Ex vivo gene transfer to full thickness wounds. A platform for autologous 'smart' tissue engineering for tissue repair.

Jan Vranckx
Persbericht

Ex vivo gene transfer to full thickness wounds. A platform for autologous 'smart' tissue engineering for tissue repair.

Slimme cellen genezen diepe wonden.

 

Grote wonden vergen complexe behandelingen. Een onderzoeksteam van Plastische Chirurgen in het UZ-Leuven is erin geslaagd om diepe wonden te repareren met autologe (lichaamseigen) ‘smart tissue engineering’. Bij varkens is het een succes. Nu mensen nog.

 

Derdegraads brandwonden, uitgebreide verwondingen na verkeersongevallen of na een operatie vanwege kanker, de plastisch reconstructief chirurg wordt er dagelijks mee geconfronteerd. Vaak is een complexe chirurgische weefsel transplantatie nodig die op zich een oorzaak is van nieuwe verwonding daar waar het donor weefsel genomen werd. Maar wanneer zulke uitgebreide behandeling niet wordt uitgevoerd, zijn stugge littekens het gevolg en die leiden al te vaak tot forse bewegingsbeperking rond gewrichten en verminderde motivatie van de patiënt voor sociale reïntegratie na het ongeval.

Bij sommige erg complexe defecten, zoals zeer uitgebreide diepe brandwonden, is er zelfs geen echt degelijke oplossing. De sinds lang gebruikte huidgreffen zijn dun en vertegenwoordigen enkel de oppervlakkige laag van huid. De recent ontwikkelde ‘dermale’ kunsthuid, die de diepere laag van de huid vertegenwoordigt, kan dienen als een tapijtje waarop huidgreffen kunnen gelegd worden. Die kunstdermis is nog steeds dun en vermits ze samengesteld is uit bindweefsel van koeien of varkens,wordt ze geleidelijk afgebroken eens op een wonde geplaatst bij de mens.

 

Jan Vranckx van de Universiteit van Leuven promoveerde op een strategie die huiddefecten behandelt met “autologe (lichaamseigen),elastische,goed doorbloede huid van volledige dikte”. Autologe smart tissue engineering, heet deze strategie.

 

‘Smart tissue engineering’ start met het kweken van cellen uit de verschillende huidlagen en van lichaamseigen oorsprong. “Ook deze methode bestaat al 20 jaar maar heeft tot weinig geleid”, zegt Vranckx. Om deze cellen een volwaardige elastische huid te doen vormen is heel wat meer nodig. “ Zie de wonde als een constructie site; je hebt bouwmateriaal nodig, mankracht maar ook een coördinator, een architect. In de wondheling fungeren groei-eiwitten ,groeifactoren, als de coördinators. Deze eiwitten worden afgescheiden door alle cellen die iets in de helende wonde te betekenen hebben. Bij ziekte of na trauma is de aanmaak van die groeifactoren fors verminderd, hoewel ze net dan meest benodigd zijn.

Vranckx: “Die groeifactoren zijn erg belangrijk vanaf het prille begin van de wondheling, van de vorming van stolsel en nieuwe bloedvaatjes in de wonde tot het zacht en wit worden van het litteken. Die groeifactoren gebruiken om de wondheling te bevorderen is dus een logische stap”.

Helaas worden die groeifactoren snel afgebroken als je ze simpelweg in de wonde zou spuiten. Dit kan omzeild worden door de cellen zélf als miniproductie eenheden de eiwitten te laten aanmaken.

Vranckx:” Nadat we in het labo het DNA van de groeifactor naar keuze in de huidcellen hebben aangebracht wordt dit DNA in de cel zelf vertaald naar het groeifactor eiwit dat dan in de wonde wordt afgescheiden; niet éénmalig maar constant. De cellen vormen zich in de wonde als kleine fabriekjes van de groeifactoren.

Om deze strategie optimaal te laten verlopen, focust Vranckx op stam cellen van de huid.“Zulke cellen zijn in staat om op 4 weken tijd uit 10 cm2 zoveel cellen te produceren dat de ganse huid oppervlakte van de mens zou kunnen bedekt worden. Evengoed kunnen die cellen fungeren als dragers van die groeifactoren die in wondheling een groot verschil kunnen maken”.

Deze strategie leidde in een varkensmodel tot snelle heling met laag per laag goed doorbloed weefsel. “Deze doorbloeding is het grote verschil met eerder voorgestelde behandelingsmethoden door tissue engineering” zegt Vranckx. De fusie van tissue engineering technieken met gen therapie brengt ons een hele stap verder in die zoektocht naar tissue engineering van weefsels en zelfs organen.

Hoe lang het nog duurt alvorens we ‘lichaamseigen weefsel als ‘take away’ kunnen  bestellen en gebruiken is onvoorspelbaar.

De euforie was nabij toen eind jaren ‘90 op een ‘naakt muismodel’ een mensen oor werd gegroeid. Door tissue engineering leek toen alles mogelijk. Dit oor was slechts een matrix in de vorm van een oorschelp, die onder de erg elastische en dunne huid van die muizensoort werd gehecht. Die matrix werd natuurlijk in korte tijd overwoekerd met littekenweefsel en bloedvaatjes en bij de muis niet afgestoten. Vranckx:“Zowel bij varken als mens komen we geen stap verder als het weefsel, aangemaakt door tissue engineering, niet goed doorbloed is en niet lichaamseigen. Met de huidige wereldwijde ambitie en motivatie in tissue engineering lijkt me nog een 10-tal jaar voor authentieke huid op bestelling rationeel.”

Tot dan je hand er zeker niet voor in het vuur steken.

 

 

Bibliografie

References: zie bibliografie in 'scriptie'  p7 en p171-178.

1. Vranckx JJ, B.Vandenhof.,D.Misselyn.,

      N. Verhelle,O.Heymans

      The gracilis free flap: more than a gracile flap for             lower leg reconstructions.

      J.Reconstructive Microsurgery 20,

      36-41,2004.

2. Vandevoort M, Vranckx JJ fabre G.

      Perforator Topography of the DIEP flap.

      In: Mosby Yearbook of Plastic &        Reconstructive Surgery 2004 ,p259, Ed.         Miller S,Illinois.

3. JJ. Vranckx, P Delaere , F.C Wei,

      B Van den hof.

      Vascularised Fascia and Supra-Fascial              Radialis Forearm Free Flaps in Head and Neck             Reconstructions.”

      Microsurgery, 20 (6), 290,2000.

4. Hunt JL.,Purdue GF.,Pownell P.,

      Rohrich R.

      Burns: acute burns,burn surgery and post-burn              reconstruction.

      Select.Read. Plast.Surg. 8: 1-37.,1997

5. Massagé P.,Vandenhof B.,Vranckx JJ.

      Full face resurfacing of third degree burns with               artificial dermis: barriers and opportunities.

      J.Plast.Reconstr.Surg.59; S1-12,2006.

6. Andreadis ST.

      Gene-modified tissue engineered skin: the next               generation of skin substitutes.         Adv.Biochem.Eng.Biotechnol.103;

      241-2007.

7. Metcalfe A.,Ferguson M.

      Harnessing wound healing and           regeneration for tissue engineering.

      Biochem.Soc.Trans. 33,413,2005

8. Hebda P.,Sandulache VC.

      The biochemistry of epidermal healing.

      In: The epidermis in wound healing. CRC          Press Boca Raton 2004 EdsRovee DT,           Maibach HI.

9. Singer A., Clark R.A.

      Mechanisms of disease,

      Cutaneous Wound Healing.

            N.Engl.J.Med. 341,738,1999

10. Burke JF, Yannas IV, Quinby WC Jr,

      Bondoc CC, Jung WK.

      Successful use of a physiologically acceptable               artificial skin in the treatment of extensive     burn injury.

      Ann Surg. 194,413,1981

11. Sheridan RL.,Tompkins RG.

      Skin substitutes in burns.

      Burns 25, 97,1999

 

12. Yarlagadda PK.,Chandrasekharan M.,

      Shyan JY.

      Recent advances and current               developments in tissue scaffolding.               Biomed.Mater.Eng.15;159-77,2005.

13. Muschler GF.,Nakamotot C.,Griffith LG.            Engineering principles of clinical cell-            based tissue engineering.

      J.Bone Joint Surg.Am.86;1541-58,2004.

14. Griffith LG.

      Emerging design principles in biomaterials and               scaffolds for tissue engineering.

      Ann.NY.Acad.Sci. 961,83,2002

15. Rosso F., Marino G.,Giordano A.,

      Barbarisi M.,Parmeggiani D.,,Barbarisi A.

      Smart materials as scaffolds for tissue               engineering.

      J.Cell.Physiol. 203,465,2005

16. Rumalla V.K.,Borah G.L.

      Cytokines,growth factors and Plastic                 Surgery.

      Plast.Reconstr.Surg.108,719,2001

17. Vranckx JJ.,Vermeulen P.,Dickens S.

      Smart Autologous Tissue Engineering:             stem cells and growthfactors, scaffolds         and genes.

      J.Plast.reconstr.Surg.59;S1-12,2006.

18. Gambardella L.,Barrandon Y.

      The multifaceted adult epidermal stem               cell.

            Curr.Opinion Cell Biol.15, 771,2003

19. Conrad C.,Huss R.

      Adult stem cell lines in regenerative   medicine                and reconstructive surgery.

            J.Surg.Research 124, 201,2005

20. Brown GL.,Nanney LB.

      Enhancement of wound healing by topical       treatment with epidermal growth factor.

      N.Engl.J.Med.321,76,1989

21. Fu X.,Li X.,Cheng B.,Chen W.,Sheng Z.           Engineered growth factors and cutaneous wound      healing, success and possible questions in the past ten               years.

      Wound Rep.Reg 13,122,2005

22. Bennett N., Schultz P.

      Growth factors and wound healing, biochemical             properties of growth factors and their receptors.         Am.J.Surg.165,728,1993

23. Vranckx JJ, Yao F.,Eriksson E.

      Gene Transfer of Growth Factors for Wound Repair”.

     In: The Epidermis in Wound healing; 2004: 265-              283,Eds. D.Rovee, H.Maibach,

            CRC Press, Boca Raton, Fl.USA.

24. Yang NS.,Burkholder J., Roberts B.,   Martinell B., Mc Cabe D.

             In vivo and in vitro gene transfer to mammalian              somatic cells by particle bombardment.

      Proc.Natl.Acad.Sci.USA 87, 9568,1990.

25. Benn SI.,Whitsitt J.,Broadley K.,

      Nanney L.,Perkins D.,He L.,Patel M.,                 Morgan J,Swain W.,Davidson JM.

      Particle mediated gene transfer with   transforming         growth factor beta1 cDNAs              enhances wound repair      in rat skin.

      J.Clin.Invest. 98,2894,1996

26. Eriksson E., Yao F., Svensjo T.,Winkler

      T., Slama J.,Macklin M., Andree C.,

      mc Gregor M., Hinshaw V., Swain W.

             In vivo gene transfer to skin by          microseeding.

      J.Surg.Res. 78, 85,1998

27. Andreadis ST.

      Gene transfer to epidermal stem cells:                implications for tissue engineering. Expert.

      Opin.Biol.Ther. 4;783-800,2004.

28. Ehrenreich M.,Ruszczak Z.

      Update on tissue-engineered biological            dressings.

      Tissue Eng.12; 2407-24,2006.

29. Wang HJ.,Pieper J.,Schotel R.,            vanBlitterswijk CA., Lamme EN.

      Stimulation of skin repair is dependent on fibroblast     source and presence of extracellular matrix.

      Tissue Eng.10;1054-64,2004.

30. Naughton GK.,Mansbridge JN.

      Human based tissue engineered implants for   reconstructive surgery.

      Clin.Pl.Surg.26, 579,1999

31. Yao F, Svensjo T, Winker T, Lu M,

      Eriksson E.

             Tetracycline repressor, tetR,rather than            tetR-mamalian cell transcription fusion          derivative, regulates inducible gene expression            in mammalian cells.

             Human Gene Therapy 9, 1939,1998.

32. Yao F.,Eriksson E.

      A novel tetracycline-inducible viral replication               switch.

      Human Gene Therapy 10, 419,1999.

33. Sullivan T.P.,Eaglstein W.H.Davis SC.,             Mertz P.

             The pig as a model for human wound                healing.

      Wound. Rep.Reg. 9; 66-76, 2001

34. Vardaxis N.J., Brans TA. Boon ME.,   Kreis RW., Marres LM.

      Confocal laser scanning microscopy of            porcine skin ; implications fro human             wound healing studies.

            J.Anat. 190; 601-11,199

35. Vranckx JJ, Yao F, Petrie N,

      Augustinova H, Hoeller D, Visovatti S, Slama J,             Eriksson E.

             In vivo gene delivery of Ad-VEGF121 to full-    thickness wounds in aged pigs results in high levels                 of VEGF expression but not in accelerated healing.

      Wound Repair Regen. 2005; 13:51-60.

36. Vranckx JJ.,Hoeller D.,Petrie N.

      Velander P., Theopold C.,,Eriksson E.,

             Yao F.

             Cell suspension cultures of allogenic                keratinocytes are efficient carriers for ex vivo gene     transfer and accelerate healing of full thickness skin      wounds by overexpression of hEGF.

             Accepted for publication in

             Wound Rep.Reg 2007.

37. Vranckx JJ, Slama J, Preuss S,

      Svensjö T, Visovatti S, Breuing K,

      Bartlett R,

      Pribaz J, Eriksson E.

      Wet wound healing.

      Plast Reconstr Surg 110(7), 1680, 2002.

38. Eriksson E.,Vranckx JJ.

      Wet wound healing: from laboratory to            patient to Gene therapy.

      Am.J.Surg. 2004; 188:36-41.

39. Vranckx JJ., Steinstrasser L,                 Mohammadi-Tabrisi A., Jacobson F.,

      Mittler D., Lehnhardt M., Langer S.,   Kuhnen C.,           Gatermann S., Steinau HU.,Eriksson E.

      A novel titanium wound chamber for the          study of wound infections in pigs.

      Comp Med. 2006 Aug;56:279-85.

      VJJ, SL : co-first authors.

40. Martin, P.

      Wound healing--aiming for perfect skin            regeneration.

             Science 1997; 276: 75-81.

41. Steed D.L.

      Modifying the wound healing response           with exogenous growth factors.Clinics in      Pl.Surg.25,397,1998

42. Robson MC.,Mustoe TA.,Hunt TK.

      The future of recombinant growth factors in wound      healing.

      Am.J.Surg.176,80 S,1998

43. Yao F., Visovatti S., Johnson S.,Chen M.,

      Slama J.,Wenger A.,Eriksson E

      Age and Growth Factors in porcine full            thickness wound healing.

      Wound Rep.& Gen.9,371,2001

44. Rivard A., Fabre JE., Silver M., Chen D.,           Murohara T., Isner J.

      Age dependent impairment of             angiogenesis.

      Circulation 99: 111,1999

45. Ashcroft GS.,Horan MA.,Ferguson MWJ.

      Aging alters the inflammatory and endothelial                adhesion molecule profiles during human     cutaneous wound healing.

      Lab.Invest. 78 :47,1998

46. Khorramizadeh MR.,Tredget EE.,

      Telasky C.,Shen Q.,Gharay A.

      Aging differentially modulates the expression of            collagen and collagenase in dermal fibroblasts.

      Mol.Cell.Biochem. 194:99,1999

47. Davidson J.M.

      Gene therapy of wounds. In: The epidermis

      in wound healing.

      CRC Press Boca Raton 2004. Eds: Rovee          DT. Maibach HI.

48. Andree C., Swain W.F.,Page C.P,

      Macklin MD.,Slama J.,Hatzis D.,

      Eriksson E.

            In vivo gene transfer and expression of            human epidermal growth factor accelerates   wound repair.

      Proc.Natl.Acad.Sci.USA.91, 12188,1994.

49. Eming SA., Whitsitt JS., He L.,Krieg T.,            Morgan JR., Davidson JM.

      Particle mediated gene transfer of PDGF           isoforms promotes wound repair.

            J.Invest.Dermatol. 112, 297,1999.

50. Liechty KW.,Sablick TJ.,Adzick NS.,                 Cromblestone TM.Recombinant adenoviral mediated gene transfer in ischaemic impaired          wound healing.

      Wound RepReg.7,148,1999.

51. Morgan JR.,Eden CA.

      Retroviral-mediated gene transfer into               transplantable human epidermal cells.

      Prog.Clin.Biolog.Res. 365, 417,1991.

52. Petrie N., Vranckx JJ., Hoeller D.,Yao F.,            Eriksson E.

      Gene delivery of PDGF for wound healing        therapy.

      J Tissue Viability 15 :16-21,2005

53. Petrie N., Vranckx JJ,Yao F.,Hoeller D.,              Eriksson E.

      Stable cell lines of PDGF-BB expressing            fibroblasts impair wound healing in a             porcine full thickness skin model.

      Plastic Surgery Research Council ,

      Las Vegas, NV, 2003.

54. Deodato B.,Arsic N.,Zentilin L.           Recombinant AAV vector encoding               human    VEGF165 enhances wound healing.

      Gene Ther.9: 777,2002.

55. Ailawadi M., Lee JM.,Lee S.,Hackett N.,           Crystal RG., Korst RJ.

      Adenovirus vector-mediated transfer of the vacular      endothelial growth factor cDNA to healing   abdominal fascia enhances

      vascularity and bursting strength in mice         with normal and impaired wound healing.

      Surgery 131,219,2002 2002

56. Gitay-Goren H.,Cohen T.,Tessler S.,   Soker S.,Gengrinovitch S.,Rockwell P.,           Klagsbrun M.,Levi BZ.,Neufeld G.

             Selective binding of VEGF121 to one of the three             VEGF receptors of vascular endothelial cells.

      J.Biochem.271;5519-23,1996.

57. Losordo D., Vale P., Symes J.,

      Gene Therapy for myocardial              angiogenesis.

      Circulation 1998; 98: 2000.

58. Gabel H.,Bitter-Suermann H.,

      Henriksson C., Save-Soderbergh J.,

      Lundholm K.,Brynger H.

             Streptozotocin diabetes in juveline    pigs.Evaulation of an experimental model.

             Horm.Metab.Res.17,275,1985.

59. Nakamura Y.,Muguruma Y.,Yahata T.,               Miyatake H.,Sakai D.,Michida J.,Hotta T.,     Ando K.

      Expression of CD90 on keratinocyte stem/progenitor cells.

      Br.J.Dermatol.154,1062,2006.

60. Hengge U., Chan E.,Hampshire V.,

      Foster RA., Vogel JC.

The derivation and characterization of Pig Keratinocyte cell lines that retain the ability to differentiate. J.Invest.Deramtol.106; 287-293,1996

61. Regauer S.,Compton C.

      Cultured porcine epithelial grafts : an                improved method.

      J.Invest.Dermatol. 94; 230-234,1990.

62. Boyce ST., Ham RG.

      Calcium regulated differentiation of   normal    human keratinocytes in chemically defined   clonal cultures and serum-free serial cultures.

      J.Invest.Dermatol. 81; 33-40, 1983.

63. Maciag T., Nemore RE., Weinstein R.,               Gilchcrest BA.

      An endocrine approach to the control of          epidermal growth: serum free cultivation of   human keratinocytes.

             Science 211; 1452-4, 1981

64. Rosdy M.,Pisani A.,Ortonne JP.

     Production of basement membrane components              by a reconstructed epidermis cultured in the                absence of serum and dermal factors.               Br.J.Dermatol.129 :227,1993.

 

65. Green H.,Kehinde O.,Thomas J.

            Growth of cultures of human keratinocytes into              multiple epithelia suitable for grafting.             Proc.Natl.Acad.Sci.USA.76,5665,1979

66. Boucher F.,Poumay Y., Degen A.,

      Paye M., Leloup R.

            Utilization of human cultured epidermal            keratinocytes : irreversibility of the                 inhibition              of proliferation induced in stored detached              cultures.

      Burns 17; 205-8, 1991.

67. Freshney RI.

      Culture of animal cells.

      4th ed. Ch.2. NewYork, Wiley-Liss. 2000

68. Park HS.,Kang HJ.,Kim Ch,Han ES.,

      Han K., Kim TH.,Gin YJ.,Son YS.

      Application of physical force is essential to     enrich for epidermal stem cells in primary human         keratinocyte isolation.

      Tissue Eng.10,343,2004

69. Hybbinette S.,Bostrom M.,Lindberg K.

      Enzymatic dissociation of keratinocytes           from human skin biopsies for in vitro cell      propagation.

      Exp.dermatol.8,30,1999.

70. Meyer-Hoffert U.,Rogalski C.,Seifert S.,            Schmeling G.,Wingertszahn J.,Proksch E.,     Wiedow O.

      Trypsin induces epidermal proliferation            and inflammation in murine skin.

      Exp.Dermatol.13,234,2004.

71. Green H.,Rheinwald JG.,Sun TT.

      Properties of an epithelial cell type in                culture: the epidermal keratinocyte and its    dependence on products of the fibroblast.

      Prog.Cln.Biol.Res.17,493,1977.

72. Boyce S.,Ham RG.

      Calcium-regulated differentiation of   normal    epidermal keratinocytes in chemically defined              clonal culture and serum-free serial culture.                 J.Inv.dermatol.81,33,1983.

73. Thompson CH.,Rose BR. Cossaert YE.

      Optimised growth of human epidermal              cells in vitro without the use of a feeder        layer or collagen substrate.Austr.J. Exp.       Biol.Med.Sci. 63;147-156, 1985

74. Clark RAF.

      Fibronectin matrix deposition and fibronectin receptor expression in healing and normal skin.

      J Invest Dermatol, 94, 128S, 1990

75. Black AF.,Bouez C.,Perrier E.,

      Schlotmann K.,Chapuis F.,Damour O.                Optimization and characterization of an         engineered human skin equivalent.

      Tissue Eng.11,723, 2005

 

76. Mitsi M, Hong Z, Costello CE,

      Nugent MA.

             Heparin-mediated conformational changes in fibronectin expose vascular endothelial growth           factor binding sites.

             Biochemistry, 45, 10319, 2006

77. Navsaria HA., Myers SR.,Leigh IM., McKay IA.

      Culturing skin in vitro for wound therapy.

      Trends. Biotechnol. 13; 91, 1995

78. Cai S.,Fatherazi S.,Presland RB.,

      Belton CM., Izutsu KT.

      TRPC channel expression durin calcium-          induced differentiation of human gingival     keratinocytes.

      J.Dermatol.Sci.40,21,2005.

79. Xie Z.,Singleton PA.,Bourguignon LY.,            Bikle DD.

      Calcium-induced keratinocyte             differentiation requires src-and fyn-mediated               phosphatidylinositol 3-kinse-dependent       activation of phospholipase C-gamma1.

      Mol.Biol.Cell.16,3236,2005

80. Hennings H., Michael D., Cheng C.,   Steinert P., Holbrook K., Yuspa SH.

      Calcium regulation of growth and       differentiation of mouse epidermal cells in     culture.

      Cell 19; 245-254,1980

81. Wille JJ., Pittelkow MP., Shipley GD.,                Scott RE.

      Integrated control of growth and        differentiation of normal human       prokeratinocytes cultured in serum-free         medium: clonal analysis, growth kinetics            and cell cycle studies.

      J.Cell.Physiol. 121; 31-44,1984.

82. Kiefer K.,Clement J.,Garidel P.,

      Peschka-Suss.

      Transfection efficiency and cytotoxicity of      non-viral gene transfer reagents in human    smooth muscle and endothelial cells.

      Pharma.Res.21,1009, 2004.

83. Thomas M.,Ge Q.,Lu JJ.,Chen J., Klibanov AM.

      Cross-linked small polyethylenimines:              while still non-toxic,deliver DNA efficiently to             mammalian cells in vitro and in vivo.

      Pharm.Res.22,373,2005.

84. Zellmer S.,Gaunitz F.,Salvetter J.,

      Surovoy A.,Reissig D.,Gebhardt R.

      Long-term expression of foreign genes in         normal human epidermal keratinocytes          after transfection with lipid/DNA complexes.

      Histochem.Cell.Biol.115,41,2001.

 

85. Distler JHW.,Kurowska-Stolarska M.,               Micehl BA.,Gay RE,Gay S.,Distler O.

     Nucleofection, a new ,highly efficient                transfection method for primary       keratinocytes.

            Experim.Dermatol.14,315, 2005

86. Vogt PM, Thompson S, Andree C, Liu P.,

     Breuing K, Hatzis D, Brown H,

     Mulligan RC, Eriksson E.

     Genetically modified keratinocytes transplanted              to wounds reconstitute the epidermis.

     Proc Natl Acad Sci USA, 91(20),

     9307, 1994

87. Hefton, J. M., Madden, M. R., Finkelstein,

     J. L., and Shires, G. T.

            Grafting of burn patients with allografts of        cultured epidermal cells.

     Lancet 1983; 2: 428-430.

88. De Luca, M., Albanese, E., Bondanza, S.,         Megna, M., Ugozzoli, L., Molina, F.,               Cancedda, R., Santi, P. L., Bormioli, M.,      Stella, M., and et al.

     Multicentre experience in the treatment of         burns with autogenic and allogenic cultured                epithelium, fresh or preserved in a frozen state.

     Burns 1989; 15: 303-309.

89. Brown GL.,Nanney LB.,Griffen J.,

     Cramer AB.,Yancey JM.,Curtsinger LJ.,             Holtzin L.,Schultz GS.,Jurkiewicz MJ.,             Lynch JB.

     Enhancement of wound healing by topical        treatment with epidermal growth factor.

     N.Engl.J.Med.1989; 321: 76-80.

90. Falanga V., Eaglestein WH., Bucalo B.,             Katz MH.,Harris B.,Carson P.

     Topical use of human recombinant epidermal   growth factor in venous ulcers.

     J.dermatol.Surg.Oncol.1992; 18: 604-6.

91. Snyder RJ.

     Treatment of nonhealing ulcers with allografts.

            Clin.dermatol. 23,388,2005.

92. Aubock J., Irschick E., Romani N.,      Kompatscher P.,Hopfl R.,Herold M.,              

     Schuler G.,Fritsch P.

     Rejection, after a slightly prolonged   survival                 time, of Langerhans cell-free allogenic cultured           epidermis used for wound coverage in humans.

     Transplantation 1988; 45: 730-737.

93. Rosenberg AS.

     Cellular basis of skin allograft rejection: an in   vivo model of immune-mediated tissue          destruction.

     Ann.Rev.Immunolog.1992; 10: 333-337

94. Morhenn VB.,Benike CJ.,Cox AJ.,

      Charron DJ.,Engleman EG.

      Cultured human epidermal cells do not             synthesize HLA-DR.

      J.Invest.Dermatol. 1982; 78; 32-36

 

95. Dierch A.,Chan SH.,Benoist C.,

      Mathis D.

      Graft rejection by T cells not restricted by        conventional major histocompatibility           complex molecules.

      Eur.J.Immunol.1993; 23: 2725-28

96. Hunt JP.,Hunter CT.,Brownstein M., Hultman C.,deSerres S.,Bracey L.,

             Frelinger J., Meyer A.

      Host priming, not target antigen type,               decides rejection rate in mice primed with      MHC-II “knock-out” cultured       keratinocytes.

      J.Surg.Research 1998; 76: 32-36.

97. Thivolet J, Faure M, Demidem A,

      Mauduit G.

      Long-term survival and immunological             tolerance of human epidermal allografts         produced in culture.

      Transplantation 1985; 42: 274-280.

98. Rouabhia M

      Permanent skin replacement using chimeric     cultured sheets comprising xenogeneic and syngeneic keratinocytes.

      Transplantation 1996; 61: 1290-1300.

99. Hubbell JA.

      Materials as morphogenetic guides in               tissue engineering.

      Curr. Opin. Biotechnol. 14,551,2003

100. Enoch S., SHaaba H.,Dunn KW. Informed      consent should be obtained from patients to use        products (skin substitutes) and dressings        containing biological material.

      J.Med.Ethics,31;2-6,2005

101. Clark RAF.

      Fibronectin matrix deposition and fibronectin receptor expression in healing and normal skin.

             J Invest Dermatol 94, 128S, 1990

102. Mitsi M, Hong Z, Costello CE,

      Nugent MA.

      Heparin-mediated conformational changes in fibronectin expose vascular endothelial         growth factor binding sites.

             Biochemistry 45(34), 10319, 2006

103. Bauer S.,Bauer RJ, Liu Z.,Chen H.,    Goldstein L.,Velazquez OC.

      Vascular endothelial growth factor-C                promotes vasculogenesis,angiogenesis,       and collagen constrition in three-dimensional    collagen gels.

            J.Vasc.Surg. 41, 699,2005.

104. Carmeliet P.,Collen D.

      Molecular analysis of blood vessel formation and          disease.

            The American Physiol.Soc. invited     review,H2091,1997.

 

105. Dvorak HF.,Detmar M.,Claffey KP., Nagy JA.,van de water L.,Senger DR.             Vascular permeability factor/vascular           endothelial factor:an important mediator of   angiogenesis in malignacy and        inflammation.

                Int.Arch.Allergy Immunol.                107:233,1995

106. Li B, Sharpe EE, Maupin AB,

          Teleron AA, Pyle AL, Carmeliet P,

          Young PP.

                VEGF and PLGF promote adult        vasculogenesis by enhancing EPC recruitment            and vessel formation at the site of tumor                 neovascularization.

                FASEB J 20(9), 1495, 2006

107. Ferrara N, Gerber HP, Lecouter J.µ

         The biology of VEGF and its receptors.

         Nat Med. 9, 669, 2003

108. Brown NJ., Smyth EA.,Cross SS.,

         Reed MW.

                Angiogenesis induction and regression in    human surgical wounds.

                Wound repair and Regen.2002;

                10:245-51

109. Tonnesen MG.,Feng X.,Clark RA.

         Angiogenesis in wound healing.     J.Invest.Dermatol.Symp.Proc.2000;

         5;40-6.

110. Rehman J, Li J, Orschell CM,

         March KL.

         Peripheral blood "endothelial progenitor       cells" are derived from       monocyte/macrophages and serete                angiogenic          growth factors.

         Circulation, 107(8), 1164, 2003

111. Relou IA, Damen CA,

         van der Schaft DW, Groenewegen G,             Griffioen AW.

         Effects of culture conditions on endothelial cell          growth and responsiveness.

         Tissue Cell, 30(5), 525, 1998

112. Cai J, Jiang WG, Ahmed A, Boulton M.

         Vascular endothelial growth factor induced                 endothelial cell proliferation is regulated by interaction between VEGFR-2,SH-PTP1 and        eNOS.

         Microvasc. Res. 71, 20, 2006

113. Dardik R.,Inbal A.

         Complex formation between tissue transgluaminase II (tTG) and vascular            endothelial growth receptor 2 (VEGFR-2):          proposed mECshanism for

         modulation of endothelial cell response to    VEGF.

         Exp.Cell Res.312; 2006: 2973-2982.

 

114. Duan HX, Cheng LM, Jian W, Hu LS, Lu GX.

         Angiogenic potential difference between      two types of endothelial progenitor cells       from human umbilical cord blood.

         Cell Biol. Int., 30(12), 1018, 2006

115. Collen A., Hanemaaijer R., Lupu F., Quax PH.,van Lent N.,

         Grimbergen J et al.

                Membrane-type matrix metalloproteinase-     mediated angiogenesis in a fibrin-collagen    matrix.

                Blood 2003; 101:1810-7.

116. Taraboletti G.,D’Ascenzo S., Borsotti P.,        Giavazzi R.,Pavan A.,Dolo V.

         Shedding of the Matrix metalloproteinases   MMP-2,MMP-9 and MT1-MMP as membrane             vesicle-associated components by endothelial     cells.

         Am.J.Pathol.2002;160: 673-680.

117. Krengel S.,Alexander M.,Brinckmann J.,         Tronnier M.

         MMP-2 ,TIMP-2 and MT1-MMP are              differentially expressed in lesional skin of     melanocytic nevi and their expression is          modulated by UVB-light.

         J.Cutan.Pathol.2002;29:390-6.

118. Tjwa M, Luttun A, Auterio M,

         Carmeliet P.

         VEGF and PLGF: two pleiotropic growth        factors with distinct roles in development and             hemeostasis.

         Cell Tissue Res, 314, 5, 2003

119. Li B, Sharpe EE, Maupin AB,

         Teleron AA, Pyle AL, Carmeliet P, Young PP.

         VEGF and PLGF promote adult         vasculogenesis by enhancing EPC                 recruitment           and vessel formation at the site of tumor               neovascularization.

         FASEB J, 20(9), 1495, 2006

120. Gossen M, Bujard H.

         Tight control of gene expression in                mammalian cells by tetracycline-responsive promoters.

         Natl. Acad. Sci. U.S.A., 89, 5547, 1992

121. Eppley B.,Woodell J.,Higgins J.

          Platelet quantification and growth factor       analysis from platelet rich plasma:   implications          for wound healing.

          J.Plast Rec.Surg. 114,1502,2004

122. Ellis WC.,Cassidy LK.,Finney AS.,   Spiwak AJ.,Riley JB.

          Thromboelastograph analysis of platelet gel                formed with different thrombin         concentrations.

          J.Extra Corpor.Technol. 37,52,2005.

 

123. Vermeulen P., Dickens S., Vranckx JJ.              Platelet-rich-plasma & keratinocytes              enhance healing & deposition of fibronectin in                 a porcine full-thickness wound model.           J.Plast.Reconstr.Aesth.Surg.A ,2007.

          In print

124. Zechner W.,Tangl S.,Tepper G.,Furst G.,        Haas R.,Mailath G.,Watzek G.

          Influence of platelet rich plasma on                osseous healing of dental implants: a             histologic and histomorphometric study in           minipigs.

          Int.J.Oral Maxillofac.Implants           16,15,2003.

125. Vranckx JJ., Segers K.,Vertriest R.,    Vermeulen P.,Massage P.,

          Van Brussel M.

          Vacuum assisted closure of complex              wounds: sense and nonsense.

          Submitted. Crit.Care Med.

126. Argenta LC.,Morykwas MJ.,

          Marks MW., DeFranzo AJ.,Molnar JA.,        David LR.

          Vacuum assisted closure: state of the            clinic art.

          Plast.Reconstr.Surg.117:127-42,2006

127. Morykwas MJ.,Simpson J.,Punger K.,             Argenta A.,Kremers L.,Argenta J.

          Vacuum assisted closure: basic research       and physiologic foundation.

          Plast.Reconstr.Surg.117,121-26,2006.

128. Pribaz, JJ., Fine, N., Orgill, DP.

          Flap prefabrication in the head and                neck: A 10-year experience.

          Plast. Reconstr. Surg. 103: 808, 1999.

129. Tanaka Y., Sung KC.,Tsutsumi A.,

          Ohba S., Ueda K.,Morrison WA.

          Tissue engineering skin flaps: which             vascular carrier,arteriovenous shunt              loops or arteriovenous bindle, has more           potential for angiogenesis and tissue            generation ?

                Plast.Reconstr.Surg.112: 1626,2003.

130. Tark KC., Tuchler RE., Shaw WW.

          Flap prefabrication: Effectiveness of              different vascular carriers.

          Ann. Plast. Surg. 37: 298, 1996.

131. Sato, Y. (Ed.).

          Frontline of Angiogenesis: Mechanism,        Pathology and Treatment.

          Tokyo: Yodosha, 1999. Pp. 10-21.

132. Vittet, D., Buchou, T., Schweizer, A.,               Dejana, E., and Huber, P.

                Targeted null-mutation in the vascular           endothelial-cadherin gene impairs the            organization of vascular-like structures in embryoid bodies.

          Proc. Natl. Acad. Sci. U.S.A. 94:

          6273, 1997

 

133. Tanaka, Y., Tajima, S., Tsutsumi, A., et al.

          New matrix flap prefabricated by     arteriovenous shunting and artificial              skin dermis in rats: II. Effect of         interpositional vein or artery grafts      and bFGF on new tissue generation.

          J. Jpn. Plast. Reconstr. Surg. 16:

          679, 1996.

134. Zhu C.,Ying D.,Zhou D.,Mi J.,

          Zhang W., Chang Q.,Li L.

                Expression of TGF-beta1 in smooth                muscle cells regulates endothelial progenitor               cells migration and differentiation.

                J Surg Res. 2005 May 15;125(2):151-6

135. Hudlicka, O., and Margaret, D. B. Physical     forces and angiogenesis.

          In G. M.Rubanyi (Ed.),       Mechanoreception             by the Vascular Wall.

                Mount Kisco, N.Y.: Futura,1993.

                Pp. 197-242.

136. Risau, W.

          Mechanisms of angiogenesis.

          Nature 386: 671, 1997.

137. Delaere P.,Vanderpoorten V.,

          Vranckx JJ., Hierner R.

          Laryngeal repair after resection of advanced                cancer: an optimal reconstructive protocol.

Eur Arch Otorhinolaryngol.262:910,2005.

138. Vranckx JJ.,Segers K.,Vertriest R.,    

          Vermeulen P.,Wever C.

          Tissue reconstruction after severe tissue      destruction by meningococcal sepis. A         story of glory and defeat.

          Submitted to JPRAS 2007.

139. Lantieri LA.,Martin-Garcia N.,

          Wechsler J., Mitrofanoff M.,

          Raulo Y.,Baruch JP.

          VEGF expression in expanded tissue : a         possible mechanism of angiogenesis in         tissue expansion.

          Plast.Reconstr.Surg.101;392-8,1998.

140. Mitchell SL., Niklason LE.

         Requirements for growing tissue engineered                vascular grafts.

         Cardiovasc.Pathol.2003; 12:59-64.

141. Kannan RY, Salacinski HJ, Sales K, Butler P, Seifalian AM.

         The roles of tissue engineering and                vascularisation in the development of            micro-vascular networks.

         Biomaterials. 2005;26:1857-75.

 

142. C. Kalka, H. Masuda, T. Takahashi,                 W.M. Kalka-Moll, M. Silver, M. Kearney,

         T. Li, J.M. Isner and T. Asahara,      Transplantation of ex vivo expanded              endothelial progenitor cells for therapeutic          neovascularization,

         Proc Natl Acad Sci USA 2000;

         97 :3422–3427.

143. Asahara T.,Murohata T.,Sullivan A.,               Silver M.,Vanderzee R.,Li T.,

         Witzenbichler B., Schatterman G.,

         Isner JM.

         Isolation of putative progenitor endothelial cells for angiogenesis.

         Science 1997; 275: 964-7.

144. Kaihara, J. Borenstein, R. Koka,

         S. Lalan, E.R. Ochoa, M. Ravens,

         H. Pien, B. Cunningham and J.P. Vacanti,

         Silicon micromachining to tissue engineer     branched vascular channels for       liver        fabrication,

                Tissue Eng 2000; 6: 105–117.

145. Moldovan NI.,Ferrari M.,

         Prospects for microtechnology and                nanotechnology in bioengineering of            replacement microvessels.

         Arch Pathol Lab Med 2002;

                126: 320–324.S.

146. Suh W.,Kim KL.,Kim JM.,Shin IS.,

         Lee YS., Jang HS.,Lee JS.,Byun J.,

         Choi JH.,Jeon ES.,Kim DK.                Transplantation of EPCs accelerates              dermal wound healing with increased          recruitment of monocytes/macrophages        and neovascularization.

                Stem Cells 23,1571,2005.

147. Yoshimura K.,Shigeura T.,

         Matsumoto D., Sato D.,Takaki Y.,

         Aiba-Kojima E.,Sato K.,Inoue K.,

         Nagase T., Koshima I.,Gonda K.      Characterization of freshly isolated and         cultured cells derived from the fatty and        fluid portions of liposuction aspirates.             J.Cell.Physiol.208,64,2006.

148. Medical rapid prototyping technologies:        state of the art and current limitations for      application in oral and maxillofacial surgery.

          J.Oral Maxillofac.Surg.63,1006,2005