM = 2.08 MPa [72]
T = thickness; D = diameter; L = length; ID = inner diameter; P = porosity; BT = branched tubes; ST = straight tubes; BP = burst pressure; UTS = ultimate tensile stress; EM = elastic modulus; PE = permeability; SRS = suture retention strength; C = compliance; hSkMDCs = human skeletal muscle cells; hMSCs = human mesenchymal cells; LD = laser degradation; PI = phase inversion; HUVECs = human umbilical vein endothelial cells; HA-VSMCs = human aortic vascular smooth muscle cells, HDF-n = human dermal fibroblasts-neonatal; FB = fibroblast; HUSMCs = human uterine smooth muscle cells.
Table 2. Mechanical Properties of Native Blood Vessels.
Vessel Types and Axial Directions Elastic Modulus [100] Ultimate Tensile Strength [100] Strain at Failure (%) Burst Pressure (mmHg) References
Saphenous vein circumferential 43/4.2/2.25 3/1.8/4 11/243/180 NA/1680–3900/1250 [101][102]
Saphenous vein longitudinal 130/23.7 13/6.3 17/83 NA/NA [102][103]
Left internal mammary artery circumferential 8 4.1 134 2000 [102]
Left internal mammary artery longitudinal 16.8 4.3 59 NA [104]
Femoral artery circumferential 9–12 1–2 63–76 NA [105]
NA = not available.
More importantly, the future perspective for developing synthetic-polymer scaffolds should focus on enhancing biophysical performance, such as neovascularization. Some vascular scaffolds proved insufficient for cell regrowth due to the porosity and fabrication techniques used [106]. As an example, vascular scaffolds developed from electrospinning have been shown to possess low capacities for cell migration, adhesion, viability, and proliferation [107]. The relatively small pore sizes support these facts within electrospun scaffolds. Small pore sizes prevent cell infiltration and metabolite, nutrients, and waste diffusion. Synthetic (polyurethane and PLGA) and natural (derived from gelatin) polymers used to create electrospun scaffolds adversely influenced cell bioactivities due to their pore size and porosity [108]. Besides, cytotoxic solvents used in forming scaffolds’ surfaces, inferior structural integrities, and limited degradation rates have been shown to inhibit vascular remodeling and recellularization [109]. As a result, the successful creation of synthetic polymeric vascular tubes demands paying attention to the properties of polymers and other easily ignorable influencers, such as the cytotoxic solvents and agents residual in tissue-engineering technologies.

References

  1. Roth, G.A.; Mensah, G.A.; Johnson, C.O.; Addolorato, G.; Ammirati, E.; Baddour, L.M.; Barengo, N.C.; Beaton, A.Z.; Benjamin, E.J.; Benziger, C.P.; et al. Global Burden of Cardiovascular Diseases and Risk Factors, 1990–2019: Update From the GBD 2019 Study. J. Am. Coll. Cardiol. 2020, 76, 2982–3021.
  2. Ricklefs, M.; Korossis, S.; Haverich, A.; Schilling, T. Polymeric Scaffolds for Bioartificial Cardiovascular Prostheses. In Scaffolds in Tissue Engineering-Materials, Technologies and Clinical Applications; IntechOpen: London, UK, 2017; pp. 267–291.
  3. Wilhelmi, M.; Jockenhoevel, S.; Mela, P. Bioartificial fabrication of regenerating blood vessel substitutes: Requirements and current strategies. Biomed. Eng./Biomed. Tech. 2014, 59, 185–195.
  4. Constantinides, C.; Carr, C.; Schneider, J. Recent Advances in Image-Based Stem-Cell Labeling and Tracking, and Scaf-fold-Based Organ Development in Cardiovascular Disease. Recent Pat. Med. Imaging Discontin. 2014, 4, 110–126.
  5. Ozlu, B.; Ergin, M.; Budak, S.; Tunali, S.; Yildirim, N.; Erisken, C. A bioartificial rat heart tissue: Perfusion decellulari-zation and characterization. Int. J. Artif. Organs 2019, 42, 757–764.
  6. Diodato, M.; Chedrawy, E.G. Coronary Artery Bypass Graft Surgery: The Past, Present, and Future of Myocardial Revascularisation. Surg. Res. Pr. 2014, 2014, 726158.
  7. Barnes, V.A.; Orme-Johnson, D.W. Prevention and Treatment of Cardiovascular Disease in Adolescents and Adults through the Transcendental Meditation® Program: A Research Review Update. Curr. Hypertens. Rev. 2012, 8, 227–242.
  8. Wang, X.; Chan, V.; Corridon, P.R. Decellularized blood vessel development: Current state-of-the-art and future directions. Front. Bioeng. Biotechnol. 2022, 10, 1400.
  9. 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.
  10. Ravi, S.; Chaikof, E.L. Biomaterials for vascular tissue engineering. Regen. Med. 2010, 5, 107–120.
  11. Devillard, C.D.; Marquette, C.A. Vascular Tissue Engineering: Challenges and Requirements for an Ideal Large Scale Blood Vessel. Front. Bioeng. Biotechnol. 2021, 9, 913.
  12. Eltom, A.; Zhong, G.; Muhammad, A. Scaffold Techniques and Designs in Tissue Engineering Functions and Purposes: A Review. Adv. Mater. Sci. Eng. 2019, 2019, 3429527.
  13. Shakeel, A.C.; Corridon, P.R. Mitigating Challenges and Expanding the Future of Vascular Tissue Engineering—Are We There Yet? Available online: https://ssrn.com/abstract=4039050 (accessed on 20 February 2022).
  14. Corridon, P.R. In vitro investigation of the impact of pulsatile blood flow on the vascular architecture of decellularized porcine kidneys. Sci. Rep. 2021, 11, 16965.
  15. Corridon, P.R. Intravital microscopy datasets examining key nephron segments of transplanted decellularized kidneys. Sci. Data 2022, 9, 561.
  16. Corridon, P.R.; Ko, I.K.; Yoo, J.J.; Atala, A. Bioartificial Kidneys. Curr. Stem Cell Rep. 2017, 3, 68–76.
  17. Pantic, I.V.; Shakeel, A.; Petroianu, G.A.; Corridon, P.R. Analysis of Vascular Architecture and Parenchymal Damage Generated by Reduced Blood Perfusion in Decellularized Porcine Kidneys Using a Gray Level Co-occurrence Matrix. Front. Cardiovasc. Med. 2022, 9, 797283.
  18. Liu, J.; Zheng, H.; Poh, P.S.P.; Machens, H.-G.; Schilling, A.F. Hydrogels for Engineering of Perfusable Vascular Networks. Int. J. Mol. Sci. 2015, 16, 15997–16016.
  19. Wang, P.; Sun, Y.; Shi, X.; Shen, H.; Ning, H.; Liu, H. 3D printing of tissue engineering scaffolds: A focus on vascular regeneration. Bio-Design Manuf. 2021, 4, 344–378.
  20. Hasan, A.; Memic, A.; Annabi, N.; Hossain, M.; Paul, A.; Dokmeci, M.R.; Dehghani, F.; Khademhosseini, A. Elec-trospun scaffolds for tissue engineering of vascular grafts. Acta Biomater. 2014, 10, 11–25.
  21. Leal, B.B.; Wakabayashi, N.; Oyama, K.; Kamiya, H.; Braghirolli, D.I.; Pranke, P. Vascular Tissue Engineering: Poly-mers and Methodologies for Small Caliber Vascular Grafts. Front. Cardiovasc. Med. 2021, 7, 592361.
  22. Wang, Z.; Liu, L.; Mithieux, S.M.; Weiss, A.S. Fabricating Organized Elastin in Vascular Grafts. Trends Biotechnol. 2021, 39, 505–518.
  23. Zbinden, J.C.; Blum, K.M.; Berman, A.G.; Ramachandra, A.B.; Szafron, J.M.; Kerr, K.E.; Anderson, J.L.; Sangha, G.S.; Earl, C.C.; Nigh, N.R.; et al. Effects of Braiding Parameters on Tissue Engineered Vascular Graft Devel-opment. Adv. Healthc. Mater. 2020, 9, 2001093.
  24. Zbinden, J.C.; Blum, K.M.; Berman, A.G.; Ramachandra, A.B.; Szafron, J.M.; Kerr, K.E.; Anderson, J.L.; Sangha, G.S.; Earl, C.C.; Nigh, N.R.; et al. Tissue-Engineered Vascular Grafts: Effects of Braiding Parameters on Tissue Engineered Vascular Graft Development. Adv. Healthc. Mater. 2020, 9, 2070086.
  25. Bazgir, M.; Zhang, W.; Zhang, X.; Elies, J.; Saeinasab, M.; Coates, P.; Youseffi, M.; Sefat, F. Degradation and Characterisation of Electrospun Polycaprolactone (PCL) and Poly(lactic-co-glycolic acid) (PLGA) Scaffolds for Vascular Tissue Engineering. Materials 2021, 14, 4773.
  26. Lee, S.J.; Liu, J.; Oh, S.H.; Soker, S.; Atala, A.; Yoo, J.J. Development of a composite vascular scaffolding system that withstands physiological vascular conditions. Biomaterials 2008, 29, 2891–2898.
  27. Centola, M.; Rainer, A.; Spadaccio, C.; De Porcellinis, S.; Genovese, J.A.; Trombetta, M. Combining electrospin-ning and fused deposition modeling for the fabrication of a hybrid vascular graft. Biofabrication 2010, 2, 014102.
  28. Spadaccio, C.; Nappi, F.; De Marco, F.; Sedati, P.; Sutherland, F.W.; Chello, M.; Trombetta, M.; Rainer, A. Pre-liminary in Vivo Evaluation of a Hybrid Armored Vascular Graft Combining Electrospinning and Additive Manufacturing Techniques: Supplementary Issue: Current Developments in Drug Eluting Devices. Drug Target Insights 2016, 10, DTI-S35202.
  29. Baudis, S.; Nehl, F.; Ligon, S.C.; Nigisch, A.; Bergmeister, H.; Bernhard, D.; Stampfl, J.; Liska, R. Elastomeric de-gradable biomaterials by photopolymerization-based CAD-CAM for vascular tissue engineering. Biomed. Mater. 2011, 6, 055003.
  30. Paguirigan, A.; Beebe, D.J. Gelatin based microfluidic devices for cell culture. Lab Chip 2006, 6, 407–413.
  31. Lee, S.J.; Heo, D.N.; Park, J.S.; Kwon, S.K.; Lee, J.H.; Lee, J.H.; Kim, W.D.; Kwon, I.K.; Park, S.A. Characterization and preparation of bio-tubular scaffolds for fabricating artificial vascular grafts by combining electrospinning and a 3D printing system. Phys. Chem. Chem. Phys. 2014, 17, 2996–2999.
  32. Visser, J.; Peters, B.; Burger, T.J.; Boomstra, J.; Dhert, W.J.; Melchels, F.P.; Malda, J. Biofabrication of mul-ti-material anatomically shaped tissue constructs. Biofabrication 2013, 5, 035007.
  33. Browning, M.; Dempsey, D.; Guiza, V.; Becerra, S.; Rivera, J.; Russell, B.; Höök, M.; Clubb, F.; Miller, M.; Fossum, T.; et al. Multilayer vascular grafts based on collagen-mimetic proteins. Acta Biomater. 2012, 8, 1010–1021.
  34. L’Heureux, N.; Dusserre, N.; Konig, G.; Victor, B.; Keire, P.; Wight, T.N.; Chronos, N.A.; Kyles, A.E.; Gregory, C.R.; Hoyt, G.; et al. Human tissue-engineered blood vessels for adult arterial revascularization. Nat. Med. 2006, 12, 361–365.
  35. Rayatpisheh, S.; Heath, D.E.; Shakouri, A.; Rujitanaroj, P.-O.; Chew, S.Y.; Chan-Park, M.B. Combining cell sheet technology and electrospun scaffolding for engineered tubular, aligned, and contractile blood vessels. Biomaterials 2014, 35, 2713–2719.
  36. 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.
  37. Hashi, C.K.; Zhu, Y.; Yang, G.-Y.; Young, W.L.; Hsiao, B.S.; Wang, K.; Chu, B.; Li, S. Antithrombogenic property of bone marrow mesenchymal stem cells in nanofibrous vascular grafts. Proc. Natl. Acad. Sci. USA 2007, 104, 11915–11920.
  38. Patterson, J.T.; Gilliland, T.; Maxfield, M.W.; Church, S.; Naito, Y.; Shinoka, T.; Breuer, C.K. Tissue-engineered vascular grafts for use in the treatment of congenital heart disease: From the bench to the clinic and back again. Regen. Med. 2012, 7, 409–419.
  39. Ghasemi, A.; Imani, R.; Yousefzadeh, M.; Bonakdar, S.; Solouk, A.; Fakhrzadeh, H. Studying the Potential Applica-tion of Electrospun Polyethylene Terephthalate/Graphene Oxide Nanofibers as Electroconductive Cardiac Patch. Macromol. Mater. Eng. 2019, 304, 1900187.
  40. Hess, F. History of (MICRO) vascular surgery and the development of small-caliber blood vessel prostheses (with some notes on patency rates and re-endothelialization). Microsurgery 1985, 6, 59–69.
  41. Panitch, A. Implantation biology: The host response and biomedical devices, Ralph S. Greco, ed. (UMDNJ-Robert Wood Johnson Medical School), CRC Press, Inc., Boca Raton, Ann Arbor, London, Tokyo, 1994, 408 pp. J. Polym. Sci. Part A Polym. Chem. 1995, 33, 603-603.
  42. Jonas, R.A.; Ziemer, G.; Schoen, F.J.; Britton, L.; Castaneda, A.R. A new sealant for knitted Dacron prostheses: Minimally cross-linked gelatin. J. Vasc. Surg. 1988, 7, 414–419.
  43. Scott, S.M.; Gaddy, L.R.; Sahmel, R.; Hoffman, H. A collagen coated vascular prosthesis. J. Cardiovasc. Surg. 1987, 28, 498–504.
  44. Xue, L.; Greisler, H.P. Biomaterials in the development and future of vascular grafts. J. Vasc. Surg. 2003, 37, 472–480.
  45. Nunn, D.B.; Carter, M.M.; Donohue, M.T.; Hudgins, P.C. Postoperative dilation of knitted Dacron aortic bifurcation graft. J. Vasc. Surg. 1990, 12, 291–297.
  46. Alimi, Y.; Juhan, C.; Morati, N.; Girard, N.; Cohen, S. Dilation of Woven and Knitted Aortic Prosthetic Grafts: CT Scan Evaluation. Ann. Vasc. Surg. 1994, 8, 238–242.
  47. Kidson, I.G. The effect of wall mechanical properties on patency of arterial grafts. Ann. R. Coll. Surg. Engl. 1983, 65, 24–29.
  48. Gupta, B.S.; Kasyanov, V.A. Biomechanics of human common carotid artery and design of novel hybrid textile compliant vascular grafts. J. Biomed. Mater. Res. 1997, 34, 341–349.
  49. Friedman, S.G.; Lazzaro, R.S.; Spier, L.N.; Moccio, C.; Tortolani, A.J. A prospective randomized comparison of Dacron and polytetrafluoroethylene aortic bifurcation grafts. Surgery 1995, 117, 7–17.
  50. Prager, M.; Polterauer, P.; Böhmig, H.-J.; Wagner, O.; Fügl, A.; Kretschmer, G.; Plohner, M.; Nanobashvili, J.; Huk, I. Collagen versus gelatin-coated Dacron versus stretch polytetrafluoroethylene in abdominal aortic bifurca-tion graft surgery: Results of a seven-year prospective, randomized multicenter trial. Surgery 2001, 130, 408–414.
  51. Vogt, L.; Ruther, F.; Salehi, S.; Boccaccini, A.R. Poly(Glycerol Sebacate) in Biomedical Applications—A Review of the Recent Literature. Adv. Healthc. Mater. 2021, 10, e2002026.
  52. Kim, I.-Y.; Seo, S.-J.; Moon, H.-S.; Yoo, M.-K.; Park, I.-Y.; Kim, B.-C.; Cho, C.-S. Chitosan and its derivatives for tissue engineering applications. Biotechnol. Adv. 2008, 26, 1–21.
  53. Begovac, P.; Thomson, R.; Fisher, J.; Hughson, A.; Gällhagen, A. Improvements in GORE-TEX® vascular graft performance by Carmeda® bioactive surface heparin immobilization. Eur. J. Vasc. Endovasc. Surg. 2003, 25, 432–437.
  54. Devine, C.; Hons, B.; McCollum, C. Heparin-bonded Dacron or polytetrafluoroethylene for femoropopliteal bypass grafting: A multicenter trial. J. Vasc. Surg. 2001, 33, 533–539.
  55. Boretos, J.W.; Pierce, W.S. Segmented Polyurethane: A New Elastomer for Biomedical Applications. Science 1967, 158, 1481–1482.
  56. Yunus, S.; Sefa-Ntiri, B.; Anderson, B.; Kumi, F.; Mensah-Amoah, P.; Sackey, S.S. Quantitative Pore Characterization of Polyurethane Foam with Cost-Effective Imaging Tools and Image Analysis: A Proof-Of-Principle Study. Polymers 2019, 11, 1879.
  57. Williamson, M.R.; Black, R.; Kielty, C. PCL–PU composite vascular scaffold production for vascular tissue engineering: At-tachment, proliferation and bioactivity of human vascular endothelial cells. Biomaterials 2006, 27, 3608–3616.
  58. Mines, L.W.D.; Park, J.H.; Mudunkotuwa, I.A.; Anthony, T.R.; Grassian, V.H.; Peters, T.M. Porous Polyurethane Foam for Use as a Particle Collection Substrate in a Nanoparticle Respiratory Deposition Sampler. Aerosol Sci. Technol. 2016, 50, 497–506.
  59. Punnakitikashem, P.; Truong, D.; Menon, J.U.; Nguyen, K.T.; Hong, Y. Electrospun biodegradable elastic polyurethane scaffolds with dipyridamole release for small diameter vascular grafts. Acta Biomater. 2014, 10, 4618–4628.
  60. Greenwald, S.E.; Berry, C.L. Improving vascular grafts: The importance of mechanical and haemodynamic properties. J. Pathol. 2000, 190, 292–299.
  61. Place, E.S.; George, J.H.; Williams, C.K.; Stevens, M.M. Synthetic polymer scaffolds for tissue engineering. Chem. Soc. Rev. 2009, 38, 1139–1151.
  62. Bonferoni, M.C.; Caramella, C.; Catenacci, L.; Conti, B.; Dorati, R.; Ferrari, F.; Genta, I.; Modena, T.; Perteghella, S.; Rossi, S.; et al. Biomaterials for Soft Tissue Repair and Regeneration: A Focus on Italian Research in the Field. Pharmaceutics 2021, 13, 1341.
  63. Chrobak, K.M.; Potter, D.R.; Tien, J. Formation of perfused, functional microvascular tubes in vitro. Microvasc. Res. 2006, 71, 185–196.
  64. Rodrigues, S.F.; Granger, D.N. Blood cells and endothelial barrier function. Tissue Barriers 2015, 3, e978720.
  65. Kumar, P.; Shen, Q.; Pivetti, C.D.; Lee, E.S.; Wu, M.H.; Yuan, S.Y. Molecular mechanisms of endothelial hyperpermeability: Implications in inflammation. Expert Rev. Mol. Med. 2009, 11, e19.
  66. Owczarzy, A.; Kurasiński, R.; Kulig, K.; Rogóż, W.; Szkudlarek, A.; Maciążek-Jurczyk, M. Collagen—Structure, properties and application. Eng. Biomater. 2020, 23, 17–23.
  67. Cross, V.L.; Zheng, Y.; Choi, N.; Verbridge, S.S.; Sutermaster, B.A.; Bonassar, L.J.; Fischbach, C.; Stroock, A.D. Dense type I collagen matrices that support cellular remodeling and microfabrication for studies of tumor angiogenesis and vasculogenesis in vitro. Biomaterials 2010, 31, 8596–8607.
  68. Su, H.-J.; Shi, H.; Yu, J. A Symbolic Formulation for Analytical Compliance Analysis and Synthesis of Flexure Mechanisms. J. Mech. Des. 2012, 134, 051009.
  69. Xia, Y.; He, Y.; Zhang, F.; Liu, Y.; Leng, J. A Review of Shape Memory Polymers and Composites: Mechanisms, Materials, and Applications. Adv. Mater. 2020, 33, 2000713.
  70. Oh, S.H.; An, D.B.; Kim, T.H.; Lee, J.H. Wide-range stiffness gradient PVA/HA hydrogel to investigate stem cell differentiation behavior. Acta Biomater. 2016, 35, 23–31.
  71. Joh, J.H.; Park, H.-C. The cutoff value of saphenous vein diameter to predict reflux. J. Korean Surg. Soc. 2013, 85, 169–174.
  72. Lee, S.J.; Yoo, J.J.; Lim, G.J.; Atala, A.; Stitzel, J. In vitro evaluation of electrospun nanofiber scaffolds for vascular graft application. J. Biomed. Mater. Res. Part A 2007, 83A, 999–1008.
  73. Brandenberg, N.; Lutolf, M.P. In Situ Patterning of Microfluidic Networks in 3D Cell-Laden Hydrogels. Adv. Mater. 2016, 28, 7450–7456.
  74. Heintz, K.A.; Bregenzer, M.E.; Mantle, J.L.; Lee, K.H.; West, J.L.; Slater, J.H. Fabrication of 3D Biomimetic Microfluidic Networks in Hydrogels. Adv. Healthc. Mater. 2016, 5, 2153–2160.
  75. Nakazawa, Y.; Sato, M.; Takahashi, R.; Aytemiz, D.; Takabayashi, C.; Tamura, T.; Enomoto, S.; Sata, M.; Asakura, T. Development of Small-Diameter Vascular Grafts Based on Silk Fibroin Fibers from Bombyx mori for Vascular Regeneration. J. Biomater. Sci. Polym. Ed. 2011, 22, 195–206.
  76. Ling, Y.; Rubin, J.; Deng, Y.; Huang, C.; Demirci, U.; Karp, J.M.; Khademhosseini, A. A cell-laden microfluidic hydrogel. Lab Chip 2007, 7, 756–762.
  77. Hinton, T.J.; Jallerat, Q.; Palchesko, R.N.; Park, J.H.; Grodzicki, M.S.; Shue, H.-J.; Ramadan, M.H.; Hudson, A.R.; Feinberg, A.W. Three-dimensional printing of complex biological structures by freeform reversible embedding of sus-pended hydrogels. Sci. Adv. 2015, 1, e1500758.
  78. Xu, Y.; Wang, X. Fluid and cell behaviors along a 3D printed alginate/gelatin/fibrin channel. Biotechnol. Bioeng. 2015, 112, 1683–1695.
  79. Pinnock, C.B.; Meier, E.M.; Joshi, N.N.; Wu, B.; Lam, M.T. Customizable engineered blood vessels using 3D printed inserts. Methods 2016, 99, 20–27.
  80. Xu, Y.; Hu, Y.; Liu, C.; Yao, H.; Liu, B.; Mi, S. A Novel Strategy for Creating Tissue-Engineered Biomimetic Blood Vessels Using 3D Bioprinting Technology. Materials 2018, 11, 1581.
  81. Melchiorri, A.J.; Hibino, N.; Best, C.A.; Yi, T.; Lee, Y.U.; Kraynak, C.A.; Kimerer, L.K.; Krieger, A.; Kim, P.; Breuer, C.K.; et al. 3D-Printed Biodegradable Polymeric Vascular Grafts. Adv. Healthc. Mater. 2015, 5, 319–325.
  82. Mishra, R.; Roux, B.M.; Posukonis, M.; Bodamer, E.; Brey, E.M.; Fisher, J.P.; Dean, D. Effect of prevascularization on in vivo vascularization of poly(propylene fumarate)/fibrin scaffolds. Biomaterials 2016, 77, 255–266.
  83. Meyer, W.; Engelhardt, S.; Novosel, E.; Elling, B.; Wegener, M.; Krüger, H. Soft Polymers for Building up Small and Smallest Blood Supplying Systems by Stereolithography. J. Funct. Biomater. 2012, 3, 257–268.
  84. Catto, V.; Farè, S.; Freddi, G.; Tanzi, M.C. Vascular Tissue Engineering: Recent Advances in Small Diameter Blood Vessel Regeneration. ISRN Vasc. Med. 2014, 2014, 923030.
  85. Roh, J.D.; Nelson, G.N.; Brennan, M.P.; Mirensky, T.L.; Yi, T.; Hazlett, T.F.; Tellides, G.; Sinusas, A.J.; Pober, J.S.; Saltzman, W.; et al. Small-diameter biodegradable scaffolds for functional vascular tissue engineering in the mouse model. Biomaterials 2008, 29, 1454–1463.
  86. Zeinali, S.; Thompson, E.K.; Gerhardt, H.; Geiser, T.; Guenat, O.T. Remodeling of an in vitro microvessel exposed to cyclic mechanical stretch. APL Bioeng. 2021, 5, 026102.
  87. Chen, Y.; Ding, X.; Li, Y.; King, M.W.; Gao, J.; Zhao, X. A bilayer prototype woven vascular prosthesis with improved radial compliance. J. Text. Inst. 2012, 103, 106–111.
  88. Singh, C.; Wong, C.S.; Wang, X. Medical Textiles as Vascular Implants and Their Success to Mimic Natural Arteries. J. Funct. Biomater. 2015, 6, 500–525.
  89. Murphy, S.; Marsh, J.; Kelly, C.; Leeke, G.; Jenkins, M. CO2 assisted blending of poly(lactic acid) and poly(ε-caprolactone). Eur. Polym. J. 2016, 88, 34–43.
  90. Browning, L.M.; Walker, C.G.; Mander, A.P.; West, A.L.; Madden, J.; Gambell, J.M.; Young, S.; Wang, L.; Jebb, S.A.; Calder, P.C. Incorporation of eicosapentaenoic and docosahexaenoic acids into lipid pools when given as supple-ments providing doses equivalent to typical intakes of oily fish. Am. J. Clin. Nutr. 2012, 96, 748–758.
  91. Foraida, Z.I.; Kamaldinov, T.; Nelson, D.A.; Larsen, M.; Castracane, J. Elastin-PLGA hybrid electrospun nanofiber scaffolds for salivary epithelial cell self-organization and polarization. Acta Biomater. 2017, 62, 116–127.
  92. Jeong, S.I.; Kim, S.Y.; Cho, S.K.; Chong, M.S.; Kim, K.S.; Kim, H.; Lee, S.B.; Lee, Y.M. Tissue-engineered vascular grafts composed of marine collagen and PLGA fibers using pulsatile perfusion bioreactors. Biomaterials 2007, 28, 1115–1122.
  93. Jin, M.M.; Shi, M.J.; Zhu, W.; Yao, H.; Wang, D.-A. Polysaccharide-Based Biomaterials in Tissue Engineering: A Review. Tissue Eng. Part B Rev. 2021, 27, 604–626.
  94. Ye, X.; Lu, L.; Kolewe, M.E.; Park, H.; Larson, B.L.; Kim, E.S.; Freed, L.E. A biodegradable microvessel scaffold as a framework to enable vascular support of engineered tissues. Biomaterials 2013, 34, 10007–10015.
  95. Mikhail, A.S. Peptide Modified PDMS: Surface Modification For Improved Vascular Cell Interactions. Master’s Thesis, McMaster University, Hamilton, ON, Canada, 2006.
  96. Shalumon, K.T.; Deepthi, S.; Anupama, M.S.; Nair, S.V.; Jayakumar, R.; Chennazhi, K.P. Fabrication of poly (L-lactic acid)/gelatin composite tubular scaffolds for vascular tissue engineering. Int. J. Biol. Macromol. 2015, 72, 1048–1055.
  97. Fu, W.; Liu, Z.; Feng, B.; Hu, R.; He, X.; Wang, H.; Yin, M.; Huang, H.; Zhang, H.; Wang, W. Electrospun gelatin/PCL and collagen/PLCL scaffolds for vascular tissue engineering. Int. J. Nanomed. 2014, 9, 2335–2344.
  98. Madhavan, K.; Elliott, W.H.; Bonani, W.; Monnet, E.; Tan, W. Mechanical and biocompatible characterizations of a readily available multilayer vascular graft. J. Biomed. Mater. Res. Part B Appl. Biomater. 2012, 101B, 506–519.
  99. Sell, S.; McClure, M.J.; Barnes, C.P.; Knapp, D.C.; Walpoth, B.H.; Simpson, D.G.; Bowlin, G.L. Electrospun polydioxanone–elastin blends: Potential for bioresorbable vascular grafts. Biomed. Mater. 2006, 1, 72–80.
  100. Mastrullo, V.; Cathery, W.; Velliou, E.; Madeddu, P.; Campagnolo, P. Angiogenesis in Tissue Engineering: As Nature Intended? Front. Bioeng. Biotechnol. 2020, 8, 188.
  101. Donovan, D.L.; Schmidt, S.P.; Townshend, S.P.; Njus, G.O.; Sharp, W.V. Material and structural characterization of human saphenous vein. J. Vasc. Surg. 1990, 12, 531–537.
  102. Stekelenburg, M.; Rutten, M.C.; Snoeckx, L.H.; Baaijens, F.P. Dynamic Straining Combined with Fibrin Gel Cell Seeding Improves Strength of Tissue-Engineered Small-Diameter Vascular Grafts. Tissue Eng. Part A 2009, 15, 1081–1089.
  103. Soletti, L.; Hong, Y.; Guan, J.; Stankus, J.J.; El-Kurdi, M.S.; Wagner, W.; Vorp, D.A. A bilayered elastomeric scaffold for tissue engineering of small diameter vascular grafts. Acta Biomater. 2010, 6, 110–122.
  104. Yamada, H.; Evans, F. Mechanical properties of circulatory organs and tissues. In Strength of Biological Materials; Krieger: New York, NY, USA, 1970; pp. 106–137.
  105. Fung, Y.C. Blood Flow in Arteries. In Biomechanics: Circulation; Fung, Y.C., Ed.; Springer: New York, NY, USA, 1997; pp. 108–205.
  106. Rouwkema, J.; Rivron, N.C.; van Blitterswijk, C.A. Vascularization in tissue engineering. Trends Biotechnol. 2008, 26, 434–441.
  107. Tan, Z.; Wang, H.; Gao, X.; Liu, T.; Tan, Y. Composite vascular grafts with high cell infiltration by co-electrospinning. Mater. Sci. Eng. C 2016, 67, 369–377.
  108. Dahlin, R.L.; Kasper, F.K.; Mikos, A.G. Polymeric Nanofibers in Tissue Engineering. Tissue Eng. Part B Rev. 2011, 17, 349–364.
  109. Nikolova, M.P.; Chavali, M.S. Recent advances in biomaterials for 3D scaffolds: A review. Bioact. Mater. 2019, 4, 271–292.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 402
Revisions: 2 times (View History)
Update Date: 21 Nov 2022
1000/1000