최소 단어 이상 선택하여야 합니다.
최대 10 단어까지만 선택 가능합니다.
다음과 같은 기능을 한번의 로그인으로 사용 할 수 있습니다.
NTIS 바로가기International journal of molecular sciences, v.20 no.3, 2019년, pp.636 -
Mariani, Erminia (Laboratorio di Immunoreumatologia e rigenerazione tissutale, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1) , Lisignoli, Gina (gina.lisignoli@ior.it (G.L.)) , Borzì, Rosa Maria (rosamaria.borzì) , Pulsatelli, Lia (@ior.it (R.M.B.))
The perspectives of regenerative medicine are still severely hampered by the host response to biomaterial implantation, despite the robustness of technologies that hold the promise to recover the functionality of damaged organs and tissues. In this scenario, the cellular and molecular events that de...
1. Vishwakarma A. Bhise N.S. Evangelista M.B. Rouwkema J. Dokmeci M.R. Ghaemmaghami A.M. Vrana N.E. Khademhosseini A. Engineering Immunomodulatory Biomaterials to Tune the Inflammatory Response Trends Biotechnol. 2016 34 470 482 10.1016/j.tibtech.2016.03.009 27138899
2. Dhandayuthapani B. Yoshida Y. Maekawa T. Kumar D.S. Polymeric scaffolds in tissue engineering application: A review Int. J. Polym. Sci. 2011 2011 290602 10.1155/2011/290602
3. Wiles K. Fishman J.M. De Coppi P. Birchall M.A. The Host Immune Response to Tissue-Engineered Organs: Current Problems and Future Directions Tissue Eng. Part B Rev. 2016 22 208 219 10.1089/ten.teb.2015.0376 26701069
4. Roseti L. Parisi V. Petretta M. Cavallo C. Desando G. Bartolotti I. Grigolo B. Scaffolds for Bone Tissue Engineering: State of the art and new perspectives Mater. Sci. Eng. C Mater. Biol. Appl. 2017 78 1246 1262 10.1016/j.msec.2017.05.017 28575964
5. Rossi F. Santoro M. Perale G. Polymeric scaffolds as stem cell carriers in bone repair J. Tissue Eng. Regen. Med. 2015 9 1093 1119 10.1002/term.1827 24668819
6. Wilson C.J. Clegg R.E. Leavesley D.I. Pearcy M.J. Mediation of biomaterial-cell interactions by adsorbed proteins: A review Tissue Eng. 2005 11 1 18 10.1089/ten.2005.11.1 15738657
7. Keselowsky B.G. Collard D.M. Garcia A.J. Surface chemistry modulates focal adhesion composition and signaling through changes in integrin binding Biomaterials 2004 25 5947 5954 10.1016/j.biomaterials.2004.01.062 15183609
8. Gasperini L. Mano J.F. Reis R.L. Natural polymers for the microencapsulation of cells J. R. Soc. Interface 2014 11 20140817 10.1098/rsif.2014.0817 25232055
9. Thevenot P. Hu W. Tang L. Surface chemistry influences implant biocompatibility Curr. Top. Med. Chem. 2008 8 270 280 18393890
10. Matlaga B.F. Yasenchak L.P. Salthouse T.N. Tissue response to implanted polymers: The significance of sample shape J. Biomed. Mater. Res. 1976 10 391 397 10.1002/jbm.820100308 1270456
11. Delves P.J. Martin S.J. Burton D.R. Roitt I.M. Roitt’s Essential Immunology John Wiley & Sons Hoboken, NJ, USA 2011
12. Anderson J.M. Rodriguez A. Chang D.T. Foreign body reaction to biomaterials Semin. Immunol. 2008 20 86 100 10.1016/j.smim.2007.11.004 18162407
13. Reid B. Gibson M. Singh A. Taube J. Furlong C. Murcia M. Elisseeff J. PEG hydrogel degradation and the role of the surrounding tissue environment J. Tissue Eng. Regen. Med. 2015 9 315 318 10.1002/term.1688 23495204
14. Anderson J.M. Inflammatory response to implants ASAIO Trans. 1988 34 101 107 10.1097/00002480-198804000-00005 3285869
15. Boccafoschi F. Mosca C. Cannas M. Cardiovascular biomaterials: When the inflammatory response helps to efficiently restore tissue functionality? J. Tissue Eng. Regen. Med. 2014 8 253 267 10.1002/term.1526 22730232
16. Brown B.N. Badylak S.F. Expanded applications, shifting paradigms and an improved understanding of host-biomaterial interactions Acta Biomater. 2013 9 4948 4955 10.1016/j.actbio.2012.10.025 23099303
17. Christo S.N. Diener K.R. Bachhuka A. Vasilev K. Hayball J.D. Innate Immunity and Biomaterials at the Nexus: Friends or Foes Biomed. Res. Int. 2015 2015 342304 10.1155/2015/342304 26247017
18. Chung L. Maestas D.R. Jr. Housseau F. Elisseeff J.H. Key players in the immune response to biomaterial scaffolds for regenerative medicine Adv. Drug Deliv. Rev. 2017 114 184 192 10.1016/j.addr.2017.07.006 28712923
19. Franz S. Rammelt S. Scharnweber D. Simon J.C. Immune responses to implants—A review of the implications for the design of immunomodulatory biomaterials Biomaterials 2011 32 6692 6709 10.1016/j.biomaterials.2011.05.078 21715002
20. Ward W.K. A review of the foreign-body response to subcutaneously-implanted devices: The role of Macrophages and cytokines in biofouling and fibrosis J. Diabetes Sci. Technol. 2008 2 768 777 10.1177/193229680800200504 19885259
21. Tang L. Eaton J.W. Fibrin(ogen) mediates acute inflammatory responses to biomaterials J. Exp. Med. 1993 178 2147 2156 10.1084/jem.178.6.2147 8245787
22. Milleret V. Buzzi S. Gehrig P. Ziogas A. Grossmann J. Schilcher K. Zinkernagel A.S. Zucker A. Ehrbar M. Protein adsorption steers blood contact activation on engineered cobalt chromium alloy oxide layers Acta Biomater. 2015 24 343 351 10.1016/j.actbio.2015.06.020 26102336
24. Zhang L. Cao Z. Bai T. Carr L. Ella-Menye J.R. Irvin C. Ratner B.D. Jiang S. Zwitterionic hydrogels implanted in mice resist the foreign-body reaction Nat. Biotechnol. 2013 31 553 556 10.1038/nbt.2580 23666011
25. Kim Y.K. Que R. Wang S.W. Liu W.F. Modification of biomaterials with a self-protein inhibits the macrophage response Adv. Healthc. Mater. 2014 3 989 994 10.1002/adhm.201300532 24573988
26. Ekdahl K.N. Lambris J.D. Elwing H. Ricklin D. Nilsson P.H. Teramura Y. Nicholls I.A. Nilsson B. Innate immunity activation on biomaterial surfaces: A mechanistic model and coping strategies Adv. Drug Deliv. Rev. 2011 63 1042 1050 10.1016/j.addr.2011.06.012 21771620
27. Gorbet M.B. Sefton M.V. Biomaterial-associated thrombosis: Roles of coagulation factors, complement, platelets and leukocytes The Biomaterials: Silver Jubilee Compendium Elsevier Ltd. Oxford, UK 2006
28. Chiumiento A. Lamponi S. Barbucci R. Role of fibrinogen conformation in platelet activation Biomacromolecules 2007 8 523 531 10.1021/bm060664m 17291077
29. Wu Y. Simonovsky F.I. Ratner B.D. Horbett T.A. The role of adsorbed fibrinogen in platelet adhesion to polyurethane surfaces: A comparison of surface hydrophobicity, protein adsorption, monoclonal antibody binding, and platelet adhesion J. Biomed. Mater. Res. A 2005 74 722 738 10.1002/jbm.a.30381 16037938
30. Engelmann B. Massberg S. Thrombosis as an intravascular effector of innate immunity Nat. Rev. Immunol. 2013 13 34 45 10.1038/nri3345 23222502
31. Andersson J. Ekdahl K.N. Lambris J.D. Nilsson B. Binding of C3 fragments on top of adsorbed plasma proteins during complement activation on a model biomaterial surface Biomaterials 2005 26 1477 1485 10.1016/j.biomaterials.2004.05.011 15522749
32. Hed J. Johansson M. Lindroth M. Complement activation according to the alternate pathway by glass and plastic surfaces and its role in neutrophil adhesion Immunol. Lett. 1984 8 295 299 10.1016/0165-2478(84)90013-0 6526424
33. Nilsson B. Ekdahl K.N. Mollnes T.E. Lambris J.D. The role of complement in biomaterial-induced inflammation Mol. Immunol. 2007 44 82 94 10.1016/j.molimm.2006.06.020 16905192
34. Flick M.J. Du X. Witte D.P. Jirouskova M. Soloviev D.A. Busuttil S.J. Plow E.F. Degen J.L. Leukocyte engagement of fibrin(ogen) via the integrin receptor alphaMbeta2/Mac-1 is critical for host inflammatory response in vivo J. Clin. Investig. 2004 113 1596 1606 10.1172/JCI20741 15173886
35. Szaba F.M. Smiley S.T. Roles for thrombin and fibrin(ogen) in cytokine/chemokine production and macrophage adhesion in vivo Blood 2002 99 1053 1059 10.1182/blood.V99.3.1053 11807012
36. Esche C. Stellato C. Beck L.A. Chemokines: Key players in innate and adaptive immunity J. Investig. Dermatol. 2005 125 615 628 10.1111/j.0022-202X.2005.23841.x 16185259
37. McFarland C.D. Thomas C.H. DeFilippis C. Steele J.G. Healy K.E. Protein adsorption and cell attachment to patterned surfaces J. Biomed. Mater. Res. 2000 49 200 210 10.1002/(SICI)1097-4636(200002)49:2 3.0.CO;2-L 10571906
38. Groth T. Zlatanov I. Altankov G. Adhesion of Human Peripheral Lymphocytes on Biomaterials Preadsorbed with Fibronectin and Vitronectin J. Biomater. Sci. Polym. Ed. 1995 10.1163/156856295X00111
39. Jenney C.R. Anderson J.M. Adsorbed serum proteins responsible for surface dependent human macrophage behavior J. Biomed. Mater. Res. 2000 49 435 447 10.1002/(SICI)1097-4636(20000315)49:4 3.0.CO;2-Y 10602077
40. McNally A.K. Jones J.A. Macewan S.R. Colton E. Anderson J.M. Vitronectin is a critical protein adhesion substrate for IL-4-induced foreign body giant cell formation J. Biomed. Mater. Res. A 2008 86 535 543 10.1002/jbm.a.31658 17994558
41. Keselowsky B.G. Bridges A.W. Burns K.L. Tate C.C. Babensee J.E. LaPlaca M.C. Garcia A.J. Role of plasma fibronectin in the foreign body response to biomaterials Biomaterials 2007 28 3626 3631 10.1016/j.biomaterials.2007.04.035 17521718
42. Sheikh Z. Brooks P.J. Barzilay O. Fine N. Glogauer M. Macrophages, Foreign Body Giant Cells and Their Response to Implantable Biomaterials Materials 2015 8 5671 5701 10.3390/ma8095269 28793529
43. Shen M. Garcia I. Maier R.V. Horbett T.A. Effects of adsorbed proteins and surface chemistry on foreign body giant cell formation, tumor necrosis factor alpha release and procoagulant activity of monocytes J. Biomed. Mater. Res. A 2004 70 533 541 10.1002/jbm.a.30069 15307157
44. Keselowsky B.G. Collard D.M. Garcia A.J. Integrin binding specificity regulates biomaterial surface chemistry effects on cell differentiation Proc. Natl. Acad. Sci. USA 2005 102 5953 5957 10.1073/pnas.0407356102 15827122
45. McNally A.K. Macewan S.R. Anderson J.M. alpha subunit partners to beta1 and beta2 integrins during IL-4-induced foreign body giant cell formation J. Biomed. Mater. Res. A 2007 82 568 574 10.1002/jbm.a.31161 17311314
46. Akira S. Uematsu S. Takeuchi O. Pathogen recognition and innate immunity Cell 2006 124 783 801 10.1016/j.cell.2006.02.015 16497588
47. Kumar H. Kawai T. Akira S. Pathogen recognition by the innate immune system Int. Rev. Immunol. 2011 30 16 34 10.3109/08830185.2010.529976 21235323
48. Babensee J.E. Interaction of dendritic cells with biomaterials Semin. Immunol. 2008 20 101 108 10.1016/j.smim.2007.10.013 18054498
49. Bianchi M.E. DAMPs, PAMPs and alarmins: All we need to know about danger J. Leukoc. Biol. 2007 81 1 5 10.1189/jlb.0306164
50. Grandjean-Laquerriere A. Tabary O. Jacquot J. Richard D. Frayssinet P. Guenounou M. Laurent-Maquin D. Laquerriere P. Gangloff S. Involvement of toll-like receptor 4 in the inflammatory reaction induced by hydroxyapatite particles Biomaterials 2007 28 400 404 10.1016/j.biomaterials.2006.09.015 17010424
51. Labow R.S. Meek E. Santerre J.P. Neutrophil-mediated biodegradation of medical implant materials J. Cell. Physiol. 2001 186 95 103 10.1002/1097-4652(200101)186:1 3.0.CO;2-0 11147818
52. Nimeri G. Majeed M. Elwing H. Ohman L. Wettero J. Bengtsson T. Oxygen radical production in neutrophils interacting with platelets and surface-immobilized plasma proteins: Role of tyrosine phosphorylation J. Biomed. Mater. Res. A 2003 67 439 447 10.1002/jbm.a.10081 14566784
53. Nimeri G. Ohman L. Elwing H. Wettero J. Bengtsson T. The influence of plasma proteins and platelets on oxygen radical production and F-actin distribution in neutrophils adhering to polymer surfaces Biomaterials 2002 23 1785 1795 10.1016/S0142-9612(01)00305-2 11950049
54. Wettero J. Bengtsson T. Tengvall P. Complement activation on immunoglobulin G-coated hydrophobic surfaces enhances the release of oxygen radicals from neutrophils through an actin-dependent mechanism J. Biomed. Mater. Res. 2000 51 742 751 10.1002/1097-4636(20000915)51:4 3.0.CO;2-D 10880124
55. Brinkmann V. Reichard U. Goosmann C. Fauler B. Uhlemann Y. Weiss D.S. Weinrauch Y. Zychlinsky A. Neutrophil extracellular traps kill bacteria Science 2004 303 1532 1535 10.1126/science.1092385 15001782
56. Kolaczkowska E. Kubes P. Neutrophil recruitment and function in health and inflammation Nat. Rev. Immunol. 2013 13 159 175 10.1038/nri3399 23435331
57. Hahn J. Schauer C. Czegley C. Kling L. Petru L. Schmid B. Weidner D. Reinwald C. Biermann M.H.C. Blunder S. Aggregated neutrophil extracellular traps resolve inflammation by proteolysis of cytokines and chemokines and protection from antiproteases FASEB J. 2018 10.1096/fj.201800752R 30130433
58. Schauer C. Janko C. Munoz L.E. Zhao Y. Kienhofer D. Frey B. Lell M. Manger B. Rech J. Naschberger E. Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines Nat. Med. 2014 20 511 517 10.1038/nm.3547 24784231
59. Selders G.S. Fetz A.E. Radic M.Z. Bowlin G.L. An overview of the role of neutrophils in innate immunity, inflammation and host-biomaterial integration Regen. Biomater. 2017 4 55 68 10.1093/rb/rbw041 28149530
60. Branzk N. Lubojemska A. Hardison S.E. Wang Q. Gutierrez M.G. Brown G.D. Papayannopoulos V. Neutrophils sense microbe size and selectively release neutrophil extracellular traps in response to large pathogens Nat. Immunol. 2014 15 1017 1025 10.1038/ni.2987 25217981
61. McNally A.K. Anderson J.M. Macrophage fusion and multinucleated giant cells of inflammation Adv. Exp. Med. Biol. 2011 713 97 111 10.1007/978-94-007-0763-4_7 21432016
62. Scapini P. Lapinet-Vera J.A. Gasperini S. Calzetti F. Bazzoni F. Cassatella M.A. The neutrophil as a cellular source of chemokines Immunol. Rev. 2000 177 195 203 10.1034/j.1600-065X.2000.17706.x 11138776
63. Yamashiro S. Kamohara H. Wang J.M. Yang D. Gong W.H. Yoshimura T. Phenotypic and functional change of cytokine-activated neutrophils: Inflammatory neutrophils are heterogeneous and enhance adaptive immune responses J. Leukoc. Biol. 2001 69 698 704 11358976
64. Altieri D.C. Mannucci P.M. Capitanio A.M. Binding of fibrinogen to human monocytes J. Clin. Investig. 1986 78 968 976 10.1172/JCI112687 3760194
65. Trezzini C. Jungi T.W. Kuhnert P. Peterhans E. Fibrinogen association with human monocytes: Evidence for constitutive expression of fibrinogen receptors and for involvement of Mac-1 (CD18, CR3) in the binding Biochem. Biophys. Res. Commun. 1988 156 477 484 10.1016/S0006-291X(88)80866-0 3178847
66. Mesure L. De Visscher G. Vranken I. Lebacq A. Flameng W. Gene expression study of monocytes/macrophages during early foreign body reaction and identification of potential precursors of myofibroblasts PLoS ONE 2010 5 e12949 10.1371/journal.pone.0012949 20886081
67. Badylak S.F. Gilbert T.W. Immune response to biologic scaffold materials Semin. Immunol. 2008 20 109 116 10.1016/j.smim.2007.11.003 18083531
68. Sridharan R. Cameron A.R. Kelly D.J. Kearney C.J. O’Brien F.J. Biomaterial Based Modulation of macrophage polarization: A review and suggested design principles Mater. Today 2015 18 313 325 10.1016/j.mattod.2015.01.019
69. Jones J.A. Chang D.T. Meyerson H. Colton E. Kwon I.K. Matsuda T. Anderson J.M. Proteomic analysis and quantification of cytokines and chemokines from biomaterial surface-adherent macrophages and foreign body giant cells J. Biomed. Mater. Res. A 2007 83 585 596 10.1002/jbm.a.31221 17503526
70. Lynn A.D. Kyriakides T.R. Bryant S.J. Characterization of the in vitro macrophage response and in vivo host response to poly(ethylene glycol)-based hydrogels J. Biomed. Mater. Res. A 2010 93 941 953 10.1002/jbm.a.32595 19708075
71. Zhao Q. Topham N. Anderson J.M. Hiltner A. Lodoen G. Payet C.R. Foreign-body giant cells and polyurethane biostability: In vivo correlation of cell adhesion and surface cracking J. Biomed. Mater. Res. 1991 25 177 183 10.1002/jbm.820250205 2055915
72. Labrousse A.M. Meunier E. Record J. Labernadie A. Beduer A. Vieu C. Ben Safta T. Maridonneau-Parini I. Frustrated phagocytosis on micro-patterned immune complexes to characterize lysosome movements in live macrophages Front. Immunol. 2011 2 51 10.3389/fimmu.2011.00051 22566841
73. Underhill D.M. Goodridge H.S. Information processing during phagocytosis Nat. Rev. Immunol. 2012 12 492 502 10.1038/nri3244 22699831
74. Mosser D.M. Edwards J.P. Exploring the full spectrum of macrophage activation Nat. Rev. Immunol. 2008 8 958 969 10.1038/nri2448 19029990
75. Garg K. Pullen N.A. Oskeritzian C.A. Ryan J.J. Bowlin G.L. Macrophage functional polarization (M1/M2) in response to varying fiber and pore dimensions of electrospun scaffolds Biomaterials 2013 34 4439 4451 10.1016/j.biomaterials.2013.02.065 23515178
76. Broughton G. 2nd Janis J.E. Attinger C.E. The basic science of wound healing Plast. Reconstr. Surg. 2006 117 12S 34S 10.1097/01.prs.0000225430.42531.c2 16799372
77. Kyriakides T.R. Foster M.J. Keeney G.E. Tsai A. Giachelli C.M. Clark-Lewis I. Rollins B.J. Bornstein P. The CC chemokine ligand, CCL2/MCP1, participates in macrophage fusion and foreign body giant cell formation Am. J. Pathol. 2004 165 2157 2166 10.1016/S0002-9440(10)63265-8 15579457
78. Brodbeck W.G. Macewan M. Colton E. Meyerson H. Anderson J.M. Lymphocytes and the foreign body response: Lymphocyte enhancement of macrophage adhesion and fusion J. Biomed. Mater. Res. A 2005 74 222 229 10.1002/jbm.a.30313 15948198
79. Burd P.R. Thompson W.C. Max E.E. Mills F.C. Activated mast cells produce interleukin 13 J. Exp. Med. 1995 181 1373 1380 10.1084/jem.181.4.1373 7535336
80. Venkayya R. Lam M. Willkom M. Grunig G. Corry D.B. Erle D.J. The Th2 lymphocyte products IL-4 and IL-13 rapidly induce airway hyperresponsiveness through direct effects on resident airway cells Am. J. Respir. Cell Mol. Biol. 2002 26 202 208 10.1165/ajrcmb.26.2.4600 11804871
81. Rezzani R. Rodella L. Tartaglia G.M. Paganelli C. Sapelli P. Bianchi R. Mast cells and the inflammatory response to different implanted biomaterials Arch. Histol. Cytol. 2004 67 211 217 10.1679/aohc.67.211 15570886
82. Tang L. Jennings T.A. Eaton J.W. Mast cells mediate acute inflammatory responses to implanted biomaterials Proc. Natl. Acad. Sci. USA 1998 95 8841 8846 10.1073/pnas.95.15.8841 9671766
83. Christenson L. Wahlberg L. Aebischer P. Mast cells and tissue reaction to intraperitoneally implanted polymer capsules J. Biomed. Mater. Res. 1991 25 1119 1131 10.1002/jbm.820250906 1778997
84. Zdolsek J. Eaton J.W. Tang L. Histamine release and fibrinogen adsorption mediate acute inflammatory responses to biomaterial implants in humans J. Transl. Med. 2007 5 31 10.1186/1479-5876-5-31 17603911
85. Kanbe N. Kurosawa M. Nagata H. Saitoh H. Miyachi Y. Cord blood-derived human cultured mast cells produce transforming growth factor beta1 Clin. Exp. Allergy 1999 29 105 113 10.1046/j.1365-2222.1999.00459.x 10051709
86. Ribatti D. Crivellato E. Mast cells, angiogenesis, and tumour growth Biochim. Biophys. Acta 2012 1822 2 8 10.1016/j.bbadis.2010.11.010 21130163
87. Tacchini-Cottier F. Zweifel C. Belkaid Y. Mukankundiye C. Vasei M. Launois P. Milon G. Louis J.A. An immunomodulatory function for neutrophils during the induction of a CD4+ Th2 response in BALB/c mice infected with Leishmania major J. Immunol. 2000 165 2628 2636 10.4049/jimmunol.165.5.2628 10946291
88. Ma Y. Yabluchanskiy A. Iyer R.P. Cannon P.L. Flynn E.R. Jung M. Henry J. Cates C.A. Deleon-Pennell K.Y. Lindsey M.L. Temporal neutrophil polarization following myocardial infarction Cardiovasc. Res. 2016 110 51 61 10.1093/cvr/cvw024 26825554
89. Chen F. Wu W. Millman A. Craft J.F. Chen E. Patel N. Boucher J.L. Urban J.F. Jr. Kim C.C. Gause W.C. Neutrophils prime a long-lived effector macrophage phenotype that mediates accelerated helminth expulsion Nat. Immunol. 2014 15 938 946 10.1038/ni.2984 25173346
90. Yokoyama M. Nakahashi T. Nishimura T. Maeda M. Inoue S. Kataoka K. Sakurai Y. Adhesion behavior of rat lymphocytes to poly(ether)-poly(amino acid) block and graft copolymers J. Biomed. Mater. Res. 1986 20 867 878 10.1002/jbm.820200702 3760003
91. Curtsinger J.M. Mescher M.F. Inflammatory cytokines as a third signal for T cell activation Curr. Opin. Immunol. 2010 22 333 340 10.1016/j.coi.2010.02.013 20363604
92. Curtsinger J.M. Schmidt C.S. Mondino A. Lins D.C. Kedl R.M. Jenkins M.K. Mescher M.F. Inflammatory cytokines provide a third signal for activation of naive CD4+ and CD8+ T cells J. Immunol. 1999 162 3256 3262 10092777
93. Rodriguez A. Voskerician G. Meyerson H. MacEwan S.R. Anderson J.M. T cell subset distributions following primary and secondary implantation at subcutaneous biomaterial implant sites J. Biomed. Mater. Res. A 2008 85 556 565 10.1002/jbm.a.31562 17729264
94. Chang D.T. Colton E. Anderson J.M. Paracrine and juxtacrine lymphocyte enhancement of adherent macrophage and foreign body giant cell activation J. Biomed. Mater. Res. A 2009 89 490 498 10.1002/jbm.a.31981 18437695
95. Anderson J.M. In Vitro and In Vivo Monocyte, Macrophage, Foreign Body Giant Cell, and Lymphocyte Interactions with Biomaterials Biological Interactions on Materials Surfaces Springer New York, NY, USA 2009
96. Shen E.C. Chou T.C. Gau C.H. Tu H.P. Chen Y.T. Fu E. Releasing growth factors from activated human platelets after chitosan stimulation: A possible bio-material for platelet-rich plasma preparation Clin. Oral Implants Res. 2006 17 572 578 10.1111/j.1600-0501.2004.01241.x 16958699
97. Bao P. Kodra A. Tomic-Canic M. Golinko M.S. Ehrlich H.P. Brem H. The role of vascular endothelial growth factor in wound healing J. Surg. Res. 2009 153 347 358 10.1016/j.jss.2008.04.023 19027922
98. Chen S. Jones J.A. Xu Y. Low H.Y. Anderson J.M. Leong K.W. Characterization of topographical effects on macrophage behavior in a foreign body response model Biomaterials 2010 31 3479 3491 10.1016/j.biomaterials.2010.01.074 20138663
99. Garg K. Sell S.A. Madurantakam P. Bowlin G.L. Angiogenic potential of human macrophages on electrospun bioresorbable vascular grafts Biomed. Mater. 2009 4 031001 10.1088/1748-6041/4/3/031001 19372619
100. Mustafa K. Rubinstein J. Lopez B.S. Arvidson K. Production of transforming growth factor beta1 and prostaglandin E2 by osteoblast-like cells cultured on titanium surfaces blasted with TiO2 particles Clin. Oral Implants Res. 2003 14 50 56 10.1034/j.1600-0501.2003.140107.x 12562365
101. Diegelmann R.F. Evans M.C. Wound healing: An overview of acute, fibrotic and delayed healing Front. Biosci. 2004 9 283 289 10.2741/1184 14766366
102. Castro P.R. Marques S.M. Campos P.P. Cardoso C.C. Sampaio F.P. Ferreira M.A. Andrade S.P. Kinetics of implant-induced inflammatory angiogenesis in abdominal muscle wall in mice Microvasc. Res. 2012 84 9 15 10.1016/j.mvr.2012.04.003 22549137
103. Oviedo-Socarras T. Vasconcelos A.C. Barbosa I.X. Pereira N.B. Campos P.P. Andrade S.P. Diabetes alters inflammation, angiogenesis, and fibrogenesis in intraperitoneal implants in rats Microvasc. Res. 2014 93 23 29 10.1016/j.mvr.2014.02.011 24594441
104. Ratner B.D. Reducing capsular thickness and enhancing angiogenesis around implant drug release systems J. Control. Release 2002 78 211 218 10.1016/S0168-3659(01)00502-8 11772462
105. Ratner B.D. Bryant S.J. Biomaterials: Where we have been and where we are going Annu. Rev. Biomed. Eng. 2004 6 41 75 10.1146/annurev.bioeng.6.040803.140027 15255762
106. Rockey D.C. Bell P.D. Hill J.A. Fibrosis—A common pathway to organ injury and failure N. Engl. J. Med. 2015 372 1138 1149 10.1056/NEJMra1300575 25785971
107. Zaiss D.M. van Loosdregt J. Gorlani A. Bekker C.P. Grone A. Sibilia M. van Bergen en Henegouwen P.M. Roovers R.C. Coffer P.J. Sijts A.J. Amphiregulin enhances regulatory T cell-suppressive function via the epidermal growth factor receptor Immunity 2013 38 275 284 10.1016/j.immuni.2012.09.023 23333074
108. Arpaia N. Green J.A. Moltedo B. Arvey A. Hemmers S. Yuan S. Treuting P.M. Rudensky A.Y. A Distinct Function of Regulatory T Cells in Tissue Protection Cell 2015 162 1078 1089 10.1016/j.cell.2015.08.021 26317471
109. Zaiss D.M.W. Gause W.C. Osborne L.C. Artis D. Emerging functions of amphiregulin in orchestrating immunity, inflammation, and tissue repair Immunity 2015 42 216 226 10.1016/j.immuni.2015.01.020 25692699
110. Burzyn D. Kuswanto W. Kolodin D. Shadrach J.L. Cerletti M. Jang Y. Sefik E. Tan T.G. Wagers A.J. Benoist C. A special population of regulatory T cells potentiates muscle repair Cell 2013 155 1282 1295 10.1016/j.cell.2013.10.054 24315098
111. Weirather J. Hofmann U.D. Beyersdorf N. Ramos G.C. Vogel B. Frey A. Ertl G. Kerkau T. Frantz S. Foxp3+ CD4+ T cells improve healing after myocardial infarction by modulating monocyte/macrophage differentiation Circ. Res. 2014 115 55 67 10.1161/CIRCRESAHA.115.303895 24786398
112. Nosbaum A. Prevel N. Truong H.A. Mehta P. Ettinger M. Scharschmidt T.C. Ali N.H. Pauli M.L. Abbas A.K. Rosenblum M.D. Cutting Edge: Regulatory T Cells Facilitate Cutaneous Wound Healing J. Immunol. 2016 196 2010 2014 10.4049/jimmunol.1502139 26826250
113. Vinish M. Cui W. Stafford E. Bae L. Hawkins H. Cox R. Toliver-Kinsky T. Dendritic cells modulate burn wound healing by enhancing early proliferation Wound Repair Regen. 2016 24 6 13 10.1111/wrr.12388 26609910
114. Anzai A. Anzai T. Nagai S. Maekawa Y. Naito K. Kaneko H. Sugano Y. Takahashi T. Abe H. Mochizuki S. Regulatory role of dendritic cells in postinfarction healing and left ventricular remodeling Circulation 2012 125 1234 1245 10.1161/CIRCULATIONAHA.111.052126 22308302
115. Rutella S. Danese S. Leone G. Tolerogenic dendritic cells: Cytokine modulation comes of age Blood 2006 108 1435 1440 10.1182/blood-2006-03-006403 16684955
116. Lutz M.B. Schuler G. Immature, semi-mature and fully mature dendritic cells: Which signals induce tolerance or immunity? Trends Immunol. 2002 23 445 449 10.1016/S1471-4906(02)02281-0 12200066
117. Babensee J.E. Stein M.M. Moore L. Interconnections between inflammatory and immune responses in tissue engineering Ann. N. Y. Acad. Sci. 2002 961 360 363 10.1111/j.1749-6632.2002.tb03124.x 12081940
118. Yoshida M. Babensee J.E. Poly(lactic-co-glycolic acid) enhances maturation of human monocyte-derived dendritic cells J. Biomed. Mater. Res. A 2004 71 45 54 10.1002/jbm.a.30131 15368253
119. Keselowsky B.G. Lewis J.S. Dendritic cells in the host response to implanted materials Semin. Immunol. 2017 29 33 40 10.1016/j.smim.2017.04.002 28487131
120. Thaiss C.A. Zmora N. Levy M. Elinav E. The microbiome and innate immunity Nature 2016 535 65 74 10.1038/nature18847 27383981
121. Wu L. Ong S. Talor M.V. Barin J.G. Baldeviano G.C. Kass D.A. Bedja D. Zhang H. Sheikh A. Margolick J.B. Cardiac fibroblasts mediate IL-17A-driven inflammatory dilated cardiomyopathy J. Exp. Med. 2014 211 1449 1464 10.1084/jem.20132126 24935258
122. Martin B. Hirota K. Cua D.J. Stockinger B. Veldhoen M. Interleukin-17-producing gammadelta T cells selectively expand in response to pathogen products and environmental signals Immunity 2009 31 321 330 10.1016/j.immuni.2009.06.020 19682928
123. Housseau F. Wu S. Wick E.C. Fan H. Wu X. Llosa N.J. Smith K.N. Tam A. Ganguly S. Wanyiri J.W. Redundant Innate and Adaptive Sources of IL17 Production Drive Colon Tumorigenesis Cancer Res. 2016 76 2115 2124 10.1158/0008-5472.CAN-15-0749 26880802
124. Ramirez K. Witherden D.A. Havran W.L. All hands on DE(T)C: Epithelial-resident gammadelta T cells respond to tissue injury Cell. Immunol. 2015 296 57 61 10.1016/j.cellimm.2015.04.003 25958272
125. Jameson J. Ugarte K. Chen N. Yachi P. Fuchs E. Boismenu R. Havran W.L. A role for skin gammadelta T cells in wound repair Science 2002 296 747 749 10.1126/science.1069639 11976459
126. Ono T. Okamoto K. Nakashima T. Nitta T. Hori S. Iwakura Y. Takayanagi H. IL-17-producing gammadelta T cells enhance bone regeneration Nat. Commun. 2016 7 10928 10.1038/ncomms10928 26965320
127. Li Z. Burns A.R. Miller S.B. Smith C.W. CCL20, gammadelta T cells, and IL-22 in corneal epithelial healing FASEB J. 2011 25 2659 2668 10.1096/fj.11-184804 21518851
128. Zhang W. Magadi S. Li Z. Smith C.W. Burns A.R. IL-20 promotes epithelial healing of the injured mouse cornea Exp. Eye Res. 2017 154 22 29 10.1016/j.exer.2016.11.006 27818315
129. Davis P.A. Corless D.J. Aspinall R. Wastell C. Effect of CD4(+) and CD8(+) cell depletion on wound healing Br. J. Surg. 2001 88 298 304 10.1046/j.1365-2168.2001.01665.x 11167885
130. Reinke S. Geissler S. Taylor W.R. Schmidt-Bleek K. Juelke K. Schwachmeyer V. Dahne M. Hartwig T. Akyuz L. Meisel C. Terminally differentiated CD8(+) T cells negatively affect bone regeneration in humans Sci. Transl. Med. 2013 5 177ra136 10.1126/scitranslmed.3004754
131. Konnecke I. Serra A. El Khassawna T. Schlundt C. Schell H. Hauser A. Ellinghaus A. Volk H.D. Radbruch A. Duda G.N. T and B cells participate in bone repair by infiltrating the fracture callus in a two-wave fashion Bone 2014 64 155 165 10.1016/j.bone.2014.03.052 24721700
132. Klose C.S. Artis D. Innate lymphoid cells as regulators of immunity, inflammation and tissue homeostasis Nat. Immunol. 2016 17 765 774 10.1038/ni.3489 27328006
133. Halim T.Y. Steer C.A. Matha L. Gold M.J. Martinez-Gonzalez I. McNagny K.M. McKenzie A.N. Takei F. Group 2 innate lymphoid cells are critical for the initiation of adaptive T helper 2 cell-mediated allergic lung inflammation Immunity 2014 40 425 435 10.1016/j.immuni.2014.01.011 24613091
134. Mirchandani A.S. Besnard A.G. Yip E. Scott C. Bain C.C. Cerovic V. Salmond R.J. Liew F.Y. Type 2 innate lymphoid cells drive CD4+ Th2 cell responses J. Immunol. 2014 192 2442 2448 10.4049/jimmunol.1300974 24470502
135. Huang Y. Paul W.E. Inflammatory group 2 innate lymphoid cells Int. Immunol. 2016 28 23 28 10.1093/intimm/dxv044 26232596
136. Sadtler K. Estrellas K. Allen B.W. Wolf M.T. Fan H. Tam A.J. Patel C.H. Luber B.S. Wang H. Wagner K.R. Developing a pro-regenerative biomaterial scaffold microenvironment requires T helper 2 cells Science 2016 352 366 370 10.1126/science.aad9272 27081073
137. Anderson J.M. McNally A.K. Biocompatibility of implants: Lymphocyte/macrophage interactions Semin. Immunopathol. 2011 33 221 233 10.1007/s00281-011-0244-1 21271251
138. Matassi F. Nistri L. Chicon Paez D. Innocenti M. New biomaterials for bone regeneration Clin. Cases Miner. Bone Metab. 2011 8 21 24 22461799
139. Gao C. Deng Y. Feng P. Mao Z. Li P. Yang B. Deng J. Cao Y. Shuai C. Peng S. Current progress in bioactive ceramic scaffolds for bone repair and regeneration Int. J. Mol. Sci. 2014 15 4714 4732 10.3390/ijms15034714 24646912
140. Vroman I. Tighzert L. Biodegradable polymers Materials 2009 2 307 344 10.3390/ma2020307
141. Bai X. Gao M. Syed S. Zhuang J. Xu X. Zhang X.Q. Bioactive hydrogels for bone regeneration Bioact. Mater. 2018 3 401 417 10.1016/j.bioactmat.2018.05.006 30003179
142. Jafari M. Paknejad Z. Rad M.R. Motamedian S.R. Eghbal M.J. Nadjmi N. Khojasteh A. Polymeric scaffolds in tissue engineering: A literature review J. Biomed. Mater. Res. B Appl. Biomater. 2017 105 431 459 10.1002/jbm.b.33547 26496456
143. Stratton S. Shelke N.B. Hoshino K. Rudraiah S. Kumbar S.G. Bioactive polymeric scaffolds for tissue engineering Bioact. Mater. 2016 1 93 108 10.1016/j.bioactmat.2016.11.001 28653043
144. Janouskova O. Synthetic polymer scaffolds for soft tissue engineering Physiol. Res. 2018 67 S335 S348 30379554
145. Malikmammadov E. Tanir T.E. Kiziltay A. Hasirci V. Hasirci N. PCL and PCL-based materials in biomedical applications J. Biomater. Sci. Polym. Ed. 2018 29 863 893 10.1080/09205063.2017.1394711 29053081
146. Li W.J. Laurencin C.T. Caterson E.J. Tuan R.S. Ko F.K. Electrospun nanofibrous structure: A novel scaffold for tissue engineering J. Biomed. Mater. Res. 2002 60 613 621 10.1002/jbm.10167 11948520
147. Nair A. Tang L. Influence of scaffold design on host immune and stem cell responses Semin. Immunol. 2017 29 62 71 10.1016/j.smim.2017.03.001 28431919
148. Zarrintaj P. Manouchehri S. Ahmadi Z. Saeb M.R. Urbanska A.M. Kaplan D.L. Mozafari M. Agarose-based biomaterials for tissue engineering Carbohydr. Polym. 2018 187 66 84 10.1016/j.carbpol.2018.01.060 29486846
149. Lee K.Y. Mooney D.J. Alginate: Properties and biomedical applications Prog. Polym. Sci. 2012 37 106 126 10.1016/j.progpolymsci.2011.06.003 22125349
150. Ahsan S.M. Thomas M. Reddy K.K. Sooraparaju S.G. Asthana A. Bhatnagar I. Chitosan as biomaterial in drug delivery and tissue engineering Int. J. Biol. Macromol. 2018 110 97 109 10.1016/j.ijbiomac.2017.08.140 28866015
151. Rnjak-Kovacina J. Tang F. Whitelock J.M. Lord M.S. Glycosaminoglycan and Proteoglycan-Based Biomaterials: Current Trends and Future Perspectives Adv. Healthc. Mater. 2018 7 e1701042 10.1002/adhm.201701042 29210510
152. Collins M.N. Birkinshaw C. Hyaluronic acid based scaffolds for tissue engineering—A review Carbohydr. Polym. 2013 92 1262 1279 10.1016/j.carbpol.2012.10.028 23399155
153. Wlodarczyk-Biegun M.K. Del Campo A. 3D bioprinting of structural proteins Biomaterials 2017 134 180 201 10.1016/j.biomaterials.2017.04.019 28477541
154. Sproul E. Nandi S. Brown A. Fibrin biomaterials for tissue regeneration and repair Peptides and Proteins as Biomaterials for Tissue Regeneration and Repair Barbosa M.A. Martins M.C.L. Woodhead Publishing Sawston, Cambridge, UK 2018 151 173
155. Melke J. Midha S. Ghosh S. Ito K. Hofmann S. Silk fibroin as biomaterial for bone tissue engineering Acta Biomater. 2016 31 1 16 10.1016/j.actbio.2015.09.005 26360593
156. Qi Y. Wang H. Wei K. Yang Y. Zheng R.Y. Kim I.S. Zhang K.Q. A Review of Structure Construction of Silk Fibroin Biomaterials from Single Structures to Multi-Level Structures Int. J. Mol. Sci. 2017 18 237 10.3390/ijms18030237 28273799
157. Park J. Gerber M.H. Babensee J.E. Phenotype and polarization of autologous T cells by biomaterial-treated dendritic cells J. Biomed. Mater. Res. A 2015 103 170 184 10.1002/jbm.a.35150 24616366
158. Morris A.H. Stamer D.K. Kyriakides T.R. The host response to naturally-derived extracellular matrix biomaterials Semin. Immunol. 2017 29 72 91 10.1016/j.smim.2017.01.002 28274693
159. Hotaling N.A. Tang L. Irvine D.J. Babensee J.E. Biomaterial Strategies for Immunomodulation Annu. Rev. Biomed. Eng. 2015 17 317 349 10.1146/annurev-bioeng-071813-104814 26421896
160. Mora-Solano C. Collier J.H. Engaging adaptive immunity with biomaterials J. Mater. Chem. B 2014 2 2409 2421 10.1039/C3TB21549K 24729870
161. Panilaitis B. Altman G.H. Chen J. Jin H.J. Karageorgiou V. Kaplan D.L. Macrophage responses to silk Biomaterials 2003 24 3079 3085 10.1016/S0142-9612(03)00158-3 12895580
162. Reeves A.R. Spiller K.L. Freytes D.O. Vunjak-Novakovic G. Kaplan D.L. Controlled release of cytokines using silk-biomaterials for macrophage polarization Biomaterials 2015 73 272 283 10.1016/j.biomaterials.2015.09.027 26421484
163. Bhattacharjee M. Schultz-Thater E. Trella E. Miot S. Das S. Loparic M. Ray A.R. Martin I. Spagnoli G.C. Ghosh S. The role of 3D structure and protein conformation on the innate and adaptive immune responses to silk-based biomaterials Biomaterials 2013 34 8161 8171 10.1016/j.biomaterials.2013.07.018 23896003
164. Nakamura K. Yokohama S. Yoneda M. Okamoto S. Tamaki Y. Ito T. Okada M. Aso K. Makino I. High, but not low, molecular weight hyaluronan prevents T-cell-mediated liver injury by reducing proinflammatory cytokines in mice J. Gastroenterol. 2004 39 346 354 10.1007/s00535-003-1301-x 15168246
165. Je J.Y. Kim S.K. Reactive oxygen species scavenging activity of aminoderivatized chitosan with different degree of deacetylation Bioorg. Med. Chem. 2006 14 5989 5994 10.1016/j.bmc.2006.05.016 16725329
166. Orenstein S.B. Qiao Y. Klueh U. Kreutzer D.L. Novitsky Y.W. Activation of human mononuclear cells by porcine biologic meshes in vitro Hernia 2010 14 401 407 10.1007/s10029-010-0634-7 20145965
167. Kawai T. Akira S. The role of pattern-recognition receptors in innate immunity: Update on Toll-like receptors Nat. Immunol. 2010 11 373 384 10.1038/ni.1863 20404851
168. Maitra R. Clement C.C. Scharf B. Crisi G.M. Chitta S. Paget D. Purdue P.E. Cobelli N. Santambrogio L. Endosomal damage and TLR2 mediated inflammasome activation by alkane particles in the generation of aseptic osteolysis Mol. Immunol. 2009 47 175 184 10.1016/j.molimm.2009.09.023 19804908
169. Seong S.Y. Matzinger P. Hydrophobicity: An ancient damage-associated molecular pattern that initiates innate immune responses Nat. Rev. Immunol. 2004 4 469 478 10.1038/nri1372 15173835
170. Moyano D.F. Goldsmith M. Solfiell D.J. Landesman-Milo D. Miranda O.R. Peer D. Rotello V.M. Nanoparticle hydrophobicity dictates immune response J. Am. Chem. Soc. 2012 134 3965 3967 10.1021/ja2108905 22339432
171. Song S. Ravensbergen K. Alabanza A. Soldin D. Hahm J.I. Distinct adsorption configurations and self-assembly characteristics of fibrinogen on chemically uniform and alternating surfaces including block copolymer nanodomains ACS Nano 2014 8 5257 5269 10.1021/nn5013397 24708538
172. Kumar N. Parajuli O. Gupta A. Hahm J.I. Elucidation of protein adsorption behavior on polymeric surfaces: Toward high-density, high-payload protein templates Langmuir 2008 24 2688 2694 10.1021/la7022456 18225924
173. Absolom D.R. Zingg W. Neumann A.W. Protein adsorption to polymer particles: Role of surface properties J. Biomed. Mater. Res. 1987 21 161 171 10.1002/jbm.820210202 3818679
174. Ouberai M.M. Xu K. Welland M.E. Effect of the interplay between protein and surface on the properties of adsorbed protein layers Biomaterials 2014 35 6157 6163 10.1016/j.biomaterials.2014.04.012 24780165
175. Kakizawa Y. Lee J.S. Bell B. Fahmy T.M. Precise manipulation of biophysical particle parameters enables control of proinflammatory cytokine production in presence of TLR 3 and 4 ligands Acta Biomater. 2017 57 136 145 10.1016/j.actbio.2017.01.025 28069499
176. Li G. Yang P. Guo X. Huang N. Shen R. An in vitro evaluation of inflammation response of titanium functionalized with heparin/fibronectin complex Cytokine 2011 56 208 217 10.1016/j.cyto.2011.06.020 21795064
177. Alfarsi M.A. Hamlet S.M. Ivanovski S. Titanium surface hydrophilicity modulates the human macrophage inflammatory cytokine response J. Biomed. Mater. Res. A 2014 102 60 67 10.1002/jbm.a.34666 23595995
178. Chang D.T. Colton E. Matsuda T. Anderson J.M. Lymphocyte adhesion and interactions with biomaterial adherent macrophages and foreign body giant cells J. Biomed. Mater. Res. A 2009 91 1210 1220 10.1002/jbm.a.32218 19148923
179. Drury J.L. Mooney D.J. Hydrogels for tissue engineering: Scaffold design variables and applications Biomaterials 2003 24 4337 4351 10.1016/S0142-9612(03)00340-5 12922147
180. Peppas N.A. Kavimandan N.J. Nanoscale analysis of protein and peptide absorption: Insulin absorption using complexation and pH-sensitive hydrogels as delivery vehicles Eur. J. Pharm. Sci. 2006 29 183 197 10.1016/j.ejps.2006.04.014 16777391
181. Tiller J.C. Bonner G. Pan L.C. Klibanov A.M. Improving biomaterial properties of collagen films by chemical modification Biotechnol Bioeng 2001 73 246 252 10.1002/bit.1057 11257607
182. Blaszykowski C. Sheikh S. Thompson M. Biocompatibility and antifouling: Is there really a link? Trends Biotechnol. 2014 32 61 62 10.1016/j.tibtech.2013.11.002 24321373
183. Maitra R. Clement C.C. Crisi G.M. Cobelli N. Santambrogio L. Immunogenecity of modified alkane polymers is mediated through TLR1/2 activation PloS ONE 2008 3 e2438 10.1371/journal.pone.0002438 18560588
184. Andorko J.I. Jewell C.M. Designing biomaterials with immunomodulatory properties for tissue engineering and regenerative medicine Bioeng. Transl. Med. 2017 2 139 155 10.1002/btm2.10063 28932817
185. Barbosa J.N. Barbosa M.A. Aguas A.P. Inflammatory responses and cell adhesion to self-assembled monolayers of alkanethiolates on gold Biomaterials 2004 25 2557 2563 10.1016/j.biomaterials.2003.09.047 14751741
186. Barbosa J.N. Madureira P. Barbosa M.A. Aguas A.P. The influence of functional groups of self-assembled monolayers on fibrous capsule formation and cell recruitment J. Biomed. Mater. Res. A 2006 76 737 743 10.1002/jbm.a.30602 16331651
187. Bartneck M. Keul H.A. Singh S. Czaja K. Bornemann J. Bockstaller M. Moeller M. Zwadlo-Klarwasser G. Groll J. Rapid uptake of gold nanorods by primary human blood phagocytes and immunomodulatory effects of surface chemistry ACS Nano 2010 4 3073 3086 10.1021/nn100262h 20507158
188. Christo S.N. Bachhuka A. Diener K.R. Mierczynska A. Hayball J.D. Vasilev K. The Role of Surface Nanotopography and Chemistry on Primary Neutrophil and Macrophage Cellular Responses Adv. Healthc. Mater. 2016 5 956 965 10.1002/adhm.201500845 26845244
189. Getts D.R. Terry R.L. Getts M.T. Deffrasnes C. Muller M. van Vreden C. Ashhurst T.M. Chami B. McCarthy D. Wu H. Therapeutic inflammatory monocyte modulation using immune-modifying microparticles Sci. Transl. Med. 2014 6 219ra217 10.1126/scitranslmed.3007563 24431111
190. Kamath S. Bhattacharyya D. Padukudru C. Timmons R.B. Tang L. Surface chemistry influences implant-mediated host tissue responses J. Biomed. Mater. Res. A 2008 86 617 626 10.1002/jbm.a.31649 18022841
191. Keselowsky B.G. Collard D.M. Garcia A.J. Surface chemistry modulates fibronectin conformation and directs integrin binding and specificity to control cell adhesion J. Biomed. Mater. Res. A 2003 66 247 259 10.1002/jbm.a.10537 12888994
192. Lan M.A. Gersbach C.A. Michael K.E. Keselowsky B.G. Garcia A.J. Myoblast proliferation and differentiation on fibronectin-coated self assembled monolayers presenting different surface chemistries Biomaterials 2005 26 4523 4531 10.1016/j.biomaterials.2004.11.028 15722121
193. Lee M.H. Ducheyne P. Lynch L. Boettiger D. Composto R.J. Effect of biomaterial surface properties on fibronectin-alpha5beta1 integrin interaction and cellular attachment Biomaterials 2006 27 1907 1916 10.1016/j.biomaterials.2005.11.003 16310247
194. Sperling C. Schweiss R.B. Streller U. Werner C. In vitro hemocompatibility of self-assembled monolayers displaying various functional groups Biomaterials 2005 26 6547 6557 10.1016/j.biomaterials.2005.04.042 15939466
195. Tang L. Wu Y. Timmons R.B. Fibrinogen adsorption and host tissue responses to plasma functionalized surfaces J. Biomed. Mater. Res. 1998 42 156 163 10.1002/(SICI)1097-4636(199810)42:1 3.0.CO;2-J 9740018
196. Nair A. Zou L. Bhattacharyya D. Timmons R.B. Tang L. Species and density of implant surface chemistry affect the extent of foreign body reactions Langmuir 2008 24 2015 2024 10.1021/la7025973 18189430
197. Sastry S.K. Burridge K. Focal adhesions: A nexus for intracellular signaling and cytoskeletal dynamics Exp. Cell Res. 2000 261 25 36 10.1006/excr.2000.5043 11082272
198. Kalltorp M. Oblogina S. Jacobsson S. Karlsson A. Tengvall P. Thomsen P. In vivo cell recruitment, cytokine release and chemiluminescence response at gold, and thiol functionalized surfaces Biomaterials 1999 20 2123 2137 10.1016/S0142-9612(99)00115-5 10555080
199. Benesch J. Svedhem S. Svensson S.C. Valiokas R. Liedberg B. Tengvall P. Protein adsorption to oligo(ethylene glycol) self-assembled monolayers: Experiments with fibrinogen, heparinized plasma, and serum J. Biomater. Sci. Polym. Ed. 2001 12 581 597 10.1163/156856201316883421 11556738
200. Hirata I. Hioki Y. Toda M. Kitazawa T. Murakami Y. Kitano E. Kitamura H. Ikada Y. Iwata H. Deposition of complement protein C3b on mixed self-assembled monolayers carrying surface hydroxyl and methyl groups studied by surface plasmon resonance J. Biomed. Mater. Res. A 2003 66 669 676 10.1002/jbm.a.10067 12918051
201. Kim J. Somorjai G.A. Molecular packing of lysozyme, fibrinogen, and bovine serum albumin on hydrophilic and hydrophobic surfaces studied by infrared-visible sum frequency generation and fluorescence microscopy J. Am. Chem. Soc. 2003 125 3150 3158 10.1021/ja028987n 12617683
202. Szott L.M. Horbett T.A. Protein interactions with surfaces: Cellular responses, complement activation, and newer methods Curr. Opin. Chem. Biol. 2011 15 677 682 10.1016/j.cbpa.2011.04.021 21665522
203. Sivaraman B. Fears K.P. Latour R.A. Investigation of the effects of surface chemistry and solution concentration on the conformation of adsorbed proteins using an improved circular dichroism method Langmuir 2009 25 3050 3056 10.1021/la8036814 19437712
204. Vieira E.P. Rocha S. Carmo Pereira M. Mohwald H. Coelho M.A. Adsorption and diffusion of plasma proteins on hydrophilic and hydrophobic surfaces: Effect of trifluoroethanol on protein structure Langmuir 2009 25 9879 9886 10.1021/la9009948 19705886
205. Neumann S. Burkert K. Kemp R. Rades T. Rod Dunbar P. Hook S. Activation of the NLRP3 inflammasome is not a feature of all particulate vaccine adjuvants Immunol. Cell Biol. 2014 92 535 542 10.1038/icb.2014.21 24687021
206. Wen Y. Waltman A. Han H. Collier J.H. Switching the Immunogenicity of Peptide Assemblies Using Surface Properties ACS Nano 2016 10.1021/acsnano.6b03409
207. Gallorini S. Berti F. Parente P. Baronio R. Aprea S. D’Oro U. Pizza M. Telford J.L. Wack A. Introduction of zwitterionic motifs into bacterial polysaccharides generates TLR2 agonists able to activate APCs J. Immunol. 2007 179 8208 8215 10.4049/jimmunol.179.12.8208 18056364
208. Betancourt T. Brannon-Peppas L. Micro- and nanofabrication methods in nanotechnological medical and pharmaceutical devices Int. J. Nanomed. 2006 1 483 495 10.2147/nano.2006.1.4.483
209. Refai A.K. Textor M. Brunette D.M. Waterfield J.D. Effect of titanium surface topography on macrophage activation and secretion of proinflammatory cytokines and chemokines J. Biomed. Mater. Res. A 2004 70 194 205 10.1002/jbm.a.30075 15227664
210. Soskolne W.A. Cohen S. Sennerby L. Wennerberg A. Shapira L. The effect of titanium surface roughness on the adhesion of monocytes and their secretion of TNF-alpha and PGE2 Clin. Oral Implants Res. 2002 13 86 93 10.1034/j.1600-0501.2002.130111.x 12005150
211. Bota P.C. Collie A.M. Puolakkainen P. Vernon R.B. Sage E.H. Ratner B.D. Stayton P.S. Biomaterial topography alters healing in vivo and monocyte/macrophage activation in vitro J. Biomed. Mater. Res. A 2010 95 649 657 10.1002/jbm.a.32893 20725970
212. Collie A.M. Bota P.C. Johns R.E. Maier R.V. Stayton P.S. Differential monocyte/macrophage interleukin-1beta production due to biomaterial topography requires the beta2 integrin signaling pathway J. Biomed. Mater. Res. A 2011 96 162 169 10.1002/jbm.a.32963 21105164
213. Mohiuddin M. Pan H.A. Hung Y.C. Huang G.S. Control of growth and inflammatory response of macrophages and foam cells with nanotopography Nanoscale Res. Lett. 2012 7 394 10.1186/1556-276X-7-394 22799434
214. Fink J. Fuhrmann R. Scharnweber T. Franke R.P. Stimulation of monocytes and macrophages: Possible influence of surface roughness Clin. Hemorheol. Microcirc. 2008 39 205 212 18503127
215. Gamboa J.R. Mohandes S. Tran P.L. Slepian M.J. Yoon J.Y. Linear fibroblast alignment on sinusoidal wave micropatterns Colloids Surf. B Biointerfaces 2013 104 318 325 10.1016/j.colsurfb.2012.11.035 23375052
216. Hulander M. Lundgren A. Faxalv L. Lindahl T.L. Palmquist A. Berglin M. Elwing H. Gradients in surface nanotopography used to study platelet adhesion and activation Colloids Surf. B Biointerfaces 2013 110 261 269 10.1016/j.colsurfb.2013.04.010 23732803
217. Gilchrist C.L. Ruch D.S. Little D. Guilak F. Micro-scale and meso-scale architectural cues cooperate and compete to direct aligned tissue formation Biomaterials 2014 35 10015 10024 10.1016/j.biomaterials.2014.08.047 25263687
218. Lopacinska J.M. Gradinaru C. Wierzbicki R. Kobler C. Schmidt M.S. Madsen M.T. Skolimowski M. Dufva M. Flyvbjerg H. Molhave K. Cell motility, morphology, viability and proliferation in response to nanotopography on silicon black Nanoscale 2012 4 3739 3745 10.1039/c2nr11455k 22614757
219. Vogel V. Sheetz M. Local force and geometry sensing regulate cell functions Nat. Rev. Mol. Cell Biol. 2006 7 265 275 10.1038/nrm1890 16607289
220. Yim E.K. Reano R.M. Pang S.W. Yee A.F. Chen C.S. Leong K.W. Nanopattern-induced changes in morphology and motility of smooth muscle cells Biomaterials 2005 26 5405 5413 10.1016/j.biomaterials.2005.01.058 15814139
221. Gittens R.A. McLachlan T. Olivares-Navarrete R. Cai Y. Berner S. Tannenbaum R. Schwartz Z. Sandhage K.H. Boyan B.D. The effects of combined micron-/submicron-scale surface roughness and nanoscale features on cell proliferation and differentiation Biomaterials 2011 32 3395 3403 10.1016/j.biomaterials.2011.01.029 21310480
222. Ma Q.L. Zhao L.Z. Liu R.R. Jin B.Q. Song W. Wang Y. Zhang Y.S. Chen L.H. Zhang Y.M. Improved implant osseointegration of a nanostructured titanium surface via mediation of macrophage polarization Biomaterials 2014 35 9853 9867 10.1016/j.biomaterials.2014.08.025 25201737
223. Tan K.S. Qian L. Rosado R. Flood P.M. Cooper L.F. The role of titanium surface topography on J774A.1 macrophage inflammatory cytokines and nitric oxide production Biomaterials 2006 27 5170 5177 10.1016/j.biomaterials.2006.05.002 16808973
224. Hulander M. Lundgren A. Berglin M. Ohrlander M. Lausmaa J. Elwing H. Immune complement activation is attenuated by surface nanotopography Int. J. Nanomed. 2011 6 2653 2666 10.2147/IJN.S24578
225. Roach P. Eglin D. Rohde K. Perry C.C. Modern biomaterials: A review—bulk properties and implications of surface modifications J. Mater. Sci. Mater. Med. 2007 18 1263 1277 10.1007/s10856-006-0064-3 17443395
226. Scopelliti P.E. Borgonovo A. Indrieri M. Giorgetti L. Bongiorno G. Carbone R. Podesta A. Milani P. The effect of surface nanometre-scale morphology on protein adsorption PLoS ONE 2010 5 e11862 10.1371/journal.pone.0011862 20686681
227. Rechendorff K. Hovgaard M.B. Foss M. Zhdanov V.P. Besenbacher F. Enhancement of protein adsorption induced by surface roughness Langmuir 2006 22 10885 10888 10.1021/la0621923 17154557
228. Hovgaard M.B. Rechendorff K. Chevallier J. Foss M. Besenbacher F. Fibronectin adsorption on tantalum: The influence of nanoroughness J. Phys. Chem. B 2008 112 8241 8249 10.1021/jp801103n 18564871
230. Mrksich M. Whitesides G.M. Using self-assembled monolayers to understand the interactions of man-made surfaces with proteins and cells Annu. Rev. Biophys. Biomol. Struct. 1996 25 55 78 10.1146/annurev.bb.25.060196.000415 8800464
231. Da Silva C.A. Chalouni C. Williams A. Hartl D. Lee C.G. Elias J.A. Chitin is a size-dependent regulator of macrophage TNF and IL-10 production J. Immunol. 2009 182 3573 3582 10.4049/jimmunol.0802113 19265136
232. Fernandez T.D. Pearson J.R. Leal M.P. Torres M.J. Blanca M. Mayorga C. Le Guevel X. Intracellular accumulation and immunological properties of fluorescent gold nanoclusters in human dendritic cells Biomaterials 2015 43 1 12 10.1016/j.biomaterials.2014.11.045 25591956
233. Lee S. Choi J. Shin S. Im Y.M. Song J. Kang S.S. Nam T.H. Webster T.J. Kim S.H. Khang D. Analysis on migration and activation of live macrophages on transparent flat and nanostructured titanium Acta Biomater. 2011 7 2337 2344 10.1016/j.actbio.2011.01.006 21232636
234. Lu J. Webster T.J. Reduced immune cell responses on nano and submicron rough titanium Acta Biomater. 2015 16 223 231 10.1016/j.actbio.2015.01.036 25660564
235. McWhorter F.Y. Wang T. Nguyen P. Chung T. Liu W.F. Modulation of macrophage phenotype by cell shape Proc. Natl. Acad. Sci. USA 2013 110 17253 17258 10.1073/pnas.1308887110 24101477
236. Veiseh O. Doloff J.C. Ma M. Vegas A.J. Tam H.H. Bader A.R. Li J. Langan E. Wyckoff J. Loo W.S. Size- and shape-dependent foreign body immune response to materials implanted in rodents and non-human primates Nat. Mater. 2015 14 643 651 10.1038/nmat4290 25985456
237. Baker D.W. Liu X. Weng H. Luo C. Tang L. Fibroblast/fibrocyte: Surface interaction dictates tissue reactions to micropillar implants Biomacromolecules 2011 12 997 1005 10.1021/bm1013487 21332193
238. Jahed Z. Molladavoodi S. Seo B.B. Gorbet M. Tsui T.Y. Mofrad M.R. Cell responses to metallic nanostructure arrays with complex geometries Biomaterials 2014 35 9363 9371 10.1016/j.biomaterials.2014.07.022 25123921
239. Hornung V. Bauernfeind F. Halle A. Samstad E.O. Kono H. Rock K.L. Fitzgerald K.A. Latz E. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization Nat. Immunol. 2008 9 847 856 10.1038/ni.1631 18604214
240. Niikura K. Matsunaga T. Suzuki T. Kobayashi S. Yamaguchi H. Orba Y. Kawaguchi A. Hasegawa H. Kajino K. Ninomiya T. Gold nanoparticles as a vaccine platform: Influence of size and shape on immunological responses in vitro and in vivo ACS Nano 2013 7 3926 3938 10.1021/nn3057005 23631767
241. Padmore T. Stark C. Turkevich L.A. Champion J.A. Quantitative analysis of the role of fiber length on phagocytosis and inflammatory response by alveolar macrophages Biochim. Biophys. Acta Gen Subj 2017 1861 58 67 10.1016/j.bbagen.2016.09.031 27784615
242. Schanen B.C. Karakoti A.S. Seal S. Drake D.R. 3rd Warren W.L. Self W.T. Exposure to titanium dioxide nanomaterials provokes inflammation of an in vitro human immune construct ACS Nano 2009 3 2523 2532 10.1021/nn900403h 19769402
243. Sunshine J.C. Perica K. Schneck J.P. Green J.J. Particle shape dependence of CD8+ T cell activation by artificial antigen presenting cells Biomaterials 2014 35 269 277 10.1016/j.biomaterials.2013.09.050 24099710
244. Vaine C.A. Patel M.K. Zhu J. Lee E. Finberg R.W. Hayward R.C. Kurt-Jones E.A. Tuning innate immune activation by surface texturing of polymer microparticles: The role of shape in inflammasome activation J. Immunol. 2013 190 3525 3532 10.4049/jimmunol.1200492 23427254
245. Kosmides A.K. Meyer R.A. Hickey J.W. Aje K. Cheung K.N. Green J.J. Schneck J.P. Biomimetic biodegradable artificial antigen presenting cells synergize with PD-1 blockade to treat melanoma Biomaterials 2017 118 16 26 10.1016/j.biomaterials.2016.11.038 27940380
246. Meyer R.A. Sunshine J.C. Perica K. Kosmides A.K. Aje K. Schneck J.P. Green J.J. Biodegradable nanoellipsoidal artificial antigen presenting cells for antigen specific T-cell activation Small 2015 11 1519 1525 10.1002/smll.201402369 25641795
247. Neacsu P. Mazare A. Cimpean A. Park J. Costache M. Schmuki P. Demetrescu I. Reduced inflammatory activity of RAW 264.7 macrophages on titania nanotube modified Ti surface Int. J. Biochem. Cell Biol. 2014 55 187 195 10.1016/j.biocel.2014.09.006 25220343
248. Rajyalakshmi A. Ercan B. Balasubramanian K. Webster T.J. Reduced adhesion of macrophages on anodized titanium with select nanotube surface features Int. J. Nanomed. 2011 6 1765 1771
249. Moura C.C. Zanetta-Barbosa D. Dechichi P. Carvalho V.F. Soares P.B. Effects of titanium surfaces on the developmental profile of monocytes/macrophages Braz. Dent. J. 2014 25 96 103 10.1590/0103-6440201302260 25140712
250. Mahyudin F. Widhiyanto L. Hermawan H. Biomaterials in orthopaedics Advanced Structured Materials Springer Publishing New York, NY, USA 2016
251. Loh Q.L. Choong C. Three-dimensional scaffolds for tissue engineering applications: Role of porosity and pore size Tissue Eng. Part B Rev. 2013 19 485 502 10.1089/ten.teb.2012.0437 23672709
252. Matena J. Petersen S. Gieseke M. Kampmann A. Teske M. Beyerbach M. Murua Escobar H. Haferkamp H. Gellrich N.C. Nolte I. SLM produced porous titanium implant improvements for enhanced vascularization and osteoblast seeding Int. J. Mol. Sci. 2015 16 7478 7492 10.3390/ijms16047478 25849656
253. Kuboki Y. Jin Q. Takita H. Geometry of carriers controlling phenotypic expression in BMP-induced osteogenesis and chondrogenesis J. Bone Joint Surg. Am. 2001 83 Suppl. 1 S105 S115 10.2106/00004623-200100002-00005 11314788
254. Artel A. Mehdizadeh H. Chiu Y.C. Brey E.M. Cinar A. An agent-based model for the investigation of neovascularization within porous scaffolds Tissue Eng. Part A 2011 17 2133 2141 10.1089/ten.tea.2010.0571 21513462
255. Jiang J. Li Z. Wang H. Wang Y. Carlson M.A. Teusink M.J. MacEwan M.R. Gu L. Xie J. Expanded 3D Nanofiber Scaffolds: Cell Penetration, Neovascularization, and Host Response Adv. Healthc. Mater. 2016 5 2993 3003 10.1002/adhm.201600808 27709840
256. Muller D. Chim H. Bader A. Whiteman M. Schantz J.T. Vascular guidance: Microstructural scaffold patterning for inductive neovascularization Stem Cells Int. 2010 2011 547247 10.4061/2011/547247 21188080
257. Kim H.J. Kim U.J. Vunjak-Novakovic G. Min B.H. Kaplan D.L. Influence of macroporous protein scaffolds on bone tissue engineering from bone marrow stem cells Biomaterials 2005 26 4442 4452 10.1016/j.biomaterials.2004.11.013 15701373
258. Madden L.R. Mortisen D.J. Sussman E.M. Dupras S.K. Fugate J.A. Cuy J.L. Hauch K.D. Laflamme M.A. Murry C.E. Ratner B.D. Proangiogenic scaffolds as functional templates for cardiac tissue engineering Proc. Natl. Acad. Sci. USA 2010 107 15211 15216 10.1073/pnas.1006442107 20696917
259. Bridges A.W. Singh N. Burns K.L. Babensee J.E. Andrew Lyon L. Garcia A.J. Reduced acute inflammatory responses to microgel conformal coatings Biomaterials 2008 29 4605 4615 10.1016/j.biomaterials.2008.08.015 18804859
260. Eming S.A. Hammerschmidt M. Krieg T. Roers A. Interrelation of immunity and tissue repair or regeneration Semin. Cell Dev. Biol. 2009 20 517 527 10.1016/j.semcdb.2009.04.009 19393325
261. Badylak S.F. TISSUE REGENERATION. A scaffold immune microenvironment Science 2016 352 298 10.1126/science.aaf7587 27081059
262. Mescher A.L. Neff A.W. King M.W. Inflammation and immunity in organ regeneration Dev. Comp. Immunol. 2017 66 98 110 10.1016/j.dci.2016.02.015 26891614
263. Julier Z. Park A.J. Briquez P.S. Martino M.M. Promoting tissue regeneration by modulating the immune system Acta Biomater. 2017 53 13 28 10.1016/j.actbio.2017.01.056 28119112
264. Gower R.M. Boehler R.M. Azarin S.M. Ricci C.F. Leonard J.N. Shea L.D. Modulation of leukocyte infiltration and phenotype in microporous tissue engineering scaffolds via vector induced IL-10 expression Biomaterials 2014 35 2024 2031 10.1016/j.biomaterials.2013.11.036 24309498
265. Hubbell J.A. Thomas S.N. Swartz M.A. Materials engineering for immunomodulation Nature 2009 462 449 460 10.1038/nature08604 19940915
266. Boontheekul T. Mooney D.J. Protein-based signaling systems in tissue engineering Curr. Opin. Biotechnol. 2003 14 559 565 10.1016/j.copbio.2003.08.004 14580589
267. Wolf M.T. Dearth C.L. Sonnenberg S.B. Loboa E.G. Badylak S.F. Naturally derived and synthetic scaffolds for skeletal muscle reconstruction Adv. Drug Deliv. Rev. 2015 84 208 221 10.1016/j.addr.2014.08.011 25174309
268. Badylak S.F. Dziki J.L. Sicari B.M. Ambrosio F. Boninger M.L. Mechanisms by which acellular biologic scaffolds promote functional skeletal muscle restoration Biomaterials 2016 103 128 136 10.1016/j.biomaterials.2016.06.047 27376561
269. Badylak S.F. Freytes D.O. Gilbert T.W. Extracellular matrix as a biological scaffold material: Structure and function Acta Biomater. 2009 5 1 13 10.1016/j.actbio.2008.09.013 18938117
270. Turner N.J. Badylak S.F. The Use of Biologic Scaffolds in the Treatment of Chronic Nonhealing Wounds Adv. Wound Care 2015 4 490 500 10.1089/wound.2014.0604 26244105
271. Brown B.N. Ratner B.D. Goodman S.B. Amar S. Badylak S.F. Macrophage polarization: An opportunity for improved outcomes in biomaterials and regenerative medicine Biomaterials 2012 33 3792 3802 10.1016/j.biomaterials.2012.02.034 22386919
272. Gowen B.B. Borg T.K. Ghaffar A. Mayer E.P. Selective adhesion of macrophages to denatured forms of type I collagen is mediated by scavenger receptors Matrix Biol. 2000 19 61 71 10.1016/S0945-053X(99)00052-9 10686426
273. Veres S.P. Brennan-Pierce E.P. Lee J.M. Macrophage-like U937 cells recognize collagen fibrils with strain-induced discrete plasticity damage J. Biomed. Mater. Res. A 2015 103 397 408 10.1002/jbm.a.35156 24616426
274. Huleihel L. Hussey G.S. Naranjo J.D. Zhang L. Dziki J.L. Turner N.J. Stolz D.B. Badylak S.F. Matrix-bound nanovesicles within ECM bioscaffolds Sci. Adv. 2016 2 e1600502 10.1126/sciadv.1600502 27386584
275. Dziki J.L. Wang D.S. Pineda C. Sicari B.M. Rausch T. Badylak S.F. Solubilized extracellular matrix bioscaffolds derived from diverse source tissues differentially influence macrophage phenotype J. Biomed. Mater. Res. A 2017 105 138 147 10.1002/jbm.a.35894 27601305
276. Keane T.J. Dziki J. Sobieski E. Smoulder A. Castleton A. Turner N. White L.J. Badylak S.F. Restoring Mucosal Barrier Function and Modifying Macrophage Phenotype with an Extracellular Matrix Hydrogel: Potential Therapy for Ulcerative Colitis J. Crohns Colitis 2017 11 360 368 10.1093/ecco-jcc/jjw149 27543807
277. Brown B.N. Valentin J.E. Stewart-Akers A.M. McCabe G.P. Badylak S.F. Macrophage phenotype and remodeling outcomes in response to biologic scaffolds with and without a cellular component Biomaterials 2009 30 1482 1491 10.1016/j.biomaterials.2008.11.040 19121538
278. Fishman J.M. Lowdell M.W. Urbani L. Ansari T. Burns A.J. Turmaine M. North J. Sibbons P. Seifalian A.M. Wood K.J. Immunomodulatory effect of a decellularized skeletal muscle scaffold in a discordant xenotransplantation model Proc. Natl. Acad. Sci. USA 2013 110 14360 14365 10.1073/pnas.1213228110 23940349
279. Keane T.J. Dziki J. Castelton A. Faulk D.M. Messerschmidt V. Londono R. Reing J.E. Velankar S.S. Badylak S.F. Preparation and characterization of a biologic scaffold and hydrogel derived from colonic mucosa J. Biomed. Mater. Res. B Appl. Biomater. 2017 105 291 306 10.1002/jbm.b.33556 26506408
280. Spiller K.L. Nassiri S. Witherel C.E. Anfang R.R. Ng J. Nakazawa K.R. Yu T. Vunjak-Novakovic G. Sequential delivery of immunomodulatory cytokines to facilitate the M1-to-M2 transition of macrophages and enhance vascularization of bone scaffolds Biomaterials 2015 37 194 207 10.1016/j.biomaterials.2014.10.017 25453950
281. Deeken C.R. Eliason B.J. Pichert M.D. Grant S.A. Frisella M.M. Matthews B.D. Differentiation of biologic scaffold materials through physicomechanical, thermal, and enzymatic degradation techniques Ann. Surg. 2012 255 595 604 10.1097/SLA.0b013e3182445341 22314328
282. Delgado L.M. Bayon Y. Pandit A. Zeugolis D.I. To cross-link or not to cross-link? Cross-linking associated foreign body response of collagen-based devices Tissue Eng. Part B Rev. 2015 21 298 313 10.1089/ten.teb.2014.0290 25517923
283. McDade J.K. Brennan-Pierce E.P. Ariganello M.B. Labow R.S. Michael Lee J. Interactions of U937 macrophage-like cells with decellularized pericardial matrix materials: Influence of crosslinking treatment Acta Biomater. 2013 9 7191 7199 10.1016/j.actbio.2013.02.021 23454057
284. Keane T.J. Londono R. Turner N.J. Badylak S.F. Consequences of ineffective decellularization of biologic scaffolds on the host response Biomaterials 2012 33 1771 1781 10.1016/j.biomaterials.2011.10.054 22137126
285. Beachley V.Z. Wolf M.T. Sadtler K. Manda S.S. Jacobs H. Blatchley M.R. Bader J.S. Pandey A. Pardoll D. Elisseeff J.H. Tissue matrix arrays for high-throughput screening and systems analysis of cell function Nat. Methods 2015 12 1197 1204 10.1038/nmeth.3619 26480475
286. Asea A. Rehli M. Kabingu E. Boch J.A. Bare O. Auron P.E. Stevenson M.A. Calderwood S.K. Novel signal transduction pathway utilized by extracellular HSP70: Role of toll-like receptor (TLR) 2 and TLR4 J. Biol. Chem. 2002 277 15028 15034 10.1074/jbc.M200497200 11836257
287. Kovalchin J.T. Wang R. Wagh M.S. Azoulay J. Sanders M. Chandawarkar R.Y. In vivo delivery of heat shock protein 70 accelerates wound healing by up-regulating macrophage-mediated phagocytosis Wound Repair Regen. 2006 14 129 137 10.1111/j.1743-6109.2006.00102.x 16630101
288. Laschke M.W. Augustin V. Kleer S. Tschernig T. Menger M.D. Locally applied macrophage-activating lipopeptide-2 (MALP-2) promotes early vascularization of implanted porous polyethylene (Medpor(R)) Acta Biomater. 2014 10 4661 4669 10.1016/j.actbio.2014.07.004 25062995
289. Yamamoto M. Sato T. Beren J. Verthelyi D. Klinman D.M. The acceleration of wound healing in primates by the local administration of immunostimulatory CpG oligonucleotides Biomaterials 2011 32 4238 4242 10.1016/j.biomaterials.2011.02.043 21421264
290. Noguchi K. Ishikawa I. The roles of cyclooxygenase-2 and prostaglandin E2 in periodontal disease Periodontology 2000 2007 43 85 101 10.1111/j.1600-0757.2006.00170.x 17214837
291. Kalinski P. Hilkens C.M. Snijders A. Snijdewint F.G. Kapsenberg M.L. IL-12-deficient dendritic cells, generated in the presence of prostaglandin E2, promote type 2 cytokine production in maturing human naive T helper cells J. Immunol. 1997 159 28 35 9200435
292. Katamura K. Shintaku N. Yamauchi Y. Fukui T. Ohshima Y. Mayumi M. Furusho K. Prostaglandin E2 at priming of naive CD4+ T cells inhibits acquisition of ability to produce IFN-gamma and IL-2, but not IL-4 and IL-5 J. Immunol. 1995 155 4604 4612 7594459
293. Paralkar V.M. Borovecki F. Ke H.Z. Cameron K.O. Lefker B. Grasser W.A. Owen T.A. Li M. DaSilva-Jardine P. Zhou M. An EP2 receptor-selective prostaglandin E2 agonist induces bone healing Proc. Natl. Acad. Sci. USA 2003 100 6736 6740 10.1073/pnas.1037343100 12748385
294. Kamolratanakul P. Hayata T. Ezura Y. Kawamata A. Hayashi C. Yamamoto Y. Hemmi H. Nagao M. Hanyu R. Notomi T. Nanogel-based scaffold delivery of prostaglandin E(2) receptor-specific agonist in combination with a low dose of growth factor heals critical-size bone defects in mice Arthritis Rheumatol. 2011 63 1021 1033 10.1002/art.30151 21190246
295. Kato N. Hasegawa U. Morimoto N. Saita Y. Nakashima K. Ezura Y. Kurosawa H. Akiyoshi K. Noda M. Nanogel-based delivery system enhances PGE2 effects on bone formation J. Cell Biochem. 2007 101 1063 1070 10.1002/jcb.21160 17520665
296. Toyoda H. Terai H. Sasaoka R. Oda K. Takaoka K. Augmentation of bone morphogenetic protein-induced bone mass by local delivery of a prostaglandin E EP4 receptor agonist Bone 2005 37 555 562 10.1016/j.bone.2005.04.042 16027058
297. Kucia M. Jankowski K. Reca R. Wysoczynski M. Bandura L. Allendorf D.J. Zhang J. Ratajczak J. Ratajczak M.Z. CXCR4-SDF-1 signalling, locomotion, chemotaxis and adhesion J. Mol. Histol. 2004 35 233 245 10.1023/B:HIJO.0000032355.66152.b8 15339043
298. Lau T.T. Wang D.A. Stromal cell-derived factor-1 (SDF-1): Homing factor for engineered regenerative medicine Expert Opin. Biol. Ther. 2011 11 189 197 10.1517/14712598.2011.546338 21219236
299. Son B.R. Marquez-Curtis L.A. Kucia M. Wysoczynski M. Turner A.R. Ratajczak J. Ratajczak M.Z. Janowska-Wieczorek A. Migration of bone marrow and cord blood mesenchymal stem cells in vitro is regulated by stromal-derived factor-1-CXCR4 and hepatocyte growth factor-c-met axes and involves matrix metalloproteinases Stem Cells 2006 24 1254 1264 10.1634/stemcells.2005-0271 16410389
300. Kim Y.H. Tabata Y. Recruitment of mesenchymal stem cells and macrophages by dual release of stromal cell-derived factor-1 and a macrophage recruitment agent enhances wound closure J. Biomed. Mater. Res. A 2016 104 942 956 10.1002/jbm.a.35635 26704185
301. Shen W. Chen X. Chen J. Yin Z. Heng B.C. Chen W. Ouyang H.W. The effect of incorporation of exogenous stromal cell-derived factor-1 alpha within a knitted silk-collagen sponge scaffold on tendon regeneration Biomaterials 2010 31 7239 7249 10.1016/j.biomaterials.2010.05.040 20615544
302. Projahn D. Simsekyilmaz S. Singh S. Kanzler I. Kramp B.K. Langer M. Burlacu A. Bernhagen J. Klee D. Zernecke A. Controlled intramyocardial release of engineered chemokines by biodegradable hydrogels as a treatment approach of myocardial infarction J. Cell. Mol. Med. 2014 18 790 800 10.1111/jcmm.12225 24512349
303. Kimura Y. Tabata Y. Controlled release of stromal-cell-derived factor-1 from gelatin hydrogels enhances angiogenesis J. Biomater. Sci. Polym. Ed. 2010 21 37 51 10.1163/156856209X410193 20040152
304. Boehler R.M. Graham J.G. Shea L.D. Tissue engineering tools for modulation of the immune response Biotechniques 2011 51 239 240 239–240, 242, 244 passim 10.2144/000113754 21988690
305. Hume P.S. He J. Haskins K. Anseth K.S. Strategies to reduce dendritic cell activation through functional biomaterial design Biomaterials 2012 33 3615 3625 10.1016/j.biomaterials.2012.02.009 22361099
306. Chen J. Li M. Yang C. Yin X. Duan K. Wang J. Feng B. Macrophage phenotype switch by sequential action of immunomodulatory cytokines from hydrogel layers on titania nanotubes Colloids Surf. B Biointerfaces 2018 163 336 345 10.1016/j.colsurfb.2018.01.007 29331905
307. Lin C.C. Boyer P.D. Aimetti A.A. Anseth K.S. Regulating MCP-1 diffusion in affinity hydrogels for enhancing immuno-isolation J. Control. Release 2010 142 384 391 10.1016/j.jconrel.2009.11.022 19951731
308. Lin C.C. Metters A.T. Anseth K.S. Functional PEG-peptide hydrogels to modulate local inflammation induced by the pro-inflammatory cytokine TNFalpha Biomaterials 2009 30 4907 4914 10.1016/j.biomaterials.2009.05.083 19560813
309. Wynn T.A. Vannella K.M. Macrophages in Tissue Repair, Regeneration, and Fibrosis Immunity 2016 44 450 462 10.1016/j.immuni.2016.02.015 26982353
310. Boehler R.M. Kuo R. Shin S. Goodman A.G. Pilecki M.A. Gower R.M. Leonard J.N. Shea L.D. Lentivirus delivery of IL-10 to promote and sustain macrophage polarization towards an anti-inflammatory phenotype Biotechnol. Bioeng. 2014 111 1210 1221 10.1002/bit.25175 24375008
311. Gonzalez R. Glaser J. Liu M.T. Lane T.E. Keirstead H.S. Reducing inflammation decreases secondary degeneration and functional deficit after spinal cord injury Exp. Neurol. 2003 184 456 463 10.1016/S0014-4886(03)00257-7 14637115
312. Mokarram N. Merchant A. Mukhatyar V. Patel G. Bellamkonda R.V. Effect of modulating macrophage phenotype on peripheral nerve repair Biomaterials 2012 33 8793 8801 10.1016/j.biomaterials.2012.08.050 22979988
313. Hachim D. LoPresti S.T. Yates C.C. Brown B.N. Shifts in macrophage phenotype at the biomaterial interface via IL-4 eluting coatings are associated with improved implant integration Biomaterials 2017 112 95 107 10.1016/j.biomaterials.2016.10.019 27760399
314. Mori R. Shaw T.J. Martin P. Molecular mechanisms linking wound inflammation and fibrosis: Knockdown of osteopontin leads to rapid repair and reduced scarring J. Exp. Med. 2008 205 43 51 10.1084/jem.20071412 18180311
315. Holladay C. Power K. Sefton M. O’Brien T. Gallagher W.M. Pandit A. Functionalized scaffold-mediated interleukin 10 gene delivery significantly improves survival rates of stem cells in vivo Mol. Ther. 2011 19 969 978 10.1038/mt.2010.311 21266957
316. Rao A.J. Nich C. Dhulipala L.S. Gibon E. Valladares R. Zwingenberger S. Smith R.L. Goodman S.B. Local effect of IL-4 delivery on polyethylene particle induced osteolysis in the murine calvarium J. Biomed. Mater. Res. A 2013 101 1926 1934 10.1002/jbm.a.34486 23225668
317. De Oliveira C.P. Magolbo N. De Aquino R. Weller C. Oral aspirin for treating venous leg ulcers Cochrane Database Syst Rev. 2016 2 CD009432 10.1002/14651858.CD009432.pub2 26889740
318. Canton I. McKean R. Charnley M. Blackwood K.A. Fiorica C. Ryan A.J. MacNeil S. Development of an Ibuprofen-releasing biodegradable PLA/PGA electrospun scaffold for tissue regeneration Biotechnol. Bioeng. 2010 105 396 408 10.1002/bit.22530 19731254
319. Varatharajan L. Thapar A. Lane T. Munster A.B. Davies A.H. Pharmacological adjuncts for chronic venous ulcer healing: A systematic review Phlebology 2016 31 356 365 10.1177/0268355515587194 26036247
320. Friedrich E.E. Sun L.T. Natesan S. Zamora D.O. Christy R.J. Washburn N.R. Effects of hyaluronic acid conjugation on anti-TNF-alpha inhibition of inflammation in burns J. Biomed. Mater. Res. A 2014 102 1527 1536 10.1002/jbm.a.34829 23765644
321. Kim Y.S. Park H.J. Hong M.H. Kang P.M. Morgan J.P. Jeong M.H. Cho J.G. Park J.C. Ahn Y. TNF-alpha enhances engraftment of mesenchymal stem cells into infarcted myocardium Front. Biosci. 2009 14 2845 2856 10.2741/3417
322. Bocker W. Docheva D. Prall W.C. Egea V. Pappou E. Rossmann O. Popov C. Mutschler W. Ries C. Schieker M. IKK-2 is required for TNF-alpha-induced invasion and proliferation of human mesenchymal stem cells J. Mol. Med. 2008 86 1183 1192 10.1007/s00109-008-0378-3 18600306
323. Penn J.W. Grobbelaar A.O. Rolfe K.J. The role of the TGF-beta family in wound healing, burns and scarring: A review Int. J. Burns Trauma 2012 2 18 28 22928164
324. Johnston C.J. Smyth D.J. Dresser D.W. Maizels R.M. TGF-beta in tolerance, development and regulation of immunity Cell. Immunol. 2016 299 14 22 10.1016/j.cellimm.2015.10.006 26617281
325. Ferguson M.W. Duncan J. Bond J. Bush J. Durani P. So K. Taylor L. Chantrey J. Mason T. James G. Prophylactic administration of avotermin for improvement of skin scarring: Three double-blind, placebo-controlled, phase I/II studies Lancet 2009 373 1264 1274 10.1016/S0140-6736(09)60322-6 19362676
326. Yang D. Wei F. Tewary P. Howard O.M. Oppenheim J.J. Alarmin-induced cell migration Eur J. Immunol. 2013 43 1412 1418 10.1002/eji.201243138 23619968
327. Rankin A.L. Mumm J.B. Murphy E. Turner S. Yu N. McClanahan T.K. Bourne P.A. Pierce R.H. Kastelein R. Pflanz S. IL-33 induces IL-13-dependent cutaneous fibrosis J. Immunol. 2010 184 1526 1535 10.4049/jimmunol.0903306 20042577
328. Lopetuso L.R. Scaldaferri F. Pizarro T.T. Emerging role of the interleukin (IL)-33/ST2 axis in gut mucosal wound healing and fibrosis Fibrogenes. Tissue Repair. 2012 5 18 10.1186/1755-1536-5-18 23062310
329. Schmitz J. Owyang A. Oldham E. Song Y. Murphy E. McClanahan T.K. Zurawski G. Moshrefi M. Qin J. Li X. IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines Immunity 2005 23 479 490 10.1016/j.immuni.2005.09.015 16286016
330. Li D. Guabiraba R. Besnard A.G. Komai-Koma M. Jabir M.S. Zhang L. Graham G.J. Kurowska-Stolarska M. Liew F.Y. McSharry C. IL-33 promotes ST2-dependent lung fibrosis by the induction of alternatively activated macrophages and innate lymphoid cells in mice J. Allergy Clin. Immunol. 2014 134 1422 1432 10.1016/j.jaci.2014.05.011 24985397
331. Smithgall M.D. Comeau M.R. Yoon B.R. Kaufman D. Armitage R. Smith D.E. IL-33 amplifies both Th1- and Th2-type responses through its activity on human basophils, allergen-reactive Th2 cells, iNKT and NK cells Int. Immunol. 2008 20 1019 1030 10.1093/intimm/dxn060 18550585
332. Gao Q. Li Y. Li M. The potential role of IL-33/ST2 signaling in fibrotic diseases J. Leukoc. Biol. 2015 98 15 22 10.1189/jlb.3RU0115-012R 25881899
333. Luzina I.G. Kopach P. Lockatell V. Kang P.H. Nagarsekar A. Burke A.P. Hasday J.D. Todd N.W. Atamas S.P. Interleukin-33 potentiates bleomycin-induced lung injury Am. J. Respir. Cell Mol. Biol. 2013 49 999 1008 10.1165/rcmb.2013-0093OC 23837438
334. McHedlidze T. Waldner M. Zopf S. Walker J. Rankin A.L. Schuchmann M. Voehringer D. McKenzie A.N. Neurath M.F. Pflanz S. Interleukin-33-dependent innate lymphoid cells mediate hepatic fibrosis Immunity 2013 39 357 371 10.1016/j.immuni.2013.07.018 23954132
335. Franchimont D. Kino T. Galon J. Meduri G.U. Chrousos G. Glucocorticoids and inflammation revisited: The state of the art. NIH clinical staff conference Neuroimmunomodulation 2002 10 247 260 10.1159/000069969 12759562
336. Crossley G.H. Brinker J.A. Reynolds D. Spencer W. Johnson W.B. Hurd H. Tonder L. Zmijewski M. Steroid elution improves the stimulation threshold in an active-fixation atrial permanent pacing lead. A randomized, controlled study. Model 4068 Investigators Circulation 1995 92 2935 2939 10.1161/01.CIR.92.10.2935 7586262
337. Zhong Y. Bellamkonda R.V. Dexamethasone-coated neural probes elicit attenuated inflammatory response and neuronal loss compared to uncoated neural probes Brain Res. 2007 1148 15 27 10.1016/j.brainres.2007.02.024 17376408
338. Morais J.M. Papadimitrakopoulos F. Burgess D.J. Biomaterials/tissue interactions: Possible solutions to overcome foreign body response AAPS J. 2010 12 188 196 10.1208/s12248-010-9175-3 20143194
339. Patil S.D. Papadimitrakopoulos F. Burgess D.J. Dexamethasone-loaded poly(lactic-co-glycolic) acid microspheres/poly(vinyl alcohol) hydrogel composite coatings for inflammation control Diabetes Technol. Ther. 2004 6 887 897 10.1089/dia.2004.6.887 15684644
340. Minardi S. Corradetti B. Taraballi F. Byun J.H. Cabrera F. Liu X. Ferrari M. Weiner B.K. Tasciotti E. IL-4 Release from a Biomimetic Scaffold for the Temporally Controlled Modulation of Macrophage Response Ann. Biomed. Eng. 2016 44 2008 2019 10.1007/s10439-016-1580-z 26951461
341. Kim D.H. Martin D.C. Sustained release of dexamethasone from hydrophilic matrices using PLGA nanoparticles for neural drug delivery Biomaterials 2006 27 3031 3037 10.1016/j.biomaterials.2005.12.021 16443270
342. Tsianakas A. Varga G. Barczyk K. Bode G. Nippe N. Kran N. Roth J. Luger T.A. Ehrchen J. Sunderkoetter C. Induction of an anti-inflammatory human monocyte subtype is a unique property of glucocorticoids, but can be modified by IL-6 and IL-10 Immunobiology 2012 217 329 335 10.1016/j.imbio.2011.10.002 22154546
343. Luo J.C. Shin V.Y. Liu E.S. Ye Y.N. Wu W.K. So W.H. Chang F.Y. Cho C.H. Dexamethasone delays ulcer healing by inhibition of angiogenesis in rat stomachs Eur. J. Pharmacol. 2004 485 275 281 10.1016/j.ejphar.2003.11.038 14757151
344. Koedam J.A. Smink J.J. van Buul-Offers S.C. Glucocorticoids inhibit vascular endothelial growth factor expression in growth plate chondrocytes Mol. Cell. Endocrinol. 2002 197 35 44 10.1016/S0303-7207(02)00276-9 12431793
345. Patil S.D. Papadmitrakopoulos F. Burgess D.J. Concurrent delivery of dexamethasone and VEGF for localized inflammation control and angiogenesis J. Control. Release 2007 117 68 79 10.1016/j.jconrel.2006.10.013 17169457
346. Norton L.W. Koschwanez H.E. Wisniewski N.A. Klitzman B. Reichert W.M. Vascular endothelial growth factor and dexamethasone release from nonfouling sensor coatings affect the foreign body response J. Biomed. Mater. Res. A 2007 81 858 869 10.1002/jbm.a.31088 17236219
347. Kastellorizios M. Papadimitrakopoulos F. Burgess D.J. Multiple tissue response modifiers to promote angiogenesis and prevent the foreign body reaction around subcutaneous implants J. Control. Release 2015 214 103 111 10.1016/j.jconrel.2015.07.021 26216396
348. Barrientos S. Stojadinovic O. Golinko M.S. Brem H. Tomic-Canic M. Growth factors and cytokines in wound healing Wound Repair Regen. 2008 16 585 601 10.1111/j.1524-475X.2008.00410.x 19128254
349. Doloff J.C. Veiseh O. Vegas A.J. Tam H.H. Farah S. Ma M. Li J. Bader A. Chiu A. Sadraei A. Colony stimulating factor-1 receptor is a central component of the foreign body response to biomaterial implants in rodents and non-human primates Nat. Mater. 2017 16 671 680 10.1038/nmat4866 28319612
350. Udipi K. Ornberg R.L. Thurmond K.B. 2nd Settle S.L. Forster D. Riley D. Modification of inflammatory response to implanted biomedical materials in vivo by surface bound superoxide dismutase mimics J. Biomed. Mater. Res. 2000 51 549 560 10.1002/1097-4636(20000915)51:4 3.0.CO;2-Z 10880102
351. Hetrick E.M. Prichard H.L. Klitzman B. Schoenfisch M.H. Reduced foreign body response at nitric oxide-releasing subcutaneous implants Biomaterials 2007 28 4571 4580 10.1016/j.biomaterials.2007.06.036 17681598
352. Schwentker A. Vodovotz Y. Weller R. Billiar T.R. Nitric oxide and wound repair: Role of cytokines? Nitric Oxide 2002 7 1 10 10.1016/S1089-8603(02)00002-2 12175813
353. Bogdan C. Regulation of lymphocytes by nitric oxide Methods Mol. Biol. 2011 677 375 393 10.1007/978-1-60761-869-0_24 20941622
354. Cha K.H. Wang X. Meyerhoff M.E. Nitric Oxide Release for Improving Performance of Implantable Chemical Sensors—A Review Appl. Mater. Today 2017 9 589 597 10.1016/j.apmt.2017.10.002 29520370
355. Hersel U. Dahmen C. Kessler H. RGD modified polymers: Biomaterials for stimulated cell adhesion and beyond Biomaterials 2003 24 4385 4415 10.1016/S0142-9612(03)00343-0 12922151
356. Kao W.J. Lee D. In vivo modulation of host response and macrophage behavior by polymer networks grafted with fibronectin-derived biomimetic oligopeptides: The role of RGD and PHSRN domains Biomaterials 2001 22 2901 2909 10.1016/S0142-9612(01)00037-0 11561896
357. VandeVondele S. Voros J. Hubbell J.A. RGD-grafted poly- l -lysine-graft-(polyethylene glycol) copolymers block non-specific protein adsorption while promoting cell adhesion Biotechnol. Bioeng. 2003 82 784 790 10.1002/bit.10625 12701144
358. Zhu J. Tang C. Kottke-Marchant K. Marchant R.E. Design and synthesis of biomimetic hydrogel scaffolds with controlled organization of cyclic RGD peptides Bioconjug. Chem. 2009 20 333 339 10.1021/bc800441v 19191566
359. Karimi F. O’Connor A.J. Qiao G.G. Heath D.E. Integrin Clustering Matters: A Review of Biomaterials Functionalized with Multivalent Integrin-Binding Ligands to Improve Cell Adhesion, Migration, Differentiation, Angiogenesis, and Biomedical Device Integration Adv. Healthc. Mater. 2018 7 e1701324 10.1002/adhm.201701324 29577678
360. Serhan C.N. Pro-resolving lipid mediators are leads for resolution physiology Nature 2014 510 92 101 10.1038/nature13479 24899309
361. Serhan C.N. Chiang N. Dalli J. The resolution code of acute inflammation: Novel pro-resolving lipid mediators in resolution Semin. Immunol. 2015 27 200 215 10.1016/j.smim.2015.03.004 25857211
362. Schwab J.M. Chiang N. Arita M. Serhan C.N. Resolvin E1 and protectin D1 activate inflammation-resolution programmes Nature 2007 447 869 874 10.1038/nature05877 17568749
363. Vasconcelos D.P. Costa M. Amaral I.F. Barbosa M.A. Aguas A.P. Barbosa J.N. Development of an immunomodulatory biomaterial: Using resolvin D1 to modulate inflammation Biomaterials 2015 53 566 573 10.1016/j.biomaterials.2015.02.120 25890752
364. Vasconcelos D.P. Costa M. Amaral I.F. Barbosa M.A. Aguas A.P. Barbosa J.N. Modulation of the inflammatory response to chitosan through M2 macrophage polarization using pro-resolution mediators Biomaterials 2015 37 116 123 10.1016/j.biomaterials.2014.10.035 25453942
365. Tang Y. Zhang M.J. Hellmann J. Kosuri M. Bhatnagar A. Spite M. Proresolution therapy for the treatment of delayed healing of diabetic wounds Diabetes 2013 62 618 627 10.2337/db12-0684 23043160
366. Hong S. Tian H. Lu Y. Laborde J.M. Muhale F.A. Wang Q. Alapure B.V. Serhan C.N. Bazan N.G. Neuroprotectin/protectin D1: Endogenous biosynthesis and actions on diabetic macrophages in promoting wound healing and innervation impaired by diabetes Am. J. Physiol. Cell Physiol. 2014 307 C1058 C1067 10.1152/ajpcell.00270.2014 25273880
368. Swartzlander M.D. Blakney A.K. Amer L.D. Hankenson K.D. Kyriakides T.R. Bryant S.J. Immunomodulation by mesenchymal stem cells combats the foreign body response to cell-laden synthetic hydrogels Biomaterials 2015 41 79 88 10.1016/j.biomaterials.2014.11.020 25522967
369. Chen Z. Wu C. Gu W. Klein T. Crawford R. Xiao Y. Osteogenic differentiation of bone marrow MSCs by beta-tricalcium phosphate stimulating macrophages via BMP2 signalling pathway Biomaterials 2014 35 1507 1518 10.1016/j.biomaterials.2013.11.014 24268199
370. Mansouri S. Merhi Y. Winnik F.M. Tabrizian M. Investigation of layer-by-layer assembly of polyelectrolytes on fully functional human red blood cells in suspension for attenuated immune response Biomacromolecules 2011 12 585 592 10.1021/bm101200c 21306170
371. Kim J.Y. Lee H. Park T. Park J. Kim M.H. Cho H. Youn W. Kang S.M. Choi I.S. Artificial Spores: Cytocompatible Coating of Living Cells with Plant-Derived Pyrogallol Chem. Asian J. 2016 11 3183 3187 10.1002/asia.201601237 27706902
372. Hasturk O. Kaplan D.L. Cell armor for protection against environmental stress: Advances, challenges and applications in micro- and nanoencapsulation of mammalian cells Acta Biomater. 2018 10.1016/j.actbio.2018.11.040
373. Weiden J. Voerman D. Dolen Y. Das R.K. van Duffelen A. Hammink R. Eggermont L.J. Rowan A.E. Tel J. Figdor C.G. Injectable Biomimetic Hydrogels as Tools for Efficient T Cell Expansion and Delivery Front. Immunol. 2018 9 2798 10.3389/fimmu.2018.02798 30546367
374. Kumar M. Coburn J. Kaplan D.L. Mandal B.B. Immuno-Informed 3D Silk Biomaterials for Tailoring Biological Responses ACS Appl. Mater. Interfaces 2016 8 29310 29322 10.1021/acsami.6b09937 27726371
375. Wang H. Morales R.T. Cui X. Huang J. Qian W. Tong J. Chen W. A Photoresponsive Hyaluronan Hydrogel Nanocomposite for Dynamic Macrophage Immunomodulation Adv. Healthc. Mater. 2018 e1801234 10.1002/adhm.201801234 30537061
376. He X.T. Wu R.X. Xu X.Y. Wang J. Yin Y. Chen F.M. Macrophage involvement affects matrix stiffness-related influences on cell osteogenesis under three-dimensional culture conditions Acta Biomater. 2018 71 132 147 10.1016/j.actbio.2018.02.015 29462712
377. Zhao S. Zhang L. Han J. Chu J. Wang H. Chen X. Wang Y. Tun N. Lu L. Bai X.F. Conformal Nanoencapsulation of Allogeneic T Cells Mitigates Graft-versus-Host Disease and Retains Graft-versus-Leukemia Activity ACS Nano 2016 10 6189 6200 10.1021/acsnano.6b02206 27224853
378. Ishida Y. Kondo T. Kimura A. Matsushima K. Mukaida N. Absence of IL-1 receptor antagonist impaired wound healing along with aberrant NF-kappaB activation and a reciprocal suppression of TGF-beta signal pathway J. Immunol. 2006 176 5598 5606 10.4049/jimmunol.176.9.5598 16622029
379. Chang J. Wang Z. Tang E. Fan Z. McCauley L. Franceschi R. Guan K. Krebsbach P.H. Wang C.Y. Inhibition of osteoblastic bone formation by nuclear factor-kappaB Nat. Med. 2009 15 682 689 10.1038/nm.1954 19448637
380. Chang J. Liu F. Lee M. Wu B. Ting K. Zara J.N. Soo C. Al Hezaimi K. Zou W. Chen X. NF-kappaB inhibits osteogenic differentiation of mesenchymal stem cells by promoting beta-catenin degradation Proc. Natl. Acad. Sci. USA 2013 110 9469 9474 10.1073/pnas.1300532110 23690607
381. Mountziaris P.M. Tzouanas S.N. Sing D.C. Kramer P.R. Kasper F.K. Mikos A.G. Intra-articular controlled release of anti-inflammatory siRNA with biodegradable polymer microparticles ameliorates temporomandibular joint inflammation Acta Biomater. 2012 8 3552 3560 10.1016/j.actbio.2012.06.031 22750740
382. Leuschner F. Dutta P. Gorbatov R. Novobrantseva T.I. Donahoe J.S. Courties G. Lee K.M. Kim J.I. Markmann J.F. Marinelli B. Therapeutic siRNA silencing in inflammatory monocytes in mice Nat. Biotechnol. 2011 29 1005 1010 10.1038/nbt.1989 21983520
383. Sager H.B. Dutta P. Dahlman J.E. Hulsmans M. Courties G. Sun Y. Heidt T. Vinegoni C. Borodovsky A. Fitzgerald K. RNAi targeting multiple cell adhesion molecules reduces immune cell recruitment and vascular inflammation after myocardial infarction Sci. Transl. Med. 2016 8 342ra380 10.1126/scitranslmed.aaf1435
384. Yin H. Kanasty R.L. Eltoukhy A.A. Vegas A.J. Dorkin J.R. Anderson D.G. Non-viral vectors for gene-based therapy Nat. Rev. Genet. 2014 15 541 555 10.1038/nrg3763 25022906
385. Mehta A. Baltimore D. MicroRNAs as regulatory elements in immune system logic Nat. Rev. Immunol. 2016 16 279 294 10.1038/nri.2016.40 27121651
386. Li D. Wang A. Liu X. Meisgen F. Grunler J. Botusan I.R. Narayanan S. Erikci E. Li X. Blomqvist L. MicroRNA-132 enhances transition from inflammation to proliferation during wound healing J. Clin. Investig. 2015 125 3008 3026 10.1172/JCI79052 26121747
387. Baumjohann D. Ansel K.M. MicroRNA-mediated regulation of T helper cell differentiation and plasticity Nat. Rev. Immunol. 2013 13 666 678 10.1038/nri3494 23907446
388. Frith J.E. Porrello E.R. Cooper-White J.J. Concise review: New frontiers in microRNA-based tissue regeneration Stem Cells Transl. Med. 2014 3 969 976 10.5966/sctm.2014-0032 24873861
389. Gori M. Trombetta M. Santini D. Rainer A. Tissue engineering and microRNAs: Future perspectives in regenerative medicine Expert Opin. Biol. Ther. 2015 15 1601 1622 10.1517/14712598.2015.1071349 26200765
390. Li J. Kooger R. He M. Xiao X. Zheng L. Zhang Y. A supramolecular hydrogel as a carrier to deliver microRNA into the encapsulated cells Chem. Commun. (Camb.) 2014 50 3722 3724 10.1039/C4CC00156G 24577244
391. Morishita Y. Imai T. Yoshizawa H. Watanabe M. Ishibashi K. Muto S. Nagata D. Delivery of microRNA-146a with polyethylenimine nanoparticles inhibits renal fibrosis in vivo Int. J. Nanomed. 2015 10 3475 3488 10.2147/IJN.S82587 25999712
해당 논문의 주제분야에서 활용도가 높은 상위 5개 콘텐츠를 보여줍니다.
더보기 버튼을 클릭하시면 더 많은 관련자료를 살펴볼 수 있습니다.
*원문 PDF 파일 및 링크정보가 존재하지 않을 경우 KISTI DDS 시스템에서 제공하는 원문복사서비스를 사용할 수 있습니다.
오픈액세스 학술지에 출판된 논문
※ AI-Helper는 부적절한 답변을 할 수 있습니다.