Bladder Reconstruction with Human Amniotic Membrane in a Xenograft Rat Model: A Preclinical Study

Dimitri Barski, Holger Gerullis, Thorsten Ecke, Jin Yang, Gabriella Varga, Mihaly Boros, Isabel Pintelon, Jean-Pierre Timmermans, Thomas Otto, Dimitri Barski, Holger Gerullis, Thorsten Ecke, Jin Yang, Gabriella Varga, Mihaly Boros, Isabel Pintelon, Jean-Pierre Timmermans, Thomas Otto

Abstract

Background: Human amniotic membranes (HAMs) are assumed to have a number of unique characteristics including durability, hypoallergenic and anti-inflammatory properties. Materials and Methods: Multilayer HAMs from caesarian sections were applied to repair defined bladder defects in male Sprague-Dawley rats. The animals were sacrificed at 7, 21 and 42 days after implantation. Bladder volume capacity after grafting was measured. Histological analyses were performed to asses a number of parameters including HAM degradation, inflammatory reaction, graft rejection and smooth muscle ingrowth. Results: One rat died from sepsis in the treated group. No severe complications or signs of leakage were observed. Bladder capacity did not change over time. The initially increased inflammation in the HAM group diminished significantly over time (p<0.05). No signs of HAM degradation were observed and smooth muscle staining increased over time. Conclusions: HAMs appear to be durable and hypoallergenic grafts. The assumed suitability for the reconstruction of urinary tract justifies further research on detailed immunological process in larger grafts.

Keywords: IDEAL.; amniotic membrane; bladder augmentation; graft; rat experiment.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interest exists.

Figures

Figure 1
Figure 1
A. Preparation of amniotic membrane with dissection from the chorion under laminar flow. B. Dried and cryopreserved multilayered amniotic membrane graft.
Figure 2
Figure 2
Bladder capacity pre- and postoperatively in the amnion treated group A and control group C1. The bladder capacity reduced significantly in the control group due to the scaring of the sutured lesion (* p˂0.05).
Figure 3
Figure 3
Macroscopic and histological evaluation (H&E staining) of inflammation and AM degradation over time (21 and 42 days after grafting). Reduction of inflammation over time. Decreased inflammatory cells (*) and increased vascularization (V) in the periamniotic transition zone were detected. No signs of AM degradation. Scale bar 200µm.
Figure 4
Figure 4
Reduction of inflammation over time, semiquantative analyses of inflammatory cells: Treated (A): Amnion repair of bladder defect. Control/Lesion (C1): Closure of bladder defect with suture. Control/Amnion (C2): Amnion bladder onlay without defect. Semi-quantitative score: 0 = 50%; 200x magnification. Data is expressed as median with range. *, significant reduction (p˂0.05).
Figure 5
Figure 5
Measurement of amnion thickness over time (7, 21 and 42 days): Treated (A): Amnion repair of bladder defect. Control/Amnion (C2): Amnion bladder onlay without defect. Data is expressed as mean with SD, p˂0.05.
Figure 6
Figure 6
Immunohistological analysis of a reconstructed urinary bladder wall at timepoint 3 (42 days). A: Amnion group, reconstruction with AM graft; B: Control group, reconstruction with suture, AM; Amniotic membrane, Ur; Urothelium, BL; lumen of urinary bladder, X; region of lesion in the bladder wall. (A) H&E staining displaying mild inflammatory infiltration. α-Actin staining reveals frequently arranged smooth muscle bundles (arrows). Strong immunoreactivity underneath the urothelium layer is observed. The region of the lesion can be clearly recognized in the control group with muscle bundles, while there is a smooth muscle ingrowth in the treated group.

References

    1. Alberti C. Tissue engineering as innovative chance for organ replacement in radical tumor surgery. Eur Rev Med Pharmacol Sci. 2013;17(5):624–31.
    1. Otto T, Klosterhalfen B, Klinge U, Boros M, Ysebaert D, Williams K. Implants in urogynecology. Biomed Res Int. 2015;2015:354342.
    1. Atala A, Bauer SB, Soker S, Yoo JJ, Retik AB. Tissue-engineered autologous bladders for patients needing cystoplasty. Lancet. 2006;367(9518):1241–6.
    1. Joseph DB, Borer JG, De Filippo RE, Hodges SJ, McLorie GA. Autologous cell seeded biodegradable scaffold for augmentation cystoplasty: phase II study in children and adolescents with spina bifida. J Urol. 2014;191(5):1389–95.
    1. Probst M, Piechota HJ, Dahiya R, Tanagho EA. Homologous bladder augmentation in dog with the bladder acellular matrix graft. BJU Int. 2000;85(3):362–71.
    1. Wefer J, Sievert KD, Schlote N, Wefer AE, Nunes L, Dahiya R. et al. Time dependent smooth muscle regeneration and maturation in a bladder acellular matrix graft: histological studies and in vivo functional evaluation. J Urol. 2001;165(5):1755–9.
    1. Kikuno N, Kawamoto K, Hirata H, Vejdani K, Kawakami K, Fandel T. et al. Nerve growth factor combined with vascular endothelial growth factor enhances regeneration of bladder acellular matrix graft in spinal cord injury-induced neurogenic rat bladder. BJU Int. 2009;103(10):1424–8.
    1. Mitsui Y, Shiina H, Hiraoka T, Arichi N, Yasumoto H, Dahiya R. et al. Simultaneous implantation of bilateral ureters into bladder acellular matrix graft after partial cystectomy in a porcine model. BJU Int. 2012;110(11 Pt C):E1212–7.
    1. Roelofs LA, Oosterwijk E, Kortmann BB, Daamen WF, Tiemessen DM, Brouwer KM. et al. Bladder Regeneration Using a Smart Acellular Collagen Scaffold with Growth Factors VEGF, FGF2 and HB-EGF. Tissue Eng Part A. 2016;22(1-2):83–92.
    1. Caione P, Boldrini R, Salerno A, Nappo SG. Bladder augmentation using acellular collagen biomatrix: a pilot experience in exstrophic patients. Pediatr Surg Int. 2012;28(4):421–8.
    1. Horst M, Madduri S, Gobet R, Sulser T, Milleret V, Hall H. et al. Engineering functional bladder tissues. J Tissue Eng Regen Med. 2013;7(7):515–22.
    1. McCulloch P, Altman DG, Campbell WB, Flum DR, Glasziou P, Marshall JC. et al. No surgical innovation without evaluation: the IDEAL recommendations. Lancet. 2009;374(9695):1105–12.
    1. Sedrakyan A, Campbell B, Merino JG, Kuntz R, Hirst A, McCulloch P. IDEAL-D: a rational framework for evaluating and regulating the use of medical devices. BMJ. 2016;353:i2372.
    1. Lundbeck F, Djurhuus JC, Vaeth M. Bladder filling in mice: an experimental in vivo model to evaluate the reservoir function of the urinary bladder in a long term study. J Urol. 1989;141:1245–1249.
    1. Kim JC, Tseng SC. Transplantation of preserved human amniotic membrane for surface reconstruction in severely damaged rabbit corneas. Cornea. 1995;14(5):473–84.
    1. Meller D, Pauklin M, Thomasen H, Westekemper H, Steuhl KP. Amniotic membrane transplantation in the human eye. Dtsch Arztebl Int. 2011;108(14):243–8.
    1. de Farias CC, Allemann N, Gomes J. Randomized Trial Comparing Amniotic Membrane Transplantation with Lamellar Corneal Graft for the Treatment of Corneal Thinning. Cornea. 2016;35(4):438–44.
    1. Abdulhalim BE, Wagih MM, Gad AA, Boghdadi G, Nagy RR. Amniotic membrane graft to conjunctival flap in treatment of non-viral resistant infectious keratitis: a randomised clinical study. Br J Ophthalmol. 2015;99(1):59–63.
    1. Loeffelbein DJ, Rohleder NH, Eddicks M, Baumann CM, Stoeckelhuber M, Wolff KD. et al. Evaluation of human amniotic membrane as a wound dressing for split- thickness skin-graft donor sites. Biomed Res Int. 2014;2014:572183.
    1. Kumar A, Alraiyes AH, Gildea TR. Amniotic Membrane Graft for Bronchial Anastomotic Dehiscence in a Lung Transplant Recipient. Ann Am Thorac Soc. 2015;12(10):1583–6.
    1. Rohleder NH, Loeffelbein DJ, Feistl W, Eddicks M, Wolff KD, Gulati A. et al. Repair of oronasal fistulae by interposition of multilayered amniotic membrane allograft. Plast Reconstr Surg. 2013;132(1):172–81.
    1. Salehi SH, As'adi K, Mousavi SJ, Shoar S. Evaluation of Amniotic Membrane Effectiveness in Skin Graft Donor Site Dressing in Burn Patients. Indian J Surg. 2015;77(Suppl 2):427–31.
    1. Tsai RJ, Li LM, Chen JK. Reconstruction of damaged corneas by transplantation of autologous limbal epithelial cells. N Engl J Med. 2000;343(2):86–93.
    1. Yuan J, Li W, Huang J, Guo X, Li X, Lu X. et al. Transplantation of human adipose stem cell-derived hepatocyte-like cells with restricted localization to liver using acellular amniotic membrane. Stem Cell Res Ther. 2015;6:217.
    1. Lenko J. [Therapy of urethral stenosis with amnion transplantation] Pol Tyg Lek (Wars) 1955;10(34):1124–5.
    1. Shakeri S, Haghpanah A, Khezri A, Yazdani M, Monabbati A, Haghpanah S. et al. Application of amniotic membrane as xenograft for urethroplasty in rabbit. Int Urol Nephrol. 2009;41(4):895–901.
    1. Wang F, Liu T, Yang L, Zhang G, Liu H, Yi X. et al. Urethral reconstruction with tissue-engineered human amniotic scaffold in rabbit urethral injury models. Med Sci Monit. 2014;20:2430–8.
    1. Koziak A, Salagierski M, Marcheluk A, Szcześniewski R, Sosnowski M. Early experience in reconstruction of long ureteral strictures with allogenic amniotic membrane. Int J Urol. 2007;14(7):607–10.
    1. Brandt FT, Albuquerque CD, Lorenzato FR. Female urethral reconstruction with amnion grafts. Int J Surg Investig. 2000;1(5):409–14.
    1. Barski D, Gerullis H, Ecke T, Varga G, Boros M, Pintelon I. et al. Repair of a vesico-vaginal fistula with amniotic membrane - Step 1 of the IDEAL recommendations of surgical innovation. Cent European J Urol. 2015;68(4):459–61.
    1. Price DT, Price TC. Robotic repair of a vesicovaginal fistula in an irradiated field using a dehydrated amniotic allograft as an interposition patch. J Robot Surg. 2016;10(1):77–80.
    1. Adamowicz J, Pokrywczyńska M, Tworkiewicz J, Kowalczyk T, van Breda SV, Tyloch D. et al. New Amniotic Membrane Based Biocomposite for Future Application in Reconstructive Urology. PLoS One. 2016;11(1):e0146012.
    1. Wall LL, Wiskind AK, Taylor PA. Simple bladder filling with a cough stress test compared with subtracted cystometry for the diagnosis of urinary incontinence. Am J Obstet Gynecol. 1994;171:1472. 1994.
    1. Resch MD, Schlötzer-Schrehardt U, Hofmann-Rummelt C, Sauer R, Kruse FE, Beckmann MW. et al. Integration patterns of cryopreserved amniotic membranes into the human cornea. Ophthalmology. 2006;113(11):1927–35.
    1. Laschke MW, Harder Y, Amon M, Martin I, Farhadi J, Ring A. et al. Angiogenesis in tissue engineering: breathing life into constructed tissue substitutes. Tissue Eng. 2006;12(8):2093–104.
    1. Koizumi N, Inatomi T, Quantock AJ, Fullwood NJ, Dota A, Kinoshita S. Amniotic membrane as a substrate for cultivating limbal corneal epithelial cells for autologous transplantation in rabbits. Cornea. 2000;19(1):65–71.
    1. Yates CC, Hebda P, Wells A. Skin wound healing and scarring: fetal wounds and regenerative restitution. Birth Defects Res C Embryo Today. 2012;96(4):325–33.
    1. Solomon A, Wajngarten M, Alviano F, Anteby I, Elchalal U, Pe'er J. et al. Suppression of inflammatory and fibrotic responses in allergic inflammation by the amniotic membrane stromal matrix. Clinical & Experimental Allergy. 2005;35(7):941–948.
    1. Akle CA, Adinolfi M, Welsh KI, Leibowitz S, McColl I. Immunogenicity of human amniotic epithelial cells after transplantation into volunteers. Lancet. 1981;2(8254):1003–5.
    1. Kesting MR, Loeffelbein DJ, Steinstraesser L, Muecke T, Demtroeder C, Sommerer F. et al. Cryopreserved human amniotic membrane for soft tissue repair in rats. Ann Plast Surg. 2008;60(6):684–91.
    1. Bhowmik DM, Dinda AK, Mahanta P, Agarwal SK. The evolution of the Banff classification schema for diagnosing renal allograft rejection and its implications for clinicians. Indian J Nephrol. 2010;20(1):2–8.

Source: PubMed

3
Abonnere