Skip to main content

Abstract

Skin flaps are surgically elevated areas of skin and subcutaneous tissue transferred on their own vascular pedicle from a donor site to a recipient site that is devoid of skin. Skin flaps have been used in clinical reconstructive surgery for over 100 years and remain common procedures to this day. This chapter will describe numerous experimental animal skin flap models that have been developed over the last 40 years, and the various techniques applied to these models to increase their survival including vascular delay, ischemic preconditioning and flap prefabrication. Most of these techniques are now known to significantly increase the blood vessel numbers and blood flow to experimental skin flaps. Skin flap models and applied techniques illustrate various mechanisms by which angiogenesis can be stimulated and manipulated. Additionally, the manipulation of large vascular pedicles associated with flap prefabrication has led to a new and developing area in intrinsic vascularisation of tissue engineered chambers where new tissues and organs can be grown. Basic experimental flap models, techniques to enhance angiogenesis in these flaps and intrinsic vascularisation of tissue engineering chambers will be described in this chapter.

Geraldine M. Mitchell, Zerina Lokmic, and Shiba Sinha contributed equally to this chapter.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 39.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 54.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 54.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Abbreviations

AAS:

(Aminopropyl) triethoxy-silane

AVL:

Arterio-venous loop

CAST:

Computer assisted stereological toolbox

CT:

Computed tomography

CTA:

Computed tomographic angiography

DAB:

Chromogen diaminobenzidine

DPX:

Distyrene, plasticizer, xylene

EC:

Endothelial cell

ECM:

Extra-cellular matrix

EPC:

Endothelial precursor cells

FGF-2:

Fibroblast growth factor 2, or basic fibroblast growth factor

HIF:

Hypoxia-inducible transcription factor

HRP:

Horse radish peroxidase

IP:

Ischemic preconditioning

OPS:

Orthogonal polarisation spectral

PBS:

Phosphate buffered saline

PDGF:

Platelet derived growth factor

PPG:

Photoplethysmography

RISK:

Reperfusion injury salvage kinase

SIE:

Superficial inferior epigastric vessels

TUNEL:

Terminal deoxynucleotidyl transferase dUTP nick end labeling

TBS:

Tris buffered saline

W:

Weight

V:

Volume

VEGF:

Vascular endothelial growth factor

References

  1. Bhishgratna KKL (1907) Sustrata: an English translation of the Sushruta Samhita (based on original Sanskrit text). Bose, Calcutta

    Google Scholar 

  2. Laschke MW, Vollmar B, Menger MD (2009) Inosculation: connecting the life-sustaining pipelines. Tissue Eng B Rev 15:455–465

    Article  Google Scholar 

  3. Penington AJ (2010) Local flap reconstruction, 2nd edn. McGraw Hill, Sydney

    Google Scholar 

  4. Manchott C (1889) Die Hautarerien des menchlichen Korpers. Vogel, Leipzig

    Google Scholar 

  5. Salmon M (1936) Arteres de la Peau. Masson et Cie, Paris

    Google Scholar 

  6. Taylor GI, Palmer JH (1987) The vascular territories (angiosomes) of the body: experimental study and clinical applications. Br J Plast Surg 40:113–141

    Article  PubMed  CAS  Google Scholar 

  7. Saint-Cyr M, Wong C, Schaverien M et al (2009) The perforasome theory: vascular anatomy and clinical implications. Plast Reconstr Surg 124:1529–1544

    Article  PubMed  CAS  Google Scholar 

  8. Taylor GI, Corlett RJ, Dhar SC et al (2011) The anatomical (angiosome) and clinical territories of cutaneous perforating arteries: development of the concept and designing safe flaps. Plast Reconstr Surg 127:1447–1459

    Article  PubMed  CAS  Google Scholar 

  9. Ghali S, Butler PE, Tepper OM et al (2007) Vascular delay revisited. Plast Reconstr Surg 119:1735–1744

    Article  PubMed  CAS  Google Scholar 

  10. Milton SH (1969) The effects of “delay” on the survival of experimental pedicled skin flaps. Br J Plast Surg 22:244–252

    Article  PubMed  CAS  Google Scholar 

  11. Myers MB, Cherry G (1969) Mechanism of the delay phenomenon. Plast Reconstr Surg 44:52–57

    Article  PubMed  CAS  Google Scholar 

  12. Finseth F, Cutting C (1978) An experimental neurovascular island skin flap for the study of the delay phenomenon. Plast Reconstr Surg 61:412–420

    Article  PubMed  CAS  Google Scholar 

  13. Morris SF, Taylor GI (1995) The time sequence of the delay phenomenon: when is a surgical delay effective? An experimental study. Plast Reconstr Surg 95:526–533

    Article  PubMed  CAS  Google Scholar 

  14. Banbury J, Siemionow M, Porvasnik S et al (1999) Muscle flaps’ triphasic microcirculatory response to sympathectomy and denervation. Plast Reconstr Surg 104:730–737

    Article  PubMed  CAS  Google Scholar 

  15. Pearl RM (1981) A unifying theory of the delay phenomenon-recovery from the hyperadrenergic state. Ann Plast Surg 7:102–112

    Article  PubMed  CAS  Google Scholar 

  16. Holzbach T, Neshkova I, Vlaskou D et al (2009) Searching for the right timing of surgical delay: angiogenesis, vascular endothelial growth factor and perfusion changes in a skin-flap model. J Plast Reconstr Aesthet Surg 62:1534–1542

    Article  PubMed  Google Scholar 

  17. Barthe Garcia P, Suarez Nieto C, Rojo Ortega JM (1991) Morphological changes in the vascularization of delayed flaps in rabbits. Br J Plast Surg 44:285–290

    Article  PubMed  CAS  Google Scholar 

  18. Callegari PR, Taylor GI, Caddy CM et al (1992) An anatomic review of the delay phenomenon: I. Experimental studies. Plast Reconstr Surg 89:397–407

    Article  PubMed  CAS  Google Scholar 

  19. Murphy RC, Lawrence WT, Robson MC et al (1985) Surgical delay and arachidonic acid metabolites: evidence for an inflammatory mechanism: an experimental study in rats. Br J Plast Surg 38:272–277

    Article  PubMed  CAS  Google Scholar 

  20. Wong MS, Erdmann D, Sweis R et al (2004) Basic fibroblast growth factor expression following surgical delay of rat transverse rectus abdominis myocutaneous flaps. Plast Reconstr Surg 113:2030–2036

    Article  PubMed  Google Scholar 

  21. Lineaweaver WC, Lei MP, Mustain W et al (2004) Vascular endothelium growth factor, surgical delay, and skin flap survival. Ann Surg 239:866–873

    Article  PubMed  Google Scholar 

  22. Arranz López JL, Suárez Nieto C, Barthe García P et al (1995) Evaluation of angiogenesis in delayed skin flaps using a monoclonal antibody for the vascular endothelium. Br J Plast Surg 48:479–486

    Article  PubMed  Google Scholar 

  23. Tepper OM, Capla JM, Galiano RD et al (2005) Adult vasculogenesis occurs through in situ recruitment, proliferation, and tubulization of circulating bone marrow-derived cells. Blood 105:1068–1077

    Article  PubMed  CAS  Google Scholar 

  24. Asahara T, Murohara T, Sullivan A et al (1997) Isolation of putative progenitor endothelial cells for angiogenesis. Science 275:964–967

    Article  PubMed  CAS  Google Scholar 

  25. Kuntscher MV, Hartmann B, Germann G (2005) Remote ischemic preconditioning of flaps: a review. Microsurgery 25:346–352

    Article  PubMed  Google Scholar 

  26. Murry CE, Jennings RB, Reimer KA (1986) Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium. Circulation 74:1124–1136

    Article  PubMed  CAS  Google Scholar 

  27. Mounsey RA, Pang CY, Forrest C (1992) Preconditioning: a new technique for improved muscle flap survival. Otolaryngol Head Neck Surg 107:549–552

    PubMed  CAS  Google Scholar 

  28. Carroll CM, Carroll SM, Overgoor ML et al (1997) Acute ischemic preconditioning of skeletal muscle prior to flap elevation augments muscle-flap survival. Plast Reconstr Surg 100:58–65

    Article  PubMed  CAS  Google Scholar 

  29. Wang WZ, Anderson G, Firrell JC et al (1998) Ischemic preconditioning versus intermittent reperfusion to improve blood flow to a vascular isolated skeletal muscle flap of rats. J Trauma 45:953–959

    Article  PubMed  CAS  Google Scholar 

  30. Adanali G, Ozer K, Siemionow M (2002) Early and late effects of ischemic preconditioning on microcirculation of skeletal muscle flaps. Plast Reconstr Surg 109:1344–1351

    Article  PubMed  Google Scholar 

  31. Zahir KS, Syed SA, Zink JR et al (1998) Ischemic preconditioning improves the survival of skin and myocutaneous flaps in a rat model. Plast Reconstr Surg 102:140–150

    Article  PubMed  CAS  Google Scholar 

  32. Tatlidede S, McCormack MC, Eberlin KR et al (2009) A novel murine island skin flap for ischemic preconditioning. J Surg Res 154:112–117

    Article  PubMed  Google Scholar 

  33. Kuntscher MV, Schirmbeck EU, Menke H et al (2002) Ischemic preconditioning by brief extremity ischemia before flap ischemia in a rat model. Plast Reconstr Surg 109:2398–2404

    Article  PubMed  Google Scholar 

  34. Downey JM, Davis AM, Cohen MV (2007) Signaling pathways in ischemic preconditioning. Heart Fail Rev 12:181–188

    Article  PubMed  CAS  Google Scholar 

  35. Hausenloy DJ, Yellon DM (2007) Reperfusion injury salvage kinase signalling: taking a RISK for cardioprotection. Heart Fail Rev 12:217–234

    Article  PubMed  CAS  Google Scholar 

  36. Bolli R, Li QH, Tang XL et al (2007) The late phase of preconditioning and its natural clinical application–gene therapy. Heart Fail Rev 12:189–199

    Article  PubMed  CAS  Google Scholar 

  37. Semenza GL, Wang GL (1992) A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Mol Cell Biol 12:5447–5454

    PubMed  CAS  Google Scholar 

  38. Bernhardt WM, Warnecke C, Willam C et al (2007) Organ protection by hypoxia and hypoxia-inducible factors. Methods Enzymol 435:221–245

    PubMed  CAS  Google Scholar 

  39. Picard-Ami LA Jr, MacKay A, Kerrigan CL (1991) Pathophysiology of ischemic skin flaps: differences in xanthine oxidase levels among rats, pigs, and humans. Plast Reconstr Surg 87:750–755

    Article  PubMed  Google Scholar 

  40. Deune EG, Khouri RK (1995) Rat strain differences in flap tolerance to ischemia. Microsurgery 16:765–767

    Article  PubMed  CAS  Google Scholar 

  41. Erol OO, Spria M (1980) New capillary bed formation with a surgically constructed arteriovenous fistula. Plast Reconstr Surg 66:109–115

    Article  PubMed  CAS  Google Scholar 

  42. Shen ZY (1981) Vascular implantation into skin flap to form an axial pattern skin flap: experimental study (author’s transl). Zhonghua Wai Ke Za Zhi 19:692–695

    PubMed  CAS  Google Scholar 

  43. Khouri RK, Upton J, Shaw WW (1992) Principles of flap prefabrication. Clin Plast Surg 19:763–771

    PubMed  CAS  Google Scholar 

  44. Morrison WA, Penington AJ, Kumpta SK et al (1997) Clinical applications and technical limitations of prefabricated flaps. Plast Reconstr Surg 99:378–385

    Article  PubMed  CAS  Google Scholar 

  45. Pribaz JJ, Fine N, Orgill DP (1999) Flap prefabrication in the head and neck: a 10-year experience. Plast Reconstr Surg 103:808–820

    Article  PubMed  CAS  Google Scholar 

  46. Pribaz JJ, Weiss DD, Mulliken JB et al (1999) Prelaminated free flaps reconstruction of complex central facial defects. Plast Reconstr Surg 104:357–365

    Article  PubMed  CAS  Google Scholar 

  47. Tanaka Y, Tsutsumi A, Crowe DM et al (2000) Generation of an autologous tissue (matrix) flap by combining an arteriovenous shunt loop with artificial skin in rats: preliminary report. Br J Plast Surg 53:51–57

    Article  PubMed  CAS  Google Scholar 

  48. Mian R, Morrison WA, Hurley JV et al (2000) Formation of new tissue from an arteriovenous loop in the absence of added extracellular matrix. Tissue Eng 6:595–603

    Article  PubMed  CAS  Google Scholar 

  49. Tananka Y, Sung KC, Tsutsumi A et al (2003) Tissue engineering skin flaps: which vascular carrier, arteriovenous shunt loop or arteriovenous bundle, has more potential for angiogenesis and tissue generation? Plast Reconstr Surg 112:1636–1644

    Article  Google Scholar 

  50. Tanaka Y, Sung KC, Fumimoto M et al (2006) Prefabricated engineered skin flap using an arteriovenous vascular bundle as a vascular carrier in rabbits. Plast Reconstr Surg 117:1860–1875

    Article  PubMed  CAS  Google Scholar 

  51. Lokmic Z, Stillaert F, Morrison WA et al (2007) An arteriovenous loop in a protected space generates a permanent, highly vascular, tissue-engineered construct. FASEB J 21:511–522

    Article  PubMed  CAS  Google Scholar 

  52. Tilkorn DJ, Bedogni A, Keramidaris E et al (2010) Implanted myoblast survival is dependent on the degree of vascularization in a novel delayed implantation/prevascularization tissue engineering model. Tissue Eng A 16:165–178

    Article  CAS  Google Scholar 

  53. German W, Finesilver EM, Davis JS (1933) Establishment of circulation in tubed skin flaps. Arch Surg 26:27

    Article  Google Scholar 

  54. Myers MB, Cherry G (1971) Differences in the delay phenomenon in the rabbit, rat, and pig. Plast Reconstr Surg 47:73–78

    Article  PubMed  CAS  Google Scholar 

  55. Donovan WE (1975) Experimental models in skin flap research. In: Grabb WC, Myers MB (eds) Skin flaps, 1st edn. Little Brown and Company, Boston

    Google Scholar 

  56. McFarlane RM, Deyoung G, Henry RA (1965) The design of a pedicle flap in the rat to study necrosis and its prevention. Plast Reconstr Surg 35:177–182

    Article  PubMed  CAS  Google Scholar 

  57. Adamson JE, Horton CE, Crawford HH et al (1966) The effects of dimethyl sulfoxide on the experimental pedicle flap: a preliminary report. Plast Reconstr Surg 37:105–110

    PubMed  CAS  Google Scholar 

  58. Thompson FM, Berakha GJ, Guthrie RH Jr (1977) The effective duration of the delay phenomenon in the rat. Plast Reconstr Surg 60:384–389

    PubMed  CAS  Google Scholar 

  59. Conoyer JM, Toomey JM (1979) Dorsal skin flaps in rats as an experimental model. Surg Forum 30:510–511

    PubMed  CAS  Google Scholar 

  60. Knox LK, Stewart AG, Hayward PG et al (1994) Nitric oxide synthase inhibitors improve skin flap survival in the rat. Microsurgery 15:708–711

    Article  PubMed  CAS  Google Scholar 

  61. Pang Y, Lineaweaver WC, Lei MP et al (2003) Evaluation of the mechanism of vascular endothelial growth factor improvement of ischemic flap survival in rats. Plast Reconstr Surg 112:556–564

    Article  PubMed  Google Scholar 

  62. Aydogan H, Gurlek A, Parlakpinar H et al (2007) Beneficial effects of caffeic acid phenethyl ester (CAPE) on the ischaemia-reperfusion injury in rat skin flaps. J Plast Reconstr Aesthet Surg 60:563–568

    Article  PubMed  Google Scholar 

  63. Angel MF, Kaufman T, Swartz WM et al (1986) Studies on the nature of the flap/bed interaction in rodents–Part I: flap survival under varying conditions. Ann Plast Surg 17:317–322

    Article  PubMed  CAS  Google Scholar 

  64. Hammond DC, Brooksher RD, Mann RJ et al (1993) The dorsal skin-flap model in the rat: factors influencing survival. Plast Reconstr Surg 91:316–321

    Article  PubMed  CAS  Google Scholar 

  65. Kaufman T, Angel MF, Eichenlaub EH et al (1985) The salutary effects of the bed on the survival of experimental flaps. Ann Plast Surg 14:64–73

    Article  PubMed  CAS  Google Scholar 

  66. Jones M, Zhang F, Blain B et al (2001) Influence of recipient-bed isolation on survival rates of skin-flap transfer in rats. J Reconstr Microsurg 17:653–658

    Article  PubMed  CAS  Google Scholar 

  67. Kelly CP, Gupta A, Keskin M et al (2010) A new design of a dorsal flap in the rat to study skin necrosis and its prevention. J Plast Reconstr Aesthet Surg 63:1553–1556

    Article  PubMed  Google Scholar 

  68. Yang D, Morris SF (1999) An extended dorsal island skin flap with multiple vascular territories in the rat: a new skin flap model. J Surg Res 87:164–170

    Article  PubMed  CAS  Google Scholar 

  69. Syed SA, Tasaki Y, Fujii T et al (1992) A new experimental model: the vascular pedicle cutaneous flap over the dorsal aspect (flank and hip) of the rat. Br J Plast Surg 45:23–25

    Article  PubMed  CAS  Google Scholar 

  70. Syed SA, Restifo RJ (1997) An economical axial-pattern flap necrosis model. Plast Reconstr Surg 99:263–264

    Article  PubMed  CAS  Google Scholar 

  71. Kayikcioglu A, Akyurek M, Safak T (1998) The importance of vascular territories in designing new experimental skin flaps. Plast Reconstr Surg 101:1155–1157

    PubMed  CAS  Google Scholar 

  72. Hosnuter M, Kargi E, Peksoy I et al (2006) An ameliorated skin flap model in rats for experimental research. J Plast Reconstr Aesthet Surg 59:299–303

    Article  PubMed  Google Scholar 

  73. Ohara H, Kishi K, Nakajima T (2008) Rat dorsal paired island skin flaps: a precise model for flap survival evaluation. Keio J Med 57:211–216

    Article  PubMed  Google Scholar 

  74. Taylor GI, Corlett RJ, Caddy CM et al (1992) An anatomic review of the delay phenomenon: II. Clinical applications. Plast Reconstr Surg 89:408–416

    Article  PubMed  CAS  Google Scholar 

  75. Ahmed SS, Pierce J, Reid M et al (1997) A new experimental model: the vascular pedicled cutaneous flap over the mid-dorsum of the rat. Ann Plast Surg 39:495–499

    Article  PubMed  CAS  Google Scholar 

  76. Adanali G, Seyhan T, Turegun M et al (2002) Effects of different vascular patterns and the delay phenomenon on rat ventral island flap viability. Ann Plast Surg 48:660–664

    Article  PubMed  Google Scholar 

  77. Goldwyn RM, Lamb DL, White WL (1963) An experimental study of large island flaps in dogs. Plast Reconstr Surg 31:528–536

    Article  PubMed  CAS  Google Scholar 

  78. Strauch B, Murray DE (1967) Transfer of composite graft with immediate suture anastomosis of its vascular pedicle measuring less than 1 mm in external diameter using microsurgical techniques. Plast Reconstr Surg 40:325–329

    Article  PubMed  CAS  Google Scholar 

  79. Angel MF, Mellow CG, Knight KR et al (1989) The effect of time of vascular island skin flap elevation on tolerance to warm ischemia. Ann Plast Surg 22:426–428

    Article  PubMed  CAS  Google Scholar 

  80. Angel MF, Mellow CG, Knight KR et al (1989) Prior elevation of vascular island skin flaps: intolerance to ischemia caused by venous obstruction. J Reconstr Microsurg 5:163–165

    Article  PubMed  CAS  Google Scholar 

  81. Roberts AP, Cohen JI, Cook TA (1996) The rat ventral island flap: a comparison of the effects of reduction in arterial inflow and venous outflow. Plast Reconstr Surg 97:610–615

    Article  PubMed  CAS  Google Scholar 

  82. Angel MF, Mellow CG, Knight KR et al (1990) Secondary ischemia time in rodents: contrasting complete pedicle interruption with venous obstruction. Plast Reconstr Surg 85:789–793

    Article  PubMed  CAS  Google Scholar 

  83. Sagi A, Ferder M, Levens D et al (1986) Improved survival of island flaps after prolonged ischemia by perfusion with superoxide dismutase. Plast Reconstr Surg 77:639–644

    Article  PubMed  CAS  Google Scholar 

  84. Mellow CG, Knight KR, Angel MF et al (1990) The effect of thromboxane synthetase inhibition on tolerance of skin flaps to secondary ischemia caused by venous obstruction. Plast Reconstr Surg 86:329–334

    Article  PubMed  CAS  Google Scholar 

  85. Willemart G, Knight KR, Morrison WA (1998) Dexamethasone treatment prior to reperfusion improves the survival of skin flaps subjected to secondary venous ischaemia. Br J Plast Surg 51:624–628

    Article  PubMed  CAS  Google Scholar 

  86. Ueda K, Nozawa M, Nakao M et al (2000) Sulfatide and monoclonal antibodies prevent reperfusion injury in skin flaps. J Surg Res 88:125–129

    Article  PubMed  CAS  Google Scholar 

  87. Machens HG, Morgan JR, Berthiaume F et al (2002) Platelet-derived growth factor-AA-mediated functional angiogenesis in the rat epigastric island flap after genetic modification of fibroblasts is ischemia dependent. Surgery 131:393–400

    Article  PubMed  Google Scholar 

  88. Gideroglu K, Yilmaz F, Aksoy F et al (2009) Montelukast protects axial pattern rat skin flaps against ischemia/reperfusion injury. J Surg Res 157:181–186

    Article  PubMed  CAS  Google Scholar 

  89. Brown DM, Hong SP, Farrell CL et al (1995) Platelet-derived growth factor BB induces functional vascular anastomoses in vivo. Proc Natl Acad Sci USA 92:5920–5924

    Article  PubMed  CAS  Google Scholar 

  90. Theile DR, Kane AJ, Romeo R et al (1998) A model of bridging angiogenesis in the rat. Br J Plast Surg 51:243–249

    Article  PubMed  CAS  Google Scholar 

  91. Kane AJ, Barker JE, Mitchell GM et al (2001) Inducible nitric oxide synthase (iNOS) activity promotes ischaemic skin flap survival. Br J Pharmacol 132:1631–1638

    Article  PubMed  CAS  Google Scholar 

  92. Furuta S, Vadiveloo P, Romeo-Meeuw R et al (2004) Early inducible nitric oxide synthase 2 (NOS 2) activity enhances ischaemic skin flap survival. Angiogenesis 7:33–43

    Article  PubMed  CAS  Google Scholar 

  93. Serafin D, Shearin JC, Georgiade NG (1977) The vascularization of free flaps: a clinical and experimental correlation. Plast Reconstr Surg 60:233–241

    Article  PubMed  CAS  Google Scholar 

  94. Nakajima T (1978) How soon do venous drainage channels develop at the periphery of a free flap? A study in rats. Br J Plast Surg 31:300–308

    PubMed  CAS  Google Scholar 

  95. Nishikawa H, Manek S, Green CJ (1991) The oblique rat groin flap. Br J Plast Surg 44:295–298

    Article  PubMed  CAS  Google Scholar 

  96. Hirase Y, Valauri FA, Buncke HJ (1989) Creation of neovascularised free flaps using vein grafts as pedicles: a preliminary report on experimental models. Br J Plast Surg 42:216–222

    Article  PubMed  CAS  Google Scholar 

  97. Falco NA, Pribaz JJ, Eriksson E (1992) Vascularization of skin following implantation of an arteriovenous pedicle: implications in flap prefabrication. Microsurgery 13:249–254

    Article  PubMed  CAS  Google Scholar 

  98. Morrison WA, Dvir E, Doi K et al (1990) Prefabrication of thin transferable axial-pattern skin flaps: an experimental study in rabbits. Br J Plast Surg 43:645–654

    Article  PubMed  CAS  Google Scholar 

  99. Wilson YT, Kumpta S, Hickey MJ et al (1994) Use of free interpositional vein grafts as pedicles for prefabrication of skin flaps. Microsurgery 15:717–721

    Article  PubMed  CAS  Google Scholar 

  100. Hickey MJ, Wilson Y, Hurley JV et al (1998) Mode of vascularization of control and basic fibroblast growth factor-stimulated prefabricated skin flaps. Plast Reconstr Surg 101:1296–1304

    Article  PubMed  CAS  Google Scholar 

  101. Khouri RK, Hong SP, Deune EG et al (1994) De novo generation of permanent neovascularized soft tissue appendages by platelet-derived growth factor. J Clin Invest 94:1757–1763

    Article  PubMed  CAS  Google Scholar 

  102. Tanaka Y, Tajima S, Tsutsumi A et al (1996) New matrix flap prefabricated by arteriovenous shunting and artificial skin dermis in rats II. Effect of interpositional vein and artery grafts and bFGF on new tissue generation. J Jpn Plast Reconstr Surg 16:679

    Google Scholar 

  103. Takato T, Zuker RM, Turley CB (1991) Prefabrication of skin flaps using vein grafts: an experimental study in rabbits. Br J Plast Surg 44:593–598

    Article  PubMed  CAS  Google Scholar 

  104. Maitz PK, Pribaz JJ, Hergruter CA (1994) Manipulating prefabricated flaps: an experimental study examining flap viability. Microsurgery 15:624–629

    Article  PubMed  CAS  Google Scholar 

  105. Myers MB, Brock D, Cohn I Jr (1971) Prevention of skin slough after radical mastectomy by the use of a vital dye to delineate devascularized skin. Ann Surg 173:920–924

    Article  PubMed  CAS  Google Scholar 

  106. Teich-Alasia S (1971) A study of the vascularisation of pedicle flaps using disulphine blue. Br J Plast Surg 24:282–288

    Article  PubMed  CAS  Google Scholar 

  107. Eren S, Rubben A, Krein R et al (1995) Assessment of microcirculation of an axial skin flap using indocyanine green fluorescence angiography. Plast Reconstr Surg 96:1636–1649

    Article  PubMed  CAS  Google Scholar 

  108. Lamby PL, Gais S et al (2008) Evaluation of the vascular integrity of free flaps based on microcirculation imaging techniques. Clin Hemorheol Microcirc 39:253–263

    PubMed  CAS  Google Scholar 

  109. Ezaki T, Baluk P, Thurston G et al (2001) Time course of endothelial cell proliferation and microvascular remodeling in chronic inflammation. Am J Pathol 158:2043–2055

    Article  PubMed  CAS  Google Scholar 

  110. Jiao C, Bronner S, Mercer KL et al (2004) Epidermal cells accelerate the restoration of the blood flow in diabetic ischemic limbs. J Cell Sci 117:1055–1063

    Article  PubMed  CAS  Google Scholar 

  111. Li Y, Song Y, Zhao L et al (2008) Direct labeling and visualization of blood vessels with lipophilic carbocyanine dye DiI. Nat Protoc 3:1703–1708

    Article  PubMed  CAS  Google Scholar 

  112. Bergeron L, Tang M, Morris SF (2006) A review of vascular injection techniques for the study of perforator flaps. Plast Reconstr Surg 117:2050–2057

    Article  PubMed  CAS  Google Scholar 

  113. Lindenblatt N, Calcagni M, Contaldo C et al (2008) A new model for studying the revascularization of skin grafts in vivo: the role of angiogenesis. Plast Reconstr Surg 122:1669–1680

    Article  PubMed  CAS  Google Scholar 

  114. Lokmic Z, Mitchell GM (2011) Visualisation and stereological assessment of blood and lymphatic vessels. Histol Histopathol 26:781–796

    PubMed  Google Scholar 

  115. Proano E, Perbeck L (1993) Changes in skin blood flow in ischaemic limbs after vascular reconstruction measured by fluorescein flowmetry and laser Doppler flowmetry. Clin Physiol 13:599–609

    Article  PubMed  CAS  Google Scholar 

  116. Proano E, Perbeck L (1994) Effect of exposure to heat and intake of ethanol on the skin circulation and temperature in ischaemic limbs. Clin Physiol 14:305–310

    Article  PubMed  CAS  Google Scholar 

  117. Proano E, Svensson L, Perbeck L (1997) Correlation between the uptake of sodium fluorescein in the tissue and xenon-133 clearance and laser Doppler fluxmetry in measuring changes in skin circulation. Int J Microcirc Clin Exp 17:22–28

    Article  PubMed  CAS  Google Scholar 

  118. Saetzler RK, Jallo J, Lehr HA et al (1997) Intravital fluorescence microscopy: impact of light-induced phototoxicity on adhesion of fluorescently labeled leukocytes. J Histochem Cytochem 45:505–513

    Article  PubMed  CAS  Google Scholar 

  119. Sloan GM, Sasaki GH (1985) Noninvasive monitoring of tissue viability. Clin Plast Surg 12:185–195

    PubMed  CAS  Google Scholar 

  120. Chubb D, Rozen WM, Ashton MW (2010) Early survival of a compromised fasciocutaneous flap without pedicle revision: monitoring with Photoplethysmography. Microsurgery 30:462–465

    Article  PubMed  Google Scholar 

  121. Groner W, Winkelman JW, Harris AG et al (1999) Orthogonal polarization spectral imaging: a new method for study of the microcirculation. Nat Med 5:1209–1212

    Article  PubMed  CAS  Google Scholar 

  122. Langer S, Biberthaler P, Harris AG et al (2001) In vivo monitoring of microvessels in skin flaps: introduction of a novel technique. Microsurgery 21:317–324

    Article  PubMed  CAS  Google Scholar 

  123. Olivier WA, Hazen A, Levine JP et al (2003) Reliable assessment of skin flap viability using orthogonal polarization imaging. Plast Reconstr Surg 112:547–555

    Article  PubMed  Google Scholar 

  124. Tokiya R, Umetani K, Imai S et al (2004) Observation of microvasculatures in athymic nude rat transplanted tumor using synchrotron radiation microangiography system. Acad Radiol 11:1039–1046

    Article  PubMed  Google Scholar 

  125. Ichioka S, Minh TC, Shibata M et al (2002) In vivo model for visualizing flap microcirculation of ischemia-reperfusion. Microsurgery 22:304–310

    Article  PubMed  Google Scholar 

  126. Bellhorn MB, Bellhorn RW, Poll DS (1977) Permeability of fluorescein-labelled dextrans in fundus fluorescein angiography of rats and birds. Exp Eye Res 24:595–605

    Article  PubMed  CAS  Google Scholar 

  127. Smith LE, Wesolowski E, McLellan A et al (1994) Oxygen-induced retinopathy in the mouse. Invest Ophthalmol Vis Sci 35:101–111

    PubMed  CAS  Google Scholar 

  128. Kurozumi K, Hardcastle J, Thakur R et al (2007) Effect of tumor microenvironment modulation on the efficacy of oncolytic virus therapy. J Natl Cancer Inst 99:1768–1781

    Article  PubMed  CAS  Google Scholar 

  129. Bonner-Weir S, Orci L (1982) New perspectives on the microvasculature of the islets of Langerhans in the rat. Diabetes 31:883–889

    Article  PubMed  CAS  Google Scholar 

  130. Wagner RC, Czymmek K, Hossler FE (2006) Confocal microscopy, computer modeling and quantification of glomerular vascular corrosion casts. Microsc Microanal 12:262–268

    Article  PubMed  CAS  Google Scholar 

  131. Erol OO, Spira M (1979) New capillary bed formation with a surgically constructed arteriovenous fistula. Surg Forum 30:530–531

    PubMed  CAS  Google Scholar 

  132. Lokmic Z, Mitchell GM (2008) Engineering the microcirculation. Tissuse Eng B 14:87–103

    Article  CAS  Google Scholar 

  133. Cronin KJ, Messina A, Knight KR et al (2004) New murine model of spontaneous autologous tissue engineering, combining an arteriovenous pedicle with matrix materials. Plast Reconstr Surg 113:260–269

    Article  PubMed  Google Scholar 

  134. Lepore DA, Thomas GP, Knight KR et al (2007) Survival and differentiation of pituitary colony-forming cells in vivo. Stem Cells 25:1730–1736

    Article  PubMed  CAS  Google Scholar 

  135. Seach N, Mattesich M, Abberton K et al (2010) Vascularized tissue engineering mouse chamber model supports thymopoiesis of ectopic thymus tissue grafts. Tissue Eng C 16:543–551

    Article  CAS  Google Scholar 

  136. Hussey AJ, Winardi M, Han XL et al (2009) Seeding of pancreatic islets into prevascularized tissue engineering chambers. Tissue Eng A 15:3823–3833

    Article  CAS  Google Scholar 

  137. Hussey AJ, Winardi M, Wilson J et al (2010) Pancreatic islet transplantation using vascularised chambers containing nerve growth factor ameliorates hyperglycaemia in diabetic mice. Cells Tissues Organs 191:382–393

    Article  PubMed  CAS  Google Scholar 

  138. Forster NA, Penington AJ, Hardikar AA et al (2011) A prevascularized tissue engineering chamber supports growth and function of islets and progenitor cells in diabetic mice. Islets 3:271–283

    Article  PubMed  Google Scholar 

  139. Forster N, Palmer JA, Yeoh G et al (2011) Expansion and hepatocytic differentiation of liver progenitor cells in vivo using a vascularized tissue engineering chamber in mice. Tissue Eng C Methods 17:359–366

    Article  CAS  Google Scholar 

  140. Kelly JL, Findlay MW, Knight KR et al (2006) Contact with existing adipose tissue is inductive for adipogenesis in Matrigel. Tissue Eng 12:2041–2047

    Article  PubMed  CAS  Google Scholar 

  141. Rophael JA, Craft RO, Palmer JA et al (2007) Angiogenic growth factor synergism in a murine tissue engineering model of angiogenesis and adipogenesis. Am J Pathol 171:2048–2057

    Article  PubMed  CAS  Google Scholar 

  142. Vashi AV, Abberton KM, Thomas GP et al (2006) Adipose tissue engineering based on the controlled release of fibroblast growth factor-2 in a collagen matrix. Tissue Eng 12:3035–3043

    Article  PubMed  CAS  Google Scholar 

  143. Findlay MW, Messina A, Thompson EW et al (2009) Long-term persistence of tissue-engineered adipose flaps in a murine model to 1 year: an update. Plast Reconstr Surg 124:1077–1084

    Article  PubMed  CAS  Google Scholar 

  144. Simcock JW, Penington AJ, Morrison WA et al (2009) Endothelial precursor cells home to a vascularised tissue engineering chamber by application of the angiogenic chemokine CXCL12. Tissue Eng A 15:655–664

    Article  CAS  Google Scholar 

  145. Cassell OC, Morrison WA, Messina A et al (2001) The influence of extracellular matrix on the generation of vascularized, engineered, transplantable tissue. Ann N Y Acad Sci 944:429–442

    Article  PubMed  CAS  Google Scholar 

  146. Hofer SO, Knight KM, Cooper-White JJ et al (2003) Increasing the volume of vascularized tissue formation in engineered constructs: an experimental study in rats. Plast Reconstr Surg 111:1186–1192

    Article  PubMed  Google Scholar 

  147. Cao Y, Mitchell G, Messina A et al (2006) The influence of architecture on degradation and tissue in growth into three-dimensional poly(lactic-co-glycolic acid) scaffolds in vitro and in vivo. Biomaterials 27:2854–2864

    Article  PubMed  CAS  Google Scholar 

  148. Dolderer JH, Thompson EW, Slavin J et al (2011) Long-term stability of adipose tissue generated from a vascularized pedicled fat flap inside a chamber. Plast Reconstr Surg 127:2283–2292

    Article  PubMed  CAS  Google Scholar 

  149. Morritt AN, Bortolotto SK, Dilley RJ et al (2007) Cardiac tissue engineering in an in vivo vascularized chamber. Circulation 115:353–360

    Article  PubMed  Google Scholar 

  150. Choi YS, Matsuda K, Dusting GJ et al (2010) Engineering cardiac tissue in vivo from human adipose-derived stem cells. Biomaterials 31:2236–2242

    Article  PubMed  CAS  Google Scholar 

  151. Messina A, Bortolotto SK, Cassell OC et al (2005) Generation of a vascularized organoid using skeletal muscle as the inductive source. FASEB J 19:1570–1572

    PubMed  CAS  Google Scholar 

  152. Scherle W (1970) A simple method for volumetry of organs in quantitative stereology. Mikroskopie 26:57–60

    PubMed  CAS  Google Scholar 

  153. Hsu SM, Raine L, Fanger H (1981) Use of avidin-biotin-peroxidase complex (ABC) in immunoperoxidase techniques: a comparison between ABC and unlabeled antibody (PAP) procedures. J Histochem Cytochem 29:577–580

    Article  PubMed  CAS  Google Scholar 

  154. Howard CV, Reed MG (2005) Unbiased stereology. BIOS Scientific Publishers, Oxford

    Google Scholar 

  155. Lokmic Z, Thomas JL, Morrison WA et al (2008) An endogenously deposited fibrin scaffold determines construct size in the surgically created arteriovenous loop chamber model of tissue engineering. J Vasc Surg 48:974–985

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

The authors appreciate the technical assistance of Ms Sue Mc Kay, Ms Liliana Pepe and Ms Anna Deftereos in the Experimental Medical and Surgical Unit at St Vincent’s Hospital, Melbourne, We also thank Jason Palmer (O’Brien Institute) for his technical advice.

The O’Brien Institute acknowledges the Victorian State Government’s Department of Innovation, Industry and Regional Development’s Operational Infrastructure Support Program.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Geraldine M. Mitchell Ph.D. .

Editor information

Editors and Affiliations

Appendices

Appendix

Methods for Rat AVL Construct and Mouse Flow Through Pedicle Construct and Skin Flap Tissue Processing, Visualization and Morphometric Assessment of Blood Vessels

2.1 Rat AVL Construct or Mouse Flow Through Pedicle Construct Weight Measurements

  1. 1.

    Place an empty 50 ml container on a four decimal balance (Mettler AE 260, DeltaRange®, Mettler-Toledo, Switzerland) and record the initial weight (W initial ).

  2. 2.

    Blot each construct (rat or mouse) carefully on Whatman filter paper then place the construct in the container on the balance and record the resulting weight (W final ).

  3. 3.

    Determine the weight of the AVL construct or mouse chamber construct (W construct ) by subtracting the initial weight from the final weight recorded: W construct = W final − W initial .

2.2 Rat AVL Construct or Mouse Flow Through Pedicle Construct Volume Measurements

AVL or mouse construct volume is determined by a method described by Scherle in 1970 [152] in which the water displacement due to construct volume is determined by weighing in saline. To weigh the newly formed AVL construct

  1. 1.

    Tie off the AVL construct artery and vein or mouse chamber construct artery and vein at the point of entry into the chamber (approximately) with a long piece of 6.0 silk thread at harvest.

  2. 2.

    Fill a 50 ml plastic container with 40 ml of isotonic sterile saline (0.9% NaCl) solution, place on a balance and record the initial weight (W 1 ).

  3. 3.

    Blot the AVL construct or mouse chamber construct on Whatman paper to remove excess fluids from the construct.

  4. 4.

    Attach the AVL or mouse flow through pedicle construct to a silk thread and lower the specimen suspended on the thread into the saline solution so that it is beneath the surface of the liquid but not touching the bottom or the sides of the container.

  5. 5.

    Record the resulting weight (W 2 ).

  6. 6.

    To calculate the weight of the fluid displaced by the organ (W O ), use the following formula: W O = W 2 -W 1

  7. 7.

    To determine the construct volume (V 0 ) divide the weight of the fluid displaced (W 0 ) by the AVL construct or mouse construct by the specific gravity of isotonic saline (g = 1.0048): V 0 = W 0 /g. As the specific gravity of saline is rounded off to g = 1.0, then: V 0 ≈ W 0 = W 2 − W 1 .

2.3 Slicing the AVL Construct Prior to Tissue Processing

Once weight and volume measurements are complete, the AVL construct should be sliced into 1 mm thick slices. However, if the construct is fragile, usually in the early stages of development (0–14 day construct), it should be fixed for 2 h in the desired fixative (10% neutral buffered formol saline or 4% paraformaldehyde) before attempting to slice. This will ensure that the construct structure does not disintegrate prematurely and therefore affect accurate histomorphometry.

The direction of slicing should be from the top of the construct, adjacent to the chamber lid, to the bottom of the construct, adjacent to the chamber base and from the femoral vessels point of entry into the construct towards the edge of the construct using a razor blade. Slice thickness should be as close as possible to 1–2 mm. This will ensure that cross sections of the AVL are obtained in these slices. After slicing, fixation should take place immediately for approximately 2 days prior to paraffin embedding. If paraffin embedding is delayed tissue should be stored in phosphate buffered saline (PBS) prior to processing. [If immediate fixation prior to slicing is not undertaken, some tissue slices can be removed and placed in liquid nitrogen for subsequent extraction of messenger RNA and total protein prior to fixation of the remaining slices].

2.4 Slicing of Skin Flaps

For histology and assessment of angiogenesis skin flaps should be either pinned out flat and fixed [or chemically cleared of all tissues and prepared as a whole mount – see [100]]. Skin flaps that are fixed must be sliced after fixation, at about 4–6 h after the fixative is applied. Flaps should then be cut ‘vertically” using a razor blade or scalpel from the epidermis to the fascia, at a slice thickness of 2–4 mm. The skin flap slices are returned to fixative for a minimum of 24 h, and then stored in phosphate buffered saline (PBS) overnight prior to histological processing (see below).

2.5 Tissue processing and sectioning

(applies to the AVL construct, mouse flow through pedicle construct and skin flap slices)

The following protocols have been determined by our group to provide optimal conditions that permit clear and consistent tissue staining and successful immunohistochemistry with a strong signal to noise ratio.

  1. 1.

    Post fixation, tissue slices are placed in a tissue processing cassette and stabilized in the cassette with biopsy pads.

  2. 2.

    The processing of tissue into paraffin blocks is done using a tissue embedding processor (HyperCenter XP, Shandon®, Shandon Scientific Ltd, England), where the tissue is dehydrated through graded serial changes of alcohol and embedded into paraffin wax as follows:

    1. i.

      70% Ethanol – 20 min

    2. ii.

      90% Ethanol – 20 min

    3. iii.

      95% Ethanol – 20 min

    4. iv.

      100% Ethanol – 20 min

    5. v.

      100% Ethanol – 30 min

    6. vi.

      100% Ethanol −60 min

    7. vii.

      Histolene – 20 min

    8. viii.

      Histolene – 20 min

    9. ix.

      Histolene – 30 min

    10. x.

      Paraffin wax – 20 min under vacuum

    11. xi.

      paraffin wax – 60 min under vacuum

  3. 3.

    Following the last step in wax, the tissue is transferred to an embedding stage and embedded into tissue moulds containing melted paraffin wax. Rat chamber slices are placed cut surface down on the base of the mould, all the slices from one construct in one mould, or if large, several slices from a construct are placed in one mould. Skin flap slices are also placed cut surface down on the base of the mould, again several slices may fit into one mould. (Mouse chamber constructs can be left whole, or cut into two halves at this point, either by a single cross section or longitudinal section cut in the mid line, and the two halves embedded cut surface down on the base of the embedding mould).

  4. 4.

    The moulds are then cooled down to 5°C to allow paraffin wax solidification.

  5. 5.

    Section paraffin blocks into 3–5 μm thick serial sections using a microtome (Reichert-Jung, Grale Scientific Australia), float on 3′ (Aminopropyl) triethoxy-silane (AAS) (Sigma®, Sigma-Aldrich, St Louis, USA) coated slides for histology or poly-L-lysine coated slides (Polysine™, Menzel-Glaser®, Germany) for immunohistochemistry and dry overnight in a 37°C oven.

2.6 Identification of Blood Vessels in Rat and Mouse Tissue Sections

To identify endothelial lined blood vessels in rat tissue, a two-step indirect immunoenzymatic method [153] is used to detect B. Simplicifolia lectin by a streptavidin biotin-based immunohistochemical protocol described below. Negative and positive control tissues were included in all immunohistochemical assessments. The negative control consisted of omitting B. Simplicifolia lectin from the reaction. The positive control tissues are rat skin tissues which label with this lectin on blood vessel endothelium. There is no absolutely definitive endothelial marker for rat tissue.

[It should be noted that a number of endothelial labels could be used, none of which are without some problems. The B. Simplicifolia lectin method described here is reliable and labels endothelial cells clearly – however it also labels macrophages and the observer needs also to be able to clearly distinguish small blood vessels from macrophages on morphological grounds. Another antibody which works very well in most rat tissues and labels endothelial cells very clearly is Factor VIII. However in the AVL chamber construct it will also label the fibrin matrix which persists for at least the first 2 weeks. This factor makes blood vessel identification very difficult and it is nearly impossible for the inexperienced to know what is true blood vessel labeling and what is not. We therefore have not included the Factor VIII protocol here. For mouse tissues we have generally used a CD31 antibody. This is a very clear label of endothelial cells, it does not label any inflammatory cells, but it is not specific to vascular endothelial cells as it also labels lymphatic endothelial cells.]

2.6.1 Lectin Iummunolabelling Protocol for Rat Tissue Blood Vessels

  1. 1.

    Dewax 3 μm thick serial tissue sections in histolene, graded ethanol (starting at 100% to 50%) and bring to water.

  2. 2.

    Wash sections in PBS, circle each tissue section with a wax pen. Apply Proteinase K for 5–8 min at room temperature, then wash the sections in three changes of PBS.

  3. 3.

    To block endogenous peroxidise and reduce the background that could be associated with the use of chromogen diaminobenzidine (DAB), blot the sections gently with tissues and treat the sections in 10% hydrogen peroxide (H2O2) in 50% methanol for 5 min. Wash sections in three changes of PBS.

  4. 4.

    Apply biotinylated B. Simplicifolia lectin solution (dilution 1:100) for 30 min at room temperature. Incubate the slides in a moist chamber to prevent drying out of the lectin solution. Wash sections in tris buffered saline (TBS) buffer.

  5. 5.

    Apply Streptavidin-HRP for 30 min at room temperature, also in a moist chamber, then wash sections in three changes of 0.05% Tween/TBS buffer.

  6. 6.

    To detect the bound lectin-streptavidin complex, use a chromogen such as DAB for 1–5 min. The sections are then washed in tap water, counterstained in Myer’s haematoxylin, dehydrated and cover-slipped with DPX mountant.

2.6.2 CD 31 Immunolabelling Protocol for Mouse Tissue Blood Vessels

  1. 1.

    For CD31 immunolabelling of mouse tissues a rat anti-mouse CD31 antibody is used (BD Pharmingen, San Jose, CA).

  2. 2.

    Paraffin sections are dewaxed and hydrated and have a TBS wash, followed by a peroxidase quench (3% H2O2 in 50%methanol/distilled water for 5 min) and another TBS wash.

  3. 3.

    Sections are digested with DAKO proteinase K that is warmed to room temperature (antigen retrieval) for 8 min, washed in TBS, and nonspecific binding blocked using DAKO protein block for 30 min. Blot excess fluid.

  4. 4.

    The primary antibody is applied at 1:100 in DAKO antibody diluent for 1 h, followed by a TBS wash.

  5. 5.

    The secondary antibody (DAKO rabbit anti-rat biotinylated immunoglobulin) is then applied at 1:300 for 30 min followed by a TBS wash.

  6. 6.

    An avidin-biotin-horseradish peroxidase (HRP) detection system (ABC Elite; Vector Laboratories, Burlingame, CA) is then used followed by a TBS wash.

  7. 7.

    Finally diaminobenzidine (DAB) color development for 5 min, is followed by a distilled water wash, and counterstaining with haematoxylin for 1 min.

  8. 8.

    Dehydrate sections in increasing concentrations of alcohol, clear and coverslip with DPX mountant.

2.7 Morphometric Analysis of Labelled Blood Vessels

To morphometrically assess the vessel development in the rat AVL model, mouse flow through pedicle model or skin flaps, it is necessary to define targets of interest and reference compartments. In our laboratory, to estimate the proportion and volume of newly developed tissue components, the morphometric technique described by Howard and Reed [154] is used. When assessing rat AVL constructs or mouse flow-through pedicle constructs, the targets of interest include new blood vessels, matrix and new connective tissue and the AVL or flow through pedicle itself. In addition to the target, a reference compartment is also defined, namely a compartment to which a target can be compared to. In AVL constructs, two reference compartments were defined. The first compartment consists of the entire AVL construct comprised of AVL, fibrin matrix, new blood vessels and connective tissue. All points counted in each compartment are added together to give the total reference compartment count. However, to determine the vascularisation of the newly developed connective tissue alone, the second reference compartment is defined as the newly synthesized connective tissue which consists of combined counts for blood vessels and the newly synthesized connective tissue. A number of software tools are available for automated and semi-automated quantification of blood vessels and tissue components. In our laboratory, the Computer Assisted Stereological Toolbox (CAST) system is used (CAST System, Olympus, Denmark).

Method: To count the targets of interest, select a random 3–5 μm tissue section which contains 4–6 slices of AVL construct (1–2 sections of mouse construct) or 2–3 sections of skin flap tissue. Only complete tissue sections should be counted (Note: each of these slices should be approximately 500 to 1,000 μm apart) and labelled with B. Simplicifolia lectin (rat tissue) or CD 31 (mouse tissue).

  1. 1.

    Place a section on the automatic stage and outline the area of interest (reference compartment- usually the entire tissue section, or only the new connective tissue area in rat AVL constructs). At this point is it necessary to determine the percentage of area to be counted so that a representation of all specimens can be achieved. This is done through trial and error and it is best to undertake a small pilot study to determine the percentage area suitable for counting and the number of points suitable for counting. In our case, depending on the model, this percentage value is between 5% and 20% of total area. The software package them randomly and uniformly selects the fields where the counts are to be completed. (Note at least 200 total points per chamber construct should be counted. We generally count between 300 and 800 per chamber construct).

  2. 2.

    To determine vascular volume density, the number of points falling on the blood vessels is divided by the number of points falling on the total reference compartment [51, 141]. The percent vascular volume is then obtained by multiplying that number by 100%. Furthermore, since AVL tissue volume (and mouse chamber construct volume) is available, the percent vascular volume can be multiplied by the organ displacement volume to obtain absolute (total) vascular volume within a construct. This parameter is of particular value when determining the effect of a treatment on the construct development. The final result is represented as a function of time or as the outcome of treatment effect.

Rights and permissions

Reprints and permissions

Copyright information

© 2012 Springer Science+Business Media Dordrecht

About this chapter

Cite this chapter

Mitchell, G.M., Lokmic, Z., Sinha, S., Morrison, W.A. (2012). Skin Flap Models for Assessment of Angiogenesis. In: Zudaire, E., Cuttitta, F. (eds) The Textbook of Angiogenesis and Lymphangiogenesis: Methods and Applications. Springer, Dordrecht. https://doi.org/10.1007/978-94-007-4581-0_24

Download citation

Publish with us

Policies and ethics