Harrick PlasmaApplicationsBiology & BiomedicalCell Adhesion

Cell adhesion plays an integral role in cell culture and tissue engineering. In the native environment, cell adhesion molecules (CAMs) bind to the extracellular matrix and neighboring cells to provide structural support and chemical cues vital for cell viability, proliferation, and differentiation. However, most cell culture materials are inert, impeding cell anchorage. Plasma treatment introduces bioactive, hydrophilic functional groups to cell culture materials, improving cell adhesion and cell viability. Below you will find information regarding cell adhesion for different cell culture materials, and how plasma treatment is used to enhance biocompatibility.

Cell culture materials impact a target cell’s ability to proliferate and function as intended. These materials provide highly specific chemical and mechanical cues that determine a cells morphology and differentiation. Most commonly, cells are cultured on plasma treated polystyrene (Tissue Culture Plastic). While TCP enables rapid growth and development, flat cell morphology can negatively impact cell function or even force cells down unintended differentiation pathways (Ex: Neuron Morphology vs Glial Cells). More recently, 3D cell culture materials have been used to reproduce the native environment in artificial constructs.  Polymer cell scaffolds are often used due to their similarity with the extracellular matrix, low cost and inert and non-toxic chemistry. Many polymeric scaffolds are biodegradable or have other interesting features that contribute to their success in these applications. However, all of these materials are hydrophobic and harmful for cell adhesion.

Plasma treatment is an essential tool for the development of bioactive cell culture materials with high cell adhesion and hydrophilicity. An air or oxygen plasma is typically used for nanoscale cleaning and the introduction of functional groups with high biological affinity (Carboxyl, Hydroxyl, Amine). Without hazardous or long wet chemistry processes, benchtop plasma cleaners create hydrophilic surfaces suitable for cell seeding or coating right in the lab. As a result, researchers have been able to more quickly and easily manipulate the chemistry of their cell scaffolds. This includes the introduction of extracellular matrix constituents, such as fibronectin, that can further enhance cell function.

POLYCAPROLACTONE (PCL)

Polycaprolactone (PCL) is often used as a cell scaffold due to its similarity with native ECM and its long and non-toxic biodegradation rate. PCL has an excellent clinical track record and is approved by the FDA in several existing medical devices. Plasma treatment is often used to increase cell attachment directly, or to prepare PCL substrates for surface coatings that will improve cell activity. Currently, PCL scaffolds research is mainly focused on bone and cartilage formation.

Cells & Tissue: Endothelial, Epithelial, Bone, Adipose, Kidney, Neurons, Skin, Liver, Cartilage, ACL, Heart Valve Leaflets, Prostate, Smooth Muscle, Tumor Models

Process Gases: Air, Oxygen, Argon, Nitrogen, Carbon Dioxide

Polycaprolactone (PCL) Articles

Abbasi N, Hamlet S, and Nguyen N. “Calcium phosphate stability on melt electrowritten PCL scaffolds”. Journal of Science: Advanced Materials and Devices 2020 5: 30-39 10.1016/j.jsamd.2020.01.001

Bate T. S. R, Forbes S. J, and Callanan A. “Controlling Electrospun Polymer Morphology for Tissue Engineering Demonstrated Using hepG2 Cell Line”. Journal of Visualized Experiments 2020 10.3791/61043

Beardslee LA, Stolwijk J, Khaladj DA, Trebak M, Halman J, Torrejon KY, Niamsiri N, and Bergkvist M. “A sacrificial process for fabrication of biodegradable polymer membranes with submicron thickness”. J. Biomed. Mater. Res. B 2015 104: 1192–1201 10.1002/jbm.b.33464

Bock N. “Bioengineered Microtissue Models of the Human Bone Metastatic Microenvironment: A Novel In Vitro Theranostics Platform for Cancer Research”. Theranostics 2019 2054: 23-57 10.1007/978-1-4939-9769-5_2

Burton TP, Corcoran A, and Callanan A. “The effect of electrospun polycaprolactone scaffold morphology on human kidney epithelial cells”. Biomed. Mater. 2018 13: 15006 10.1088/1748-605X/aa8dde

Calhoun M, Chowdhury S, Nelson M, Lannutti J, Dupaix R, and Winter J. “Effect of Electrospun Fiber Mat Thickness and Support Method on Cell Morphology”. Nanomaterials 2019 9  https://doi.org/10.3390/nano9040644

Cohn C, Leung SL, Crosby J, Lafuente B, Zha Z, Teng W, Downs R, and Wu X. “Lipid-mediated protein functionalization of electrospun polycaprolactone fibers.”. eXPRESS Polym. Lett. 2016 10: 430–437 10.3144/expresspolymlett.2016.40

Costa DO, Prowse PD, Chrones T, Sims SM, Hamilton DW, Rizkalla AS, and Dixon SJ. “The differential regulation of osteoblast and osteoclast activity by surface topography of hydroxyapatite coatings”. Biomaterials 2013 34: 7215–7226 10.1016/j.biomaterials.2013.06.014

Damanik FFR, Rothuizen TC, van Blitterswijk C, Rotmans JI, and Moroni L. “Towards an in vitro model mimicking the foreign body response: tailoring the surface properties of biomaterials to modulate extracellular matrix”. Sci. Rep. 2014 4: 6325 10.1038/srep06325

Donoghue PS, Sun T, Gadegaard N, Riehle MO, and Barnett SC. “Development of a Novel 3D Culture System for Screening Features of a Complex Implantable Device for CNS Repair”. Mol. Pharmaceutics 2014 11: 2143–2150 10.1021/mp400526n

Fu L, Xie J, Carlson MA, and Reilly DA. “Three-dimensional nanofiber scaffolds with arrayed holes for engineering skin tissue constructs”. MRS Commun. 2017 7: 361-366 10.1557/mrc.2017.49

Grant R, Hay DC, and Callanan A. “A Drug-Induced Hybrid Electrospun Poly-Capro-Lactone: Cell-Derived Extracellular Matrix Scaffold for Liver Tissue Engineering”. Tissue Eng. Part A 2017 23: 650-662 10.1089/ten.tea.2016.0419

Joshi VS, Lei NY, Walthers CM, Wu B, and Dunn JC. “Macroporosity enhances vascularization of electrospun scaffolds”. J. Surg. Res. 2013 183: 18–26 10.1016/j.jss.2013.01.005

Leong NL, Kabir N, Arshi A, Nazemi A, Jiang J, Wu BM, Petrigliano FA, and McAllister DR. “Use of ultra-high molecular weight polycaprolactone scaffolds for ACL reconstruction”. J. Orthop. Res. 2016 34: 828–835 10.1002/jor.23082

Leong NL, Arshi A, Kabir N, Nazemi A, Petrigliano FA, Wu BM, and McAllister DR. “In vitro and in vivo evaluation of heparin mediated growth factor release from tissue-engineered constructs for anterior cruciate ligament reconstruction”. J. Orthop. Res. 2015 33: 229–236 10.1002/jor.22757

Leong NL, Kabir N, Arshi A, Nazemi A, Wu BM, McAllister DR, and Petrigliano FA. “Athymic Rat Model for Evaluation of Engineered Anterior Cruciate Ligament Grafts”. J. Vis. Exp. 2015 97: e52797 10.3791/52797

Leong NL, Kabir N, Arshi A, Nazemi A, Wu B, Petrigliano FA, and McAllister DR. “Evaluation of polycaprolactone scaffold with basic fibroblast growth factor and fibroblasts in an athymic rat model for anterior cruciate ligament reconstruction”. Tissue Eng. 2015 21: 1859–1868 10.1089/ten.tea.2014.0366

Ma Z, He W, Yong T, and Ramakrishna S. “Grafting of gelatin on electrospun poly(caprolactone) nanofibers to improve endothelial cell spreading and proliferation and to control cell orientation”. Tissue Eng. 2005 11: 1149–1158 10.1089/ten.2005.11.1149

Macdonald M, Samuel R, Shah N, Padera R, Beben Y, and Hammond P. “Tissue integration of growth factor-eluting layer-by-layer polyelectrolyte multilayer coated implants”. Biomaterials 2011 32: 1446 — 1453 10.1016/j.biomaterials.2010.10.052

Masoumi N, Annabi N, Assmann A, Larson BL, Hjortnaes J, Alemdar N, Kharaziha M, Manning KB, Mayer JE, and Khademhosseini A. “Tri-layered elastomeric scaffolds for engineering heart valve leaflets”. Biomaterials 2014 35: 7774–7785 10.1016/j.biomaterials.2014.04.039

Munj HR, and Tomasko DL. “Polycaprolactone-polymethyl methacrylate electrospun blends for biomedical applications”. Polym. Sci. Ser. A 2017 59: 695-707 10.1134/S0965545X17050121

Munir N, McDonald A, and Callanan A. “A combinatorial approach: Cryo-printing and electrospinning hybrid scaffolds for cartilage tissue engineering”. Bioprinting 2019 16 10.1016/j.bprint.2019.e00056

Prabhakaran MP, Venugopal J, Chan CK, and Ramakrishna S. “Surface modified electrospun nanofibrous scaffolds for nerve tissue engineering”. Nanotechnology 2008 19: 455102 10.1088/0957-4484/19/45/455102

Pereira B, Lister N, Hashimoto K, Teng L, Flandes-Iparraquirre M, Eder A, Sanchez-Herrero A, and Niranjan B. “Tissue engineered human prostate microtissues reveal key role of mast cell-derived tryptase in potentiating cancer-associated fibroblast (CAF)-induced morphometric transition in vitro”. Biomaterials 2019 197: 72-85 10.1016/j.biomaterials.2018.12.030

Rath B, Nam J, Deschner J, Schaumburger J, Tingart M, Grassel S, Grifka J, and Agarwal S. “Biomechanical forces exert anabolic effects on osteoblasts by activation of SMAD 1/5/8 through type 1 BMP receptor”. Biorheology 2011 48: 37 — 48 10.3233/BIR-2011-0580

Sarkar S, Lee GY, Wong JY, and Desai TA. “Development and characterization of a porous micro-patterned scaffold for vascular tissue engineering applications”. Biomaterials 2006 27: 4775–4782 10.1016/j.biomaterials.2006.04.038

Sridhar R, Madhaiyan K, Sundarrajan S, Gora A, Venugopal JR, and Ramakrishna S. “Cross-linking of protein scaffolds for therapeutic applications: PCL nanofibers delivering riboflavin for protein cross-linking”. J. Mater. Chem. B 2014 2: 1626–1633 10.1039/c3tb21789b

Sun T, Donoghue PS, Higginson JR, Gadegaard N, Barnett SC, and Riehle MO. “A miniaturized bioreactor system for the evaluation of cell interaction with designed substrates in perfusion culture”. J. Tissue Eng. Regener. Med. 2012 6: s4–s14 10.1002/term.510

Sun T, Donoghue P, Higginson J, Gadegaard N, Barnett S, and Riehle M. “The interactions of astrocytes and fibroblasts with defined pore structures in static and perfusion cultures”. Biomaterials 2011 32: 2021 — 2031 10.1016/j.biomaterials.2010.11.046

Valence Sd, Tille J-C, Chaabane C, Gurny R, Bochaton-Piallat M-L, Walpoth BH, and Muller M. “Plasma treatment for improving cell biocompatibility of a biodegradable polymer scaffold for vascular graft applications”. Eur. J. Pharm. Biopharm. 2013 85: 78–86 10.1016/j.ejpb.2013.06.012

Walthers CM, Nazemi AK, Patel SL, Wu BM, and Dunn JCY. “The effect of scaffold macroporosity on angiogenesis and cell survival in tissue-engineered smooth muscle”. Biomaterials 2014 35: 5129–5137 10.1016/j.biomaterials.2014.03.025

Walthers CM, Lyall CJ, Nazemi AK, Rana PV, and Dunn JCY. “Collagen and heparan sulfate coatings differentially alter cell proliferation and attachment in vitro and in vivo”. Technology 2016 4: 1640003 10.1142/s2339547816400033

Yang F, Wolke JGC, and Jansen JA. “Biomimetic calcium phosphate coating on electrospun poly (epsilon-caprolactone) scaffolds for bone tissue engineering”. Chem. Eng. J. 2008 137: 154–161 10.1016/j.cej.2007.07.076

Yildirim ED, Pappas D, Guceri S, and Sun W. “Enhanced Cellular Functions on Polycaprolactone Tissue Scaffolds by O2 Plasma Surface Modification”. Plasma Processes Polym. 2011 8: 256–267 10.1002/ppap.201000009

Zander N, Orlicki J, Rawlett A, and Beebe T. “Surface-modified nanofibrous biomaterial bridge for the enhancement and control of neurite outgrowth”. Biointerphases 2010 5: 149–158 10.1116/1.3526140

Zhang W, Lee WY, and Zilberberg J. “Tissue Engineering Platforms to Replicate the Tumor Microenvironment of Multiple Myeloma”. Methods Mol. Biol. 2017 1513: 171-191 10.1007/978-1-4939-6539-7_12

Zhao X, Lui YS, Choo CKC, Sow WT, Huang CL, Ng KW, Tan LP, and Loo JSC. “Calcium phosphate coated keratin-PCL scaffolds for potential bone tissue regeneration”. Mater. Sci. Eng. C 2015 49: 746–753 10.1016/j.msec.2015.01.084

Zhao X, Irvine SA, Agrawal A, Cao Y, Lim PQ, Tan SY, and Venkatraman SS. “3D patterned substrates for bioartificial blood vessels-The effect of hydrogels on aligned cells on a biomaterial surface”. Acta Biomater. 2015 26: 159–168 10.1016/j.actbio.2015.08.024

POLYLACTIC ACID (PLA, PLLA)

Polylactic acid (PLA) scaffolds are both biocompatible and biodegradable. As a result, PLA is highly suitable for short term surgical products such as sutures and mesh implants. Literature profiling the mechanical properties of PLA is extensive, enabling fine control of polymer porosity and degradation. Plasma treatment of PLA cleans the surface and increases cell spreading, proliferation and differentiation. Currently, PLA is featured in osteogenesis, neurogenesis and vascularization for cell grafting.

Cells & Tissue: Bone, Endothelial, Adipose, Neurons, Skin, Blood vessels

Process Gases: Air, Oxygen, Argon, Nitrogen, Neon

POLYLACTIC ACID (PLA, PLLA) Articles

Chim H, Ong JL, Schantz JT, Hutmacher DW, and Agrawal CM. “Efficacy of glow discharge gas plasma treatment as a surface modification process for three-dimensional poly (D,L-lactide) scaffolds”. J. Biomed. Mater. Res. A 2003 65A: 327–335 10.1002/jbm.a.10478

Chou Y-F, Zuk PA, Chang T-L, Benhaim P, and Wu BM. “Adipose-derived stem cells and BMP2: Part 1. BMP2-treated adipose-derived stem cells do not improve repair of segmental femoral defects”. Connect. Tissue Res. 2011 52: 109–118 10.3109/03008207.2010.484514

Demina TS, Gilman AB, and Zelenetskii AN. “Application of high-energy chemistry methods to the modification of the structure and properties of polylactide (a review)”. High Energy Chem. 2017 51: 302-314 10.1134/S0018143917040038

Koh H, Yong T, Chan C, and Ramakrishna S. “Enhancement of neurite outgrowth using nano-structured scaffolds coupled with laminin”. Biomaterials 2008 29: 3574—3582 10.1016/j.biomaterials.2008.05.014

Liu W, Cai Q, Zhang F, Wei Y, Zhang X, Wang Y, Deng X, and Deng X. “Dose-dependent enhancement of bone marrow stromal cells adhesion, spreading and osteogenic differentiation on atmospheric plasma-treated poly(L-lactic acid) nanofibers”. J. Bioact. Compat. Polym. 2013 28: 453–467 10.1177/0883911513494623

Lu HJ, Feng ZQ, Gu ZZ, and Liu CJ. “Growth of outgrowth endothelial cells on aligned PLLA nanofibrous scaffolds”. J. Mater. Sci. 2009 20: 1937–1944 10.1007/s10856-009-3744-y

Mohiti-Asli M, Saha S, Murphy SV, Gracz H, Pourdeyhimi B, Atala A, and Loboa EG. “Ibuprofen loaded PLA nanofibrous scaffolds increase proliferation of human skin cells in vitro and promote healing of full thickness incision wounds in vivo”. J. Biomed. Mater. Res. B 2015 105: 327–339 10.1002/jbm.b.33520

Shah A, Shah S, Oh S, Ong J, Wenke J, and Agrawal C. “Migration of Co-cultured Endothelial Cells and Osteoblasts in Composite Hydroxyapatite/Polylactic Acid Scaffolds”. Ann. Biomed. Eng. 2011 39: 2501–2509 10.1007/s10439-011-0344-z

Zuidema JM, Hyzinski-Garcia MC, Van Vlasselaer K, Zaccor NW, Plopper GE, Mongin AA, and Gilbert RJ. “Enhanced GLT-1 mediated glutamate uptake and migration of primary astrocytes directed by fibronectin-coated electrospun poly-l-lactic acid fibers”. Biomaterials 2014 35: 1439–1449 10.1016/j.biomaterials.2013.10.079

Supplemental References (Do Not Report Using Harrick Plasma Cleaners)

Lasprilla, A. J. R., Martinez, G. A. R., Lunelli, B. H., Jardini, A. L., & Filho, R. M. (2012). Poly-lactic acid synthesis for application in biomedical devices—A review. Biotechnology Advances, 30(1), 321–328. https://doi.org/10.1016/j.biotechadv.2011.06.019

POLY(LACTIC-CO-GLYCOLIC ACID) (PLGA)

Poly(Lactic-co-Glycolic Acid) (PLGA) is biocompatible, with tunable degradation and topography based on the ratio of lactic acid to glycolic acid available during polymerization. PLGA scaffolds are often coated with minerals such as hyaluronic acid or fibrin to improve fiber strength, or cell activity. Plasma treatment is highly effective at cleaning PLGA, without changing the morphology of the scaffold. Plasma can also be used to improve surface wetting and coating uniformity. With an excellent track record of safety and approved by the FDA, PLGA is often used for studies on bone formation and differentiation.

Cells & Tissue: Bone, Tendon-Bone, Adipose, Salivary glands

Process Gases: Air, Oxygen, Argon

POLY(LACTIC-CO-GLYCOLIC ACID) (PLGA) Articles

Cowan CM, Shi YY, Aalami OO, Chou YF, Mari C, Thomas R, Quarto N, Contag CH, Wu B, and Longaker MT. “Adipose-derived adult stromal cells heal critical-size mouse calvarial defects”. Nat. Biotechnol. 2004 22: 560–567 10.1038/nbt958

Foraida ZI, Kamaldinov T, Nelson DA, Larsen M, and Castracane J. “Elastin-PLGA hybrid electrospun nanofiber scaffolds for salivary epithelial cell self-organization and polarization”. Acta Biomater. 2017 62: 116-127 10.1016/j.actbio.2017.08.009

Holy CE, Cheng C, Davies JE, and Shoichet MS. “Optimizing the sterilization of PLGA scaffolds for use in tissue engineering”. Biomaterials 2001 22: 25–31 10.1016/S0142-9612(00)00136-8

James AW, Zara JN, Corselli M, Chiang M, Yuan W, Nguyen V, Askarinam A, Goyal R, Siu RK, Scott V, Lee M, Ting K, Peault B, and Soo C. “Use of Human Perivascular Stem Cells for Bone Regeneration”. J. Vis. Exp. 2012 63: e2952 10.3791/2952

Kolluru PV, Lipner J, Liu W, Xia Y, Thomopoulos S, Genin GM, and Chasiotis I. “Strong and tough mineralized PLGA nanofibers for tendon-to-bone scaffolds”. Acta Biomater. 2013 9: 9442–9450 10.1016/j.actbio.2013.07.042

Lipner J, Boyle JJ, Xia Y, Birman V, Genin GM, and Thomopoulos S. “Toughening of fibrous scaffolds by mobile mineral deposits”. Acta Biomater. 2017 58: 492-501 10.1016/j.actbio.2017.05.033

Lipner J, Shen H, Cavinatto L, Liu W, Havlioglu N, Xia Y, Galatz LM, and Thomopoulos S. “In vivo evaluation of adipose-derived stromal cells delivered with a nanofiber scaffold for tendon-to-bone repair”. Tissue Eng. Part A 2015 21: 2766–2774 10.1089/ten.tea.2015.0101

Lo DD, Hyun JS, Chung MT, Montoro DT, Zimmermann A, Grova MM, Lee M, Wan DC, and Longaker MT. “Repair of a Critical-sized Calvarial Defect Model Using Adipose-derived Stromal Cells Harvested from Lipoaspirate”. J. Vis. Exp. 2012 68: e4221 10.3791/4221

Son J, Appleford M, Ong J, Wenke J, Kim J, Choi S, and Oh D. “Porous hydroxyapatite scaffold with three-dimensional localized drug delivery system using biodegradable microspheres”. J. Controlled Release 2011 153: 133 — 140 10.1016/j.jconrel.2011.03.010

Supplemental References (Do Not Report Using Harrick Plasma Equipment)

Gentile, P., Chiono, V., Carmagnola, I., & Hatton, P. V. (2014). An Overview of Poly(lactic-co-glycolic) Acid (PLGA)-Based Biomaterials for Bone Tissue Engineering. International Journal of Molecular Sciences, 15(3), 3640–3659. https://doi.org/10.3390/ijms15033640

Poly(l-lactic acid)-co-poly (ε-caprolactone) (PLACL)

Poly(L-Lactic Acid)-Co-Poly(E-Caprolactone) is a tunable copolymer, due to the different properties provided by Poly(L-Lactic Acid) (PLA) and Poly(E-Caprolactone) (PCL). This tunability makes it a highly suitable candidate for drug or cell delivery. Plasma treatment increases cell adhesion and density while also providing the opportunity for PLACL substrates to be coated with materials such as collagen or hyaluronic acid. These scaffolds are then leveraged for usage in bone, vascular and muscular formation.

Cells & Tissue: Bone, Mesenschymal Stem Cells, Skin, Endothelial Cells and Vascular grafts

Process Gases: Air

POLY(L-LACTIC ACID)-CO-POLY (Ε-CAPROLACTONE) (PLACL) Articles

Chan C, Liao S, Li B, Lareu R, Larrick J, Ramakrishna S, and Raghunath M. “Early adhesive behavior of bone-marrow-derived mesenchymal stem cells on collagen electrospun fibers”. Biomed. Mater. 2009 4: 35006 10.1088/1748-6041/4/3/035006

Chandrasekaran A, Venugopal J, Sundarrajan S, and Ramakrishna S. “Fabrication of a nanofibrous scaffold with improved bioactivity for culture of human dermal fibroblasts for skin regeneration”. Biomed. Mater. 2011 6: 15001 10.1088/1748-6041/6/1/015001

Gupta D, Venugopal J, Mitra S, Dev VRG, and Ramakrishna S. “Nanostructured biocomposite substrates by electrospinning and electrospraying for the mineralization of osteoblasts”. Biomaterials 2009 30: 2085–2094 10.1016/j.biomaterials.2008.12.079

He W, Yong T, Ma ZW, Inai R, Teo WE, and Ramakrishna S. “Biodegradable polymer nanofiber mesh to maintain functions of endothelial cells”. Tissue Eng. 2006 12: 2457–2466 10.1089/ten.2006.12.ft-195

He W, Ma ZW, Yong T, Teo WE, and Ramakrishna S. “Fabrication of collagen-coated biodegradable polymer nanofiber mesh and its potential for endothelial cells growth”. Biomaterials 2005 26: 7606–7615 10.1016/j.biomaterials.2005.05.049

Supplemental References (Do Not Report Using Harrick Plasma Cleaners)

Mo, X. M., Xu, C. Y., Kotaki, M., & Ramakrishna, S. (2004). Electrospun P(LLA-CL) nanofiber: A biomimetic extracellular matrix for smooth muscle cell and endothelial cell proliferation. Biomaterials, 25(10), 1883–1890. https://doi.org/10.1016/j.biomaterials.2003.08.042

Polystyrene (TPS, LCP, SRP, LDPE)

While polystyrene is often used for 2D cell culture (Tissue Culture Plastic), it lacks biocompatibility sufficient for medical usage. Plasma treatment significantly improves polystyrene biocompatibility and seeding efficiency. Spin coating polystyrene allows researchers to control the substrate’s topography, while increasing surface area and permeability. Though non-biodegradable, porous PS membranes are beneficial for the the study of bone formation, cancer progression and cell differentiation.

Cells & Tissue: Bone, Adipose, Lymphoma cancer, Epithelial, Mesenchymal stem cells

Process Gases: Oxygen, Air

POLYSTYRENE (TPS, LCP, SRP, LDPE) Articles

Alvarez-Barreto JF, Linehan SM, Shambaugh RL, and Sikavitsas VI. “Flow perfusion improves seeding of tissue engineering scaffolds with different architectures”. Ann. Biomed. Eng. 2007 35: 429–442 10.1007/s10439-006-9244-z

Caicedo-Carvajal C, Liu Q, Remache Y, Goy A, and Suh K. “Cancer Tissue Engineering: A Novel 3D Polystyrene Scaffold for In Vitro Isolation and Amplification of Lymphoma Cancer Cells from Heterogeneous Cell Mixtures”. J. Tissue Eng. 2011 2011: N/A 10.4061/2011/362326

Kumar A, Lau W, and Starly B. “Human Mesenchymal Stem Cells Expansion on Three-Dimensional (3D) Printed Poly-Styrene (PS) Scaffolds in a Perfusion Bioreactor”. Procedia CIRP 2017 65: 115-120 10.1016/j.procir.2017.04.012

Naeem F, Prestayko R, Saem S, Nowicki L, Imit M, Adronov A, and Moran-Mirabal JM. “Fabrication of conductive polymer nanofibers through SWNT supramolecular functionalization and aqueous solution processing”. Nanotechnology 2015 26: 395301 10.1088/0957-4484/26/39/395301

Lee K, Koon Y, Kim J, Chiam K, and Park S. “Astral microtubules determine the final division axis of cells confined on anisotropic surface topography”. Journal of Experimental Nanoscience 2020 70-86 10.1080/17458080.2020.1729355

 Wongkaew N, Simsek M, Heider J, Wegener J, Baeumnet A, Schreml S, and Stolwijk J. “Cytocompatibility of Mats Prepared from Different Electrospun Polymer Nanofibers”. ACS Applied Bio Materials 2020 3: 4912-4921 10.1021/acsabm.0c00426

Polyethylene Terephthalate (PET, PETE)

Polyethylene Terephthalate (PET) is a common material used for ligament or vasculature grafts due to its stability, mechanical strength, and biocompatibility. Untreated PET has poor cell adhesion properties, which can be improved by plasma. Plasma treatment increases cell and protein adsorption as well as cell distribution by increasing hydrophilicity of the surface. 

Cells & Tissue: Ligament, Bone, Endothelial, Vascular

Process Gases: Air, Oxygen

POLYETHYLENE TEREPHTHALATE (PET, PETE) Articles

Bruinink A, Siragusano D, Ettel G, Brandsberg T, Brandsberg F, Petitmermet M, Muller B, Mayer J, and Wintermantel E. “The stiffness of bone marrow cell-knit composites is increased during mechanical load”. Biomaterials 2001 22: 3169–3178 10.1016/s0142-9612(01)00069-2

Moczulska M, Bitar M, Swieszkowski W, and Bruinink A. “Biological characterization of woven fabric using two- and three-dimensional cell cultures”. J. Biomed. Mater. Res. A 2012 100A: 882–893 10.1002/jbm.a.34023

Pratt KJ, Williams SK, and Jarrell BE. “Enhanced Adherence Of Human Adult Endothelial-Cells To Plasma Discharge Modified Polyethylene Terephthalate”. J. Biomed. Mater. Res. 1989 23: 1131–1147 10.1002/jbm.820231004

Thurner P, Muller B, Beckmann F, Weitkamp T, Rau C, Muller R, Hubbell J, and Sennhauser U. “Tomography studies of human foreskin fibroblasts on polymer yarns”. Nucl. Instrum. Methods Phys. Res., Sect. B 2003 200: 397–405 10.1016/s0168-583x(02)01729-9

Supplemental References (Do Not Report Using Harrick Plasma Cleaners)

Subramaniam, A., & Sethuraman, S. (2014). Chapter 18—Biomedical Applications of Nondegradable Polymers. In S. G. Kumbar, C. T. Laurencin, & M. Deng (Eds.), Natural and Synthetic Biomedical Polymers (pp. 301–308). Elsevier. 10.1016/B978-0-12-396983-5.00019-3

Poly(ethylene oxide)/poly(butylene terephthalate) (PEOT/PBT)

Poly(ethylene oxide)/poly(butylene terephthalate) (PEOT/PBT) is a low-cost block copolymer with high cell attachment and tunable properties. Both mechanical strength and degradation can be modified, with PEOT degrading due to oxidation and PBT degrading due to hydrolysis, resulting in unique biodegradation rates.  It has been shown to be a suitable scaffold for many different types of cells including chondrocytes, fibroblasts, and keratinocytes. When exposed to plasma the substrate has improved cell attachment.

Cells & Tissue: Epithelial, Chondrocye, Fibroblasts, Kidney

Process Gases: Oxygen, Argon

POLY(ETHYLENE OXIDE)/POLY(BUTYLENE TEREPHTHALATE) (PEOT/PBT) Articles

Bettahalli N, Vicente J, Moroni L, Higuera G, van Blitterswijk C, Wessling M, and Stamatialis D. “Integration of hollow fiber membranes improves nutrient supply in three-dimensional tissue constructs”. Acta Biomater. 2011 7: 3312 — 3324 10.1016/j.actbio.2011.06.012

Moroni L, Curti M, Welti M, Korom S, Weder W, De Wijn JR, and Van Blitterswijk CA. “Anatomical 3D fiber-deposited scaffolds for tissue engineering: Designing a neotrachea”. Tissue Eng. 2007 13: 2483–2493 10.1089/ten.2006.0385

van den Bogaerdt AJ, Ulrich MMW, van Galen MJM, Reijnen L, Verkerk M, Pieper J, Lamme EN, and Middelkoop E. “Upside-down transfer of porcine keratinocytes from a porous, synthetic dressing to experimental full-thickness wounds”. Wound Repair Regen. 2004 12: 225–234 10.1111/j.1067-1927.2004.012115.x

* Damanik F, Brunelli M, Pastorino L, Ruggiero C, van Blitterswijk C, Rotmans J, and Moroni L. “Sustained delivery of growth factors with high loading efficiency in a layer by layer assembly”. Biomaterials Science 2020 8: 174-188 10.1039/C9BM00979E

Supplemental References (Do Not Report Using Harrick Plasma Cleaners)

Deschamps, A. A., Grijpma, D. W., & Feijen, J. (2001). Poly(ethylene oxide)/poly(butylene terephthalate) segmented block copolymers: The effect of copolymer composition on physical properties and degradation behavior. Polymer, 42(23), 9335–9345. https://doi.org/10.1016/S0032-3861(01)00453-0

Chao, G., Fan, L., Jia, W., Qian, Z., Gu, Y., Liu, C., Ni, X., Li, J., Deng, H., Gong, C., Gou, M., Lei, K., Huang, A., Huang, C., Yang, J., Kan, B., & Tu, M. (2007). Synthesis, characterization and hydrolytic degradation of degradable poly(butylene terephthalate)/poly(ethylene glycol) (PBT/PEG) copolymers. Journal of Materials Science: Materials in Medicine, 18(3), 449–455. https://doi.org/10.1007/s10856-007-2004-2

Other Cell Scaffold Materials

Other cell scaffolding materials that have been used in conjunction with Harrick Plasma Cleaners include Acrylonitrile Butadiene Styrene (ABS), Deproteinized Bovine, Hydroxyapatite, Paper & Cellulose, Polycarbonate, Polylysine (PLL, PDL),  Poly(methyl methacrylate) (PMMA),  Poly(propylene fumarate) (PPF), Polysulfone PSU, Polyurethane (PUA), SU-8, and Titanium Fiber Mesh.

For more information on these scaffold materials, refer to the articles listed below.

Other Cell Scaffold Materials Articles

Acrylonitrile Butadiene Styrene (ABS)

Mozdzen LC, Rodgers R, Banks JM, Bailey RC, and Harley BAC. “Increasing the strength and bioactivity of collagen scaffolds using customizable arrays of 3D-printed polymer fibers”. Acta Biomater. 2016 33: 25–33 10.1016/j.actbio.2016.02.004

Deproteinized Bovine

Li Q, Zhou G, Yu X, Wang T, Xi Y, and Tang Z. “Porous deproteinized bovine bone scaffold with three-dimensional localized drug delivery system using chitosan microspheres”. Biomed. Eng. Online 2015 14: 1 10.1186/s12938-015-0028-2

Hydroxyapatite

Burgio F, Rimmer N, Pieles U, Buschmann J, and Beaufils-Hugot M. “Characterization and in ovo vascularisation of a 3D-printed hydroxyapatite scaffold with different extracellular matrix coatings under perfusion culture”. Biology Open 2018 10.1242/bio.034488

Kim J, Han T, Kim M, Oh D, Kang S, Kim G, Kwon T-Y, Kim K-H, Lee K-B, Son J, and Choi S. “Osteogenic evaluation of calcium phosphate scaffold with drug-loaded poly (lactic-co-glycolic acid) microspheres in beagle dogs”. Tiss Eng. Regen. Med. 2012 9: 175–183 10.1007/s13770-012-0175-5

Zhou G, Yu X, Tai J, Han F, Yan M, Xi Y, Liu M, Wu Q, and Fan Y. “Research on a novel chitosan microsphere/scaffold system by double crosslinkers”. Dent. Mater. J. 2016 10.4012/dmj.2015-227

Paper & Cellulose

Dermutz H, Thompson-Steckel G, Forro C, de Lange V, Dorwling-Carter L, Voros J, and Demko L. “Paper-based patterned 3D neural cultures as a tool to study network activity on multielectrode arrays”. RSC Adv. 2017 7: 39359-39371 10.1039/C7RA00971B

Polycarbonate

Neiman JAS, Raman R, Chan V, Rhoads MG, Raredon MSB, Velazquez JJ, Dyer RL, Bashir R, Hammond PT, and Griffith LG. “Photopatterning of hydrogel scaffolds coupled to filter materials using stereolithography for perfused 3D culture of hepatocytes”. Biotechnol. Bioeng. 2015 112: 777–787 10.1002/bit.25494

Polylysine (PLL, PDL)

Johnson CD, D’Amato AR, Puhl DL, Wich DM, Vesperman A, and Gilbert RJ. “Electrospun fiber surface nanotopography influences astrocyte-mediated neurite outgrowth”. Biomed. Mater. 2018 13: 54101 10.1088/1748-605X/aac4de

Poly(methyl methacrylate) (PMMA)

Steiner G, Zimmerer C, and Salzer R. “Characterization of metal-supported poly(methyl methacrylate) microstructures by FTIR imaging spectroscopy”. Langmuir 2006 22: 4125–4130 10.1021/la053221x

Poly(propylene fumarate) (PPF)

Dean D, Wallace J, Siblani A, Wang M, Kim K, Mikos A, and Fisher J. “Continuous digital light processing (cDLP): highly accurate additive manufacturing of tissue engineered bone scaffolds”. Virtual and Physical Prototyping 2012 7: 13 — 24 10.1080/17452759.2012.673152

Vehof JWM, Fisher JP, Dean D, van der Waerden JPCM, Spauwen PHM, Mikos AG, and Jansen JA. “Bone formation in transforming growth factor beta-1-coated porous poly(propylene fumarate) scaffolds”. J. Biomed. Mater. Res. 2002 60: 241–251 10.1002/jbm.10073

Polysulfone PSU

Ma ZW, Kotaki M, and Ramarkrishna S. “Surface modified nonwoven polysulphone (PSU) fiber mesh by electrospinning: A novel affinity membrane”. J. Membr. Sci. 2006 272: 179–187 10.1016/j.memsci.2005.07.038

Polyurethane (PUA)

Adolph EJ, Pollins AC, Cardwell NL, Davidson JM, Guelcher SA, and Nanney LB. “Biodegradable lysine-derived polyurethane scaffolds promote healing in a porcine full-thickness excisional wound model”. J. Biomater. Sci., Polym. Ed. 2014 25: 1973–1985 10.1080/09205063.2014.965997

Lee MR, Kwon KW, Jung H, Kim HN, Suh KY, Kim K, and Kim KS. “Direct differentiation of human embryonic stem cells into selective neurons on nanoscale ridge/groove pattern arrays”. Biomaterials 2010 31: 4360–4366 10.1016/j.biomaterials.2010.02.012

SU-8

Ouyang X, Zhang K, Wu J, Wong DS-H, Feng Q, Bian L, and Zhang AP. “Optical μ-Printing of Cellular-Scale Microscaffold Arrays for 3D Cell Culture”. Sci. Rep. 2017 7: 8880 10.1038/s41598-017-08598-3

Titanium Fiber Mesh

Vehof JWM, Mahmood J, Takita H, van’t Hof MA, Kuboki Y, Spauwen PHM, and Jansen JA. “Ectopic bone formation in titanium mesh loaded with bone morphogenetic protein and coated with calcium phosphate”. Plast. Reconstr. Surg. 2001 108: 434–443 10.1097/00006534-200108000-00024

Vehof JWM, Haus MTU, de Ruijter AE, Spauwen PHM, and Jansen JA. “Bone formation in Transforming Growth Factor beta-I-loaded titanium fiber mesh implants”. Clin. Oral Implants Res. 2002 13: 94–102 10.1034/j.1600-0501.2002.130112.x

Vehof JWM, Spauwen PHM, and Jansen JA. “Bone formation in calcium-phosphate-coated titanium mesh”. Biomaterials 2000 21: 2003–2009 10.1016/s0142-9612(00)00094-6

Harrick Plasma is a leading supplier of plasma equipment to the research community. We have been providing quality tabletop plasma devices specifically designed for laboratory and R&D use for over 30 years.