C. Kugler, J. Gottlieb, G. Warnecke, A. Schwarz, K. Weissenborn et al., Health-related quality of life after solid organ transplantation: a prospective, multiorgan cohort study, Transplantation, vol.96, pp.316-323, 2013.

J. R. Nuñez, D. Rio, F. Lopez, E. Moreno, M. A. Soria et al., Non-heartbeating donors: an excellent choice to increase the donor pool, Transplant Proc, vol.37, pp.3651-3654, 2005.

J. H. Daemen, R. J. De-wit, M. W. Bronkhorst, M. Yin, E. Heineman et al., Non-heart-beating donor program contributes 40% of kidneys for transplantation, Transplant Proc, vol.28, pp.105-106, 1996.

A. C. Farney, R. P. Singh, M. H. Hines, J. Rogers, E. L. Hartmann et al., Experience in renal and extrarenal transplantation with donation after cardiac death donors with selective use of extracorporeal support, J Am Coll Surg, vol.206, pp.1028-1037, 2008.

A. D. Barlow, M. S. Metcalfe, Y. Johari, R. Elwell, P. S. Veitch et al., Case-matched comparison of long-term results of non-heart beating and heart-beating donor renal transplants, Br J Surg, vol.96, pp.685-691, 2009.

G. Benoit, H. Bensadoun, I. Borie, G. Vavasseur, and C. Gillot, A double balloon catheter for the procurement of abdominal organs, Ann Urol, vol.24, pp.199-201, 1990.

M. G. Snoeijs, A. J. Dekkers, W. A. Buurman, L. Van-den-akker, R. J. Welten et al., situ preservation of kidneys from donors after cardiac death: results and complications, vol.246, pp.844-852, 2007.

R. Valero, C. Cabrer, F. Oppenheimer, E. Trias, J. Sánchez-ibáñez et al., Normothermic recirculation reduces primary graft dysfunction of kidneys obtained from non-heart-beating donors, Transpl Int, vol.13, pp.303-310, 2000.

C. Fondevila, A. J. Hessheimer, A. Ruiz, D. Calatayud, J. Ferrer et al., Liver transplant using donors after unexpected cardiac death: novel preservation protocol and acceptance criteria, Am J Transplant, vol.7, pp.1849-1855, 2007.

W. J. Ko, Y. S. Chen, P. R. Tsai, and P. H. Lee, Extracorporeal membrane oxygenation support of donor abdominal organs in non-heart-beating donors, Clin Transplant, vol.14, pp.152-156, 2000.

M. T. Gravel, J. D. Arenas, . Chenault-r-2nd, J. C. Magee, S. Rudich et al., Kidney transplantation from organ donors following cardiopulmonary death using extracorporeal membrane oxygenation support, Ann Transplant, vol.9, pp.57-58, 2004.

C. Billault, F. Godfroy, T. F. Bart, S. Arzouk, N. Van-glabeke et al., Organ procurement from donors deceased from cardiac death: a single-center efficiency assessment, Transplant Proc, vol.43, pp.3396-3397, 2011.

C. Antoine, F. Brun, A. Tenaillon, and B. Loty, Organ procurement and transplantation from non-heart-beating donors, Nephrol Ther, vol.4, pp.5-14, 2008.

B. Barrou, C. Billault, and A. Nicolas-robin, The use of extracorporeal membraneous oxygenation in donors after cardiac death, Curr Opin Organ Transplant, vol.18, pp.148-153, 2013.

T. Hauet, J. M. Goujon, H. Baumert, I. Petit, M. Carretier et al., Polyethylene glycol reduces the inflammatory injury due to cold ischemia/reperfusion in autotransplanted pig kidneys, Kidney Int, vol.62, pp.654-667, 2002.

S. Giraud, R. Thuillier, A. Belliard, W. Hebrard, C. Nadeau et al., Direct thrombin inhibitor prevents delayed graft function in a porcine model of renal transplantation, Transplantation, vol.87, pp.1636-1644, 2009.

S. Giraud, F. Favreau, N. Chatauret, R. Thuillier, S. Maiga et al., Contribution of large pig for renal ischemia-reperfusion and transplantation studies: the preclinical model, J Biomed Biotechnol, p.532127, 2011.

C. Fondevila, A. J. Hessheimer, E. Flores, A. Ruiz, N. Mestres et al., Applicability and results of Maastricht type 2 donation after cardiac death liver transplantation, Am J Transplant, vol.12, pp.162-170, 2012.

J. R. References-1.-nuñez, F. Delrio, E. Lopez, M. A. Moreno, A. Soria et al., Non-heart-beating donors: an excellent choice to increase the donor pool, Transplant Proc, vol.37, issue.9, pp.3651-3654, 2005.

J. H. Daemen, R. J. Dewit, M. W. Bronkhorst, M. Yin, E. Heineman et al., Non-heart-beating donor program contributes 40% of kidneys for transplantation, Transplant Proc, vol.28, issue.1, pp.105-106, 1996.

J. W. Daemen, A. P. Oomen, W. P. Kelders, and G. Kootstra, The potential pool of non-heart-beating kidney donors, Clin Transplant, vol.11, issue.2, pp.149-154, 1997.

J. C. García-valdecasas and C. Fondevila, In-vivo normothermic recirculation: an update, Curr Opin Organ Transplant, vol.15, issue.2, pp.173-176, 2010.

M. D. Doshi and L. G. Hunsicker, Short-and long-term outcomes with the use of kidneys and livers donated after cardiac death, Am J Transplant

A. Gallinat, A. Paul, and P. Efferz, Role of oxygenation in hypothermic machine perfusion of kidneys from heart beating donors, Transplantation, vol.94, p.809, 2012.

N. Vaziri, R. Thuillier, and F. D. Favreau, Analysis of machine perfusion benefits in kidney grafts: a preclinical study, J Transl Med, vol.9, p.15, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-00663684

I. Jochmans, E. Lerut, J. Van-pelt, D. Monbaliu, and J. Pirenne, Circulating AST, H-FABP, and NGAL are early and accurate biomarkers of graft injury and dysfunction in a preclinical model of kidney transplantation, Ann Surg, vol.254, p.784, 2011.

P. C. Grimm, P. Nickerson, and J. Gough, Computerized image analysis of Sirius Red-stained renal allograft biopsies as a surrogate marker to predict long-term allograft function, J Am Soc Nephrol, vol.14, p.1662, 2003.

R. Thuillier, R. Codas, and E. Marchand, Chronic renoprotective effect of pulsatile perfusion machine RM3 and IGL-1 solution in a preclinical kidney transplantation model, J Transl Med, vol.10, p.233, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00773201

S. Bedi, A. Vidyasagar, and A. Djamali, Epithelial-to-mesenchymal transition and chronic allograft tubulointerstitial fibrosis, Transplant Rev, vol.22, p.1, 2008.
DOI : 10.1016/j.trre.2007.09.004

URL : http://europepmc.org/articles/pmc2184838?pdf=render

D. Ferenbach, D. C. Kluth, and J. Hughes, Inflammatory cells in renal injury and repair, Semin Nephrol, vol.27, p.250, 2007.

J. T. Fletcher, B. J. Nankivell, and S. I. Alexander, Chronic allograft nephropathy, Pediatr Nephrol, vol.24, p.1465, 2009.

B. J. Nankivell and J. R. Chapman, Chronic allograft nephropathy: current concepts and future directions, Transplantation, vol.81, p.643, 2006.
DOI : 10.1097/01.tp.0000190423.82154.01

R. Thuillier and R. B. Mannon, Chronic allograft failure: natural history, pathogenesis, diagnosis and management, 2007.

J. W. Copeland, B. W. Beaumont, M. J. Merrilees, and H. L. Pilmore, Epithelial-to-mesenchymal transition of human proximal tubular epithelial cells: effects of rapamycin, mycophenolate, cyclosporin, azathioprine, and methylprednisolone, Transplantation, vol.83, p.809, 2007.

J. B. Cowland and N. Borregaard, Molecular characterization and pattern of tissue expression of the gene for neutrophil gelatinase-associated lipocalin from humans, Genomics, vol.45, p.17, 1997.

M. Haase, R. Bellomo, P. Devarajan, P. Schlattmann, and A. Haasefielitz, Accuracy of neutrophil gelatinase-associated lipocalin (NGAL) in diagnosis and prognosis in acute kidney injury: a systematic review and meta-analysis, Am J Kidney Dis, vol.54, p.1012, 2009.

T. Desurmont, S. Giraud, and J. Cau, Trophic factor and FR167653 supplementation during cold storage rescue chronic renal injury, J Urol, vol.185, p.1139, 2011.
DOI : 10.1016/j.juro.2010.10.032

Z. M. El-zoghby, M. D. Stegall, and D. J. Lager, Identifying specific causes of kidney allograft loss, Am J Transpl, vol.9, p.527, 2009.
DOI : 10.1111/j.1600-6143.2008.02519.x

URL : https://onlinelibrary.wiley.com/doi/pdf/10.1111/j.1600-6143.2008.02519.x

R. Codas, R. Thuillier, T. Hauet, and L. Badet, Renoprotective effect of pulsatile perfusion machine RM3: pathophysiological and kidney injury biomarker characterization in a preclinical model of autotransplanted pig, BJU Int, vol.109, p.141, 2012.

R. Thuillier, D. Dutheil, and M. T. Trieu, Supplementation with a new therapeutic oxygen carrier reduces chronic fibrosis and organ dysfunction in kidney static preservation, 1845. journal of surgical research, vol.11, pp.1174-1181, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01250926

R. Thuillier, G. Allain, G. S. Saintyves, T. Delpech, P. O. Couturier et al.,

, Keywords: Antioxidants, graft outcome, ischemia reperfusion injury, kidney transplantation, preservation Abbreviations: aSMA, a-smooth muscle actin, Am J Transplant, vol.14, issue.5, pp.1073-83, 2014.

, AUC, area under the curve; CDC, cyclodextrin-complexed curcumin; DCD, donation after circulatory death; EMT, epithelial-to-mesenchymal transition; HBSS, Hank's Balanced Salt Solution; IFTA, interstitial fibrosis and tubular atrophy

, IRI, ischemia reperfusion injury

, LDH, lactate dehydrogenase

, MAPK, mitogen-activated protein kinase

H. U. Nf-kb-;-meier-kriesche, J. D. Schold, T. R. Srinivas, and B. Kaplan, Lack of improvement in renal allograft survival despite a marked decrease in acute rejection rates over the most recent era, nuclear factor kappa beta; RTqPCR, real-time quantitative polymerase chain reaction, vol.4, pp.378-383, 2004.

F. K. Port, J. L. Bragg-gresham, and R. A. Metzger, Donor characteristics associated with reduced graft survival: An approach to expanding the pool of kidney donors, Transplantation, vol.74, pp.1281-1286, 2002.

R. W. Jamieson and P. J. Friend, Organ reperfusion and preservation, Front Biosci, vol.13, pp.221-235, 2008.
DOI : 10.2741/2672

A. K. Salahudeen, Consequences of cold ischemic injury of kidneys in clinical transplantation, J Investig Med, vol.52, pp.296-298, 2004.

A. K. Salahudeen, Cold ischemic injury of transplanted kidneys: New insights from experimental studies, Am J Physiol, vol.287, pp.181-187, 2004.

A. K. Salahudeen, Cold ischemic injury of transplanted organs: Some new strategies against an old problem, Am J Transplant, vol.4, p.1, 2004.

J. P. Faure, H. Baumert, and Z. Han, Evidence for a protective role of trimetazidine during cold ischemia: Targeting inflammation and nephron mass, Biochem Pharmacol, vol.66, pp.2241-2250, 2003.

T. Hauet, J. M. Goujon, and A. Vandewalle, Trimetazidine reduces renal dysfunction by limiting the cold ischemia/reperfusion injury in autotransplanted pig kidneys, J Am Soc Nephrol, vol.11, pp.138-148, 2000.

C. Jayle, F. Favreau, and K. Zhang, Comparison of protective effects of trimetazidine against experimental warm ischemia of different durations: Early and long-term effects in a pig kidney model, Am J Physiol, vol.292, pp.1082-1093, 2007.

L. Rossard, F. Favreau, and S. Giraud, Role of warm ischemia on innate and adaptive responses in a preclinical renal auto-transplanted porcine model, J Transl Med, vol.11, p.129, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00827220

L. Rossard, F. Favreau, and J. Demars, Evaluation of early regenerative processes in a preclinical pig model of acute kidney injury, Curr Mol Med, vol.12, pp.502-505, 2012.

A. Djamali and M. Samaniego, Fibrogenesis in kidney transplantation: Potential targets for prevention and therapy, Transplantation, vol.88, pp.1149-1156, 2009.

H. R. Jang and H. Rabb, The innate immune response in ischemic acute kidney injury, Clin Immunol, vol.130, pp.41-50, 2009.

L. Li, L. Huang, and S. S. Sung, NKT cell activation mediates neutrophil IFN-gamma production and renal ischemia-reperfusion injury, J Immunol, vol.178, pp.5899-5911, 2007.
DOI : 10.4049/jimmunol.178.9.5899

URL : http://www.jimmunol.org/content/jimmunol/178/9/5899.full.pdf

G. R. Kinsey, R. Sharma, and L. Huang, Regulatory T cells suppress innate immunity in kidney ischemia-reperfusion injury, J Am Soc Nephrol, vol.20, pp.1744-1753, 2009.

D. P. Basile, D. Donohoe, K. Roethe, and J. L. Osborn, Renal ischemic injury results in permanent damage to peritubular capillaries and influences long-term function, Am J Physiol, vol.281, pp.887-899, 2001.

G. Cicco, P. C. Panzera, G. Catalano, and V. Memeo, Microcirculation and reperfusion injury in organ transplantation, Adv Exp Med Biol, vol.566, pp.363-373, 2005.
DOI : 10.1007/0-387-26206-7_48

T. A. Sutton, Alteration of microvascular permeability in acute kidney injury, Microvasc Res, vol.77, pp.4-7, 2009.

A. S. Strimpakos and R. A. Sharma, Curcumin: Preventive and therapeutic properties in laboratory studies and clinical trials, Antioxid Redox Signal, vol.10, pp.511-545, 2008.
DOI : 10.1089/ars.2007.1769

B. Mishra, K. I. Priyadarsini, M. K. Bhide, R. M. Kadam, and H. Mohan, Reactions of superoxide radicals with curcumin: Probable mechanisms by optical spectroscopy and EPR, Free Radic Res, vol.38, pp.355-362, 2004.

C. Jobin, C. A. Bradham, and M. P. Russo, Curcumin blocks cytokinemediated NF-kappa B activation and proinflammatory gene expression by inhibiting inhibitory factor I-kappa B kinase activity, J Immunol, vol.163, pp.3474-3483, 1999.

B. B. Aggarwal and B. Sung, Pharmacological basis for the role of curcumin in chronic diseases: An age-old spice with modern targets, Trends Pharmacol Sci, vol.30, pp.85-94, 2009.

Y. S. Kim, Y. Ahn, and M. H. Hong, Curcumin attenuates inflammatory responses of TNF-alpha-stimulated human endothelial cells, J Cardiovasc Pharmacol, vol.50, pp.41-49, 2007.

D. A. Shoskes, Effect of bioflavonoids quercetin and curcumin on ischemic renal injury: A new class of renoprotective agents, Transplantation, vol.66, pp.147-152, 1998.

E. A. Jones, A. Shahed, and D. A. Shoskes, Modulation of apoptotic and inflammatory genes by bioflavonoids and angiotensin II inhibition in ureteral obstruction, Urology, vol.56, pp.346-351, 2000.

O. Bayrak, E. Uz, and R. Bayrak, Curcumin protects against ischemia/reperfusion injury in rat kidneys, World J Urol, vol.26, pp.285-291, 2008.

J. Gaedeke, N. A. Noble, and W. A. Border, Curcumin blocks fibrosis in antiThy 1 glomerulonephritis through up-regulation of heme oxygenase 1, Kidney Int, vol.68, pp.2042-2049, 2005.

S. S. Ghosh, H. D. Massey, and R. Krieg, Curcumin ameliorates renal failure in 5/6 nephrectomized rats: Role of inflammation, Am J Physiol, vol.296, pp.1146-1157, 2009.

N. M. Rogers, M. D. Stephenson, A. R. Kitching, J. D. Horowitz, and P. T. Coates, Amelioration of renal ischaemia-reperfusion injury by liposomal delivery of curcumin to renal tubular epithelial and antigen-presenting cells, Br J Pharmacol, vol.166, pp.194-209, 2012.

V. R. Yadav, S. Prasad, and R. Kannappan, Cyclodextrin-complexed curcumin exhibits anti-inflammatory and antiproliferative activities superior to those of curcumin through higher cellular uptake, Biochem Pharmacol, vol.80, pp.1021-1032, 2010.

M. N. Simmons, M. J. Schreiber, and I. S. Gill, Surgical renal ischemia: A contemporary overview, J Urol, vol.180, pp.19-30, 2008.

S. Giraud, F. Favreau, N. Chatauret, R. Thuillier, S. Maiga et al., Contribution of large pig for renal ischemia-reperfusion and transplantation studies: The preclinical model, J Biomed Biotechnol, p.532127, 2011.

F. Favreau, R. Thuillier, and J. Cau, Anti-thrombin therapy during warm ischemia and cold preservation prevents chronic kidney graft fibrosis in a DCD model, Am J Transplant, vol.10, pp.30-39, 2010.

R. Thuillier, F. Favreau, O. Celhay, L. Macchi, S. Milin et al., Thrombin inhibition during kidney ischemia-reperfusion reduces chronic graft inflammation and tubular atrophy, Transplantation, vol.90, pp.612-621, 2010.

J. W. Copeland, B. W. Beaumont, M. J. Merrilees, and H. L. Pilmore, Epithelial-to-mesenchymal transition of human proximal tubular epithelial cells: Effects of rapamycin, mycophenolate, cyclosporin, azathioprine, and methylprednisolone, Transplantation, vol.83, pp.809-814, 2007.

A. Kunwar, A. Barik, R. Pandey, and K. I. Priyadarsini, Transport of liposomal and albumin loaded curcumin to living cells: An absorption and fluorescence spectroscopic study, Biochim Biophys Acta, vol.1760, pp.1513-1520, 2006.

S. Giraud, R. Thuillier, and A. Belliard, Direct thrombin inhibitor prevents delayed graft function in a porcine model of renal transplantation, Transplantation, vol.87, pp.1636-1644, 2009.

T. Hauet, J. M. Goujon, and H. Baumert, Polyethylene glycol reduces the inflammatory injury due to cold ischemia/reperfusion in autotransplanted pig kidneys, Kidney Int, vol.62, pp.654-667, 2002.

R. Thuillier, D. Dutheil, and M. T. Trieu, Supplementation with a new therapeutic oxygen carrier reduces chronic fibrosis and organ dysfunction in kidney static preservation, Am J Transplant, vol.11, pp.1845-1860, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01250926

K. Solez, R. B. Colvin, and L. C. Racusen, Banff 07 classification of renal allograft pathology: Updates and future directions, Am J Transplant, vol.8, pp.753-760, 2008.

P. C. Grimm, P. Nickerson, and J. Gough, Computerized image analysis of Sirius Red-stained renal allograft biopsies as a surrogate marker to predict long-term allograft function, J Am Soc Nephrol, vol.14, pp.1662-1668, 2003.

A. Dahan, J. M. Miller, A. Hoffman, G. E. Amidon, and G. L. Amidon, The solubility-permeability interplay in using cyclodextrins as pharmaceutical solubilizers: Mechanistic modeling and application to progesterone, J Pharm Sci, vol.99, pp.2739-2749, 2010.

V. J. Stella, Q. He, and . Cyclodextrins, Toxicol Pathol, vol.36, pp.30-42, 2008.

E. P. Cummins, E. Berra, and K. M. Comerford, Prolyl hydroxylase-1 negatively regulates IkappaB kinase-beta, giving insight into hypoxia-induced NFkappaB activity, Proc Natl Acad Sci, vol.103, pp.18154-18159, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00320775

L. Baum and A. Ng, Curcumin interaction with copper and iron suggests one possible mechanism of action in Alzheimer's disease animal models, J Alzheimers Dis, vol.6, pp.443-449, 2004.

R. Sreejayan and . Mn, Curcuminoids as potent inhibitors of lipid peroxidation, J Pharm Pharmacol, vol.46, pp.1013-1016, 1994.

T. Nakagawa, D. H. Kang, and R. Ohashi, Tubulointerstitial disease: Role of ischemia and microvascular disease, Curr Opin Nephrol Hypertens, vol.12, pp.233-241, 2003.

B. B. Gourdinmj and D. Kockm, The impactof ischaemia-reperfusion on the blood vessel, Eur J Anaesthesiol, vol.26, pp.537-547, 2009.

X. Guo, R. E. Gerl, and J. W. Schrader, Defining the involvement of p38alpha MAPK in the production of anti-and proinflammatory cytokines using an SB 203580-resistant form of the kinase, J Biol Chem, vol.278, pp.22237-22242, 2003.

E. Herlaar and Z. Brown, p38 MAPK signalling cascades in inflammatory disease, Mol Med Today, vol.5, pp.439-447, 1999.

L. Shen and H. F. Ji, The pharmacology of curcumin: Is it the degradation products?, Trends Mol Med, vol.18, pp.138-144, 2012.

B. J. Nankivell and J. R. Chapman, Chronic allograft nephropathy: Current concepts and future directions, Transplantation, vol.81, pp.643-654, 2006.

Z. M. El-zoghby, M. D. Stegall, and D. J. Lager, Identifying specific causes of kidney allograft loss, Am J Transplant, vol.9, pp.527-535, 2009.

N. Chatauret, R. Thuillier, and T. Hauet, Preservation strategies to reduce ischemic injury in kidney transplantation: Pharmacological and genetic approaches, Curr Opin Organ Transplant, vol.16, pp.180-187, 2011.

R. Codas, T. Hauet, and L. Badet, Renoprotective effect of Pulsatile perfusion machine RM3: Pathophysiologic and kidney injury biomarker characterization in preclinical model of autotransplanted pig, BJU Int, vol.109, pp.141-147, 2012.

C. Moers, J. M. Smits, and M. H. Maathuis, Machine perfusion or cold storage in deceased-donor kidney transplantation, N Engl J Med, vol.360, pp.7-19, 2009.

N. Vaziri, R. Thuillier, and F. D. Favreau, Analysis of machine perfusion benefits in kidney grafts: A preclinical study, J Transl Med, vol.9, p.15, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-00663684

. Valorisation and . Travail,

G. Allain, S. Giraud, W. Hébrard, R. Thuillier, B. Barrou et al., Journal de la Société de Chirurgie Thoracique et Cardio-Vasculaire, vol.16, pp.219-224, 2012.

, -Reconditionnement des greffons rénaux issus des donneurs décédés par arrêt cardiaque : comparaison des techniques de refroidissement in situ et de circulation régionale normothermique sur un modèle porcin d'allogreffe

G. Allain, S. Giraud, R. Thuret, T. Hauet, P. Corbi et al., Journal de la Société de Chirurgie Thoracique et Cardio-Vasculaire, vol.19, pp.1-6, 2015.

, Communications orales

, Société Française de Chirurgie Thoracique et Cardio-Vasculaire : -Evaluation pré-clinique de la transplantation rénale issue de donneur à coeur arrêté

G. Allain, S. Giraud, W. Hebrard, R. Thuillier, B. Barrou et al., , vol.64

, Prix de la meilleure communication orale -Greffons rénaux issus des donneurs décédés par arrêt cardiaque : la circulation régionale normothermique permet un meilleur reconditionnement

G. Allain, S. Giraud, R. Thuret, T. Hauet, P. Corbi et al., , vol.68

, Mise au point d'un modèle expérimental porcin de prise en charge d'un donneur décédé par arrêt cardiaque par mise en place d'une circulation régionale normothermique

G. Allain, S. Giraud, R. Thuillier, C. Jayle, W. Hebrard et al., , vol.10

, European Society for Organ Transplantation: A pre-clinical evaluation of renal transplant from deceased after cardiac arrest donors

G. Allain, S. Giraud, W. Hebrard, C. Jayle, R. Thuillier et al., , vol.15

, Académie Nationale de Chirurgie -Association Française de Chirurgie : Evaluation pré-clinique des greffons rénaux issus des donneurs décédés par arrêt cardiaque reconditionnés par circulation régionale normothermique

G. Allain, S. Giraud, W. Hebrard, R. Thuillier, B. Barrou et al., Forum de recherche chirurgicale

, Donneur décédé par arrêt cardiaque : étude préclinique de l'hémostase et de l'inflammation lors d'une circulation régionale normothermique

G. Allain, T. Kerforne, S. Giraud, W. Hébrard, V. Ameteau et al., Jayle, vol.70

M. Tonelli, N. Wiebe, G. Knoll, A. Bello, S. Browne et al., Systematic review: kidney transplantation compared with dialysis in clinically relevant outcomes, Am J Transplant, vol.11, issue.10, pp.2093-109, 2011.

D. A. Axelrod, M. A. Schnitzler, H. Xiao, W. Irish, E. Tuttle-newhall et al.,

T. and L. Kl, An economic assessment of contemporary kidney transplant practice, Am J Transplant, vol.18, issue.5, pp.1168-1176, 2018.

B. Bozkurt and M. K?l?ç, Marginal Donors in Renal Transplantation, Transplant Proc, vol.015, issue.5, pp.1273-81

A. Hart, J. M. Smith, M. A. Skeans, S. K. Gustafson, A. R. Wilk et al., Annual Data Report: Kidney. Am J Transplant, vol.18, issue.1, pp.18-113, 2016.

U. Maggiore, R. Oberbauer, J. Pascual, O. Viklicky, C. Dudley et al., Strategies to increase the donor pool and access to kidney transplantation: an international perspective, Nephrol Dial Transplant, vol.30, issue.2, pp.217-239, 2015.

P. L. Tso, W. A. Dar, and M. L. Henry, With respect to elderly patients: finding kidneys in the context of new allocation concepts, Am J Transplant, vol.12, issue.5, pp.1091-1099, 2012.

J. Pascual, J. Zamora, and J. D. Pirsch, A systematic review of kidney transplantation from expanded criteria donors, Am J Kidney Dis, vol.52, issue.3, pp.553-86, 2008.

B. Palkoci, M. Vojtko, J. Fialová, D. Osinová, and M. Laj?iaková, Results of Kidney Transplantation from Expanded Criteria Donors: A Single-Center Experience, Int J Organ Transplant Med

C. L. Davis and F. L. Delmonico, Living-donor kidney transplantation: a review of the current practices for the live donor, J Am Soc Nephrol, vol.16, issue.7, pp.2098-110, 2005.

G. Kootstra, J. H. Daemen, and A. P. Oomen, Categories of non-heart-beating donors, Transplant Proc, vol.27, issue.5, pp.2893-2897, 1995.

B. Domínguez-gil, B. Haase-kromwijk, H. Van-leiden, J. Neuberger, L. Coene et al., European Committee (Partial Agreement) on Organ Transplantation. Council of Europe (CD-P-TO). Current situation of donation after circulatory death in European countries, Transpl Int, vol.24, issue.7, pp.676-86, 2011.

B. Domínguez-gil, J. Duranteau, A. Mateos, J. R. Núñez, G. Cheisson et al.,

F. Río, R. Valero, E. Coll, M. Thuong, M. Z. Akhtar et al., Uncontrolled donation after circulatory death: European practices and recommendations for the development and optimization of an effective programme, Transpl Int, vol.29, issue.8, pp.842-59, 2016.

G. C. Oniscu, L. V. Randle, P. Muiesan, A. J. Butler, I. S. Currie et al., In situ normothermic regional perfusion for controlled donation after circulatory death--the United Kingdom experience, Am J Transplant, vol.14, issue.12, pp.2846-54, 2014.

M. Thuong, A. Ruiz, P. Evrard, M. Kuiper, C. Boffa et al., New classification of donation after circulatory death donors definitions and terminology, Transpl Int, 2016.

G. Benoit, H. Bensadoun, I. Borie, G. Vavasseur, and C. Gillot, A double balloon catheter for the procurement of abdominal organs, Ann Urol (Paris), vol.24, issue.3, pp.199-201, 1990.

C. Lazzeri, M. Bonizzoli, S. Valente, G. Cianchi, M. L. Migliaccio et al., The role of extracorporeal membrane oxygenation in donation after circulatory death, Minerva Anestesiol, 2014.

L. B. Johnson, J. S. Plotkin, C. D. Howell, M. J. Njoku, P. C. Kuo et al., Successful emergency transplantation of a liver allograft from a donor maintained on extracorporeal membrane oxygenation, Transplantation, vol.63, issue.6, pp.910-911, 1997.

I. M. Shapey and P. Muiesan, Regional perfusion by extracorporeal membrane oxygenation of abdominal organs from donors after circulatory death: a systematic review, Liver Transpl, 2013.

R. Valero, C. Cabrer, F. Oppenheimer, E. Trias, J. Sánchez-ibáñez et al., Normothermic recirculation reduces primary graft dysfunction of kidneys obtained from non-heart-beating donors, Transpl Int, vol.13, issue.4, pp.303-313, 2000.

J. Demiselle, J. F. Augusto, M. Videcoq, E. Legeard, L. Dubé et al., Transplantation of kidneys from uncontrolled donation after circulatory determination of death: comparison with brain death donors with or without extended criteria and impact of normothermic regional perfusion, Transpl Int, vol.29, issue.4, pp.432-474, 2016.

C. Delsuc, A. Faure, J. Berthiller, D. Dorez, X. Matillon et al., Uncontrolled donation after circulatory death: comparison of two kidney preservation protocols on graft outcomes, BMC Nephrol, vol.19, issue.1, p.3, 2018.

J. Arias-diaz, J. Alvarez, M. R. Del-barrio, and J. L. Balibrea, Non-heart-beating donation: current state of the art, Transplant Proc, vol.36, issue.7, pp.1891-1894, 2004.

A. Alonso, C. Fernández-rivera, P. Villaverde, J. Oliver, S. Cillero et al., Renal transplantation from non-heart-beating donors: a single-center 10-year experience, Transplant Proc, vol.37, issue.9, pp.3658-60, 2005.

A. O. Sanni, C. H. Wilson, H. Wyrley-birch, D. Vijayanand, A. Navarro et al.,

B. Rix, D. Soomro, N. Manas, D. Talbot, and D. , Non-heart-beating kidney transplantation: 6-year outcomes, Transplant Proc, vol.38, issue.10, pp.3396-3403, 2006.

F. Dupriez, D. Pauw, L. , D. T. Mourad, M. Penaloza et al.,

F. , Fourteen years of experience in uncontrolled organ donation after cardio-circulatory death, Transplant Proc, vol.46, issue.9, pp.3134-3141, 2014.

M. L. Nicholson, M. S. Metcalfe, S. A. White, J. R. Waller, T. M. Doughman et al., A comparison of the results of renal transplantation from non-heart-beating, conventional cadaveric, and living donors, Kidney Int, vol.58, issue.6, pp.2585-91, 2000.

A. D. Barlow, M. S. Metcalfe, Y. Johari, R. Elwell, P. S. Veitch et al., Case-matched comparison of long-term results of non-heart beating and heart-beating donor renal transplants, Br J Surg, vol.96, issue.6, pp.685-91, 2009.

F. Fieux, M. R. Losser, E. Bourgeois, F. Bonnet, M. O. Gaudez et al.,

F. , M. F. Adnet, F. Jacob, and L. , Kidney retrieval after sudden out of hospital refractory cardiac arrest: a cohort of uncontrolled non heart beating donors, Crit Care, vol.13, issue.4, p.141, 2009.

M. C. De-gracia, J. M. Osorio, J. M. Pérez-villares, P. Galindo, M. C. Ruiz et al., A new program of kidney transplantation from donors after cardiac death in Spain, Transplant Proc, vol.44, issue.9, pp.2518-2538, 2012.

T. Gaibor, N. G. González-roncero, F. M. , C. Cabrera, M. et al., Survival and Evolution of Renal Function in Kidney Transplant Recipients From Type II Asystolic Donations: A Single-center Experience, Transplant Proc, vol.50, issue.2, pp.565-568, 2018.

A. C. Farney, R. P. Singh, M. H. Hines, J. Rogers, E. L. Hartmann et al., Experience in renal and extrarenal transplantation with donation after cardiac death donors with selective use of extracorporeal support, J Am Coll Surg, 2008.

A. I. Sánchez-fructuoso, D. Prats, J. Torrente, M. J. Pérez-contín, C. Fernández et al., Renal transplantation from non-heart beating donors: a promising alternative to enlarge the donor pool, J Am Soc Nephrol, vol.11, issue.2, pp.350-358, 2000.

C. Y. Lee, M. K. Tsai, W. J. Ko, C. J. Chang, R. H. Hu et al., Expanding the donor pool: use of renal transplants from non-heart-beating donors supported with extracorporeal membrane oxygenation, Clin Transplant, vol.19, issue.3, pp.383-90, 2005.

M. Molina, F. Guerrero-ramos, M. Fernández-ruiz, E. González, J. Cabrera et al.,

E. , H. E. Polanco, N. Hernández, A. Praga, M. Rodriguez-antolín et al., Kidney transplant from uncontrolled donation after circulatory death donors maintained by nECMO has long-term outcomes comparable to standard criteria donation after brain death, Am J Transplant, 2018.

I. Ortega-deballon, L. Hornby, and S. D. Shemie, Protocols for uncontrolled donation after circulatory death: a systematic review of international guidelines, practices and transplant outcomes, Crit Care, vol.19, p.268, 2015.

L. W. Van-heurn, D. Talbot, M. L. Nicholson, M. Z. Akhtar, A. I. Sanchez-fructuoso et al.,

B. , Recommendations for donation after circulatory death kidney transplantation in Europe, Transpl Int, vol.29, issue.7, pp.780-789, 2016.

J. Wind, M. Faut, T. C. Van-smaalen, and E. L. Van-heurn, Variability in protocols on donation after circulatory death in Europe, Crit Care, vol.17, issue.5, p.217, 2013.

H. Peters-sengers, J. J. Homan-van-der-heide, M. Heemskerk, T. Berge, I. Ultee et al., Similar 5-Year Estimated Glomerular Filtration Rate Between Kidney Transplants From Uncontrolled and Controlled Donors After Circulatory Death-A Dutch Cohort Study, Transplantation, vol.101, issue.6, pp.1144-1151, 2017.

C. Antoine, F. Brun, A. Tenaillon, and B. Loty, Organ procurement and transplantation from non-heartbeating donors, Nephrol Ther, vol.4, issue.1, pp.5-14, 2007.

C. C. Chen, W. C. Chapman, and D. W. Hanto, Ischemia-reperfusion injury in kidney transplantation, Front Biosci, vol.7, pp.117-151, 2015.

S. Soullier, N. Gayrard, C. Méjean, I. Swarcz, G. Mourad et al.,

, Nephrol Ther, vol.1, issue.5, pp.315-336, 2005.

A. M. Sheridan and J. V. Bonventre, Cell biology and molecular mechanisms of injury in ischemic acute renal failure, Curr Opin Nephrol Hypertens, vol.9, issue.4, pp.427-461, 2000.

C. Ponticelli, Ischaemia-reperfusion injury: a major protagonist in kidney transplantation, Nephrol Dial Transplant, vol.29, issue.6, pp.1134-1174, 2014.

M. Kosieradzki and W. Rowi?ski, Ischemia/reperfusion injury in kidney transplantation: mechanisms and prevention, Transplant Proc, vol.40, issue.10, pp.3279-88, 2008.
DOI : 10.1016/j.transproceed.2008.10.004

N. Chatauret, L. Badet, B. Barrou, and T. Hauet, Ischemia-reperfusion: From cell biology to acute kidney injury, Prog Urol, vol.24, issue.1, pp.4-12, 2014.
DOI : 10.1016/s1166-7087(14)70057-0

URL : https://hal.archives-ouvertes.fr/hal-01930900

M. Salvadori, G. Rosso, and E. Bertoni, Update on ischemia-reperfusion injury in kidney transplantation: Pathogenesis and treatment, World J Transplant, vol.5, issue.2, pp.52-67, 2015.

A. Fung, H. Zhao, B. Yang, Q. Lian, and D. Ma, Ischaemic and inflammatory injury in renal graft from brain death donation: an update review, J Anesth, vol.30, issue.2, pp.307-323, 2016.

J. V. Bonventre and J. M. Weinberg, Recent advances in the pathophysiology of ischemic acute renal failure, J Am Soc Nephrol, vol.14, issue.8, pp.2199-210, 2003.

T. C. Saat, E. K. Van-den-akker, J. N. Ijzermans, F. J. Dor, and R. W. De-bruin, Improving the outcome of kidney transplantation by ameliorating renal ischemia reperfusion injury: lost in translation?, J Transl Med, vol.14, p.20, 2016.

J. Menke, D. Sollinger, B. Schamberger, U. Heemann, and J. Lutz, The effect of ischemia/reperfusion on the kidney graft, Curr Opin Organ Transplant, vol.19, issue.4, pp.395-400, 2014.

N. S. Chandel and G. R. Budinger, The cellular basis for diverse responses to oxygen. Free Radic Biol Med, vol.42, pp.165-74, 2007.

K. A. Nath and S. M. Norby, Reactive oxygen species and acute renal failure, Am J Med, vol.109, issue.8, pp.665-78, 2000.
DOI : 10.1016/s0002-9343(00)00612-4

C. Li and R. M. Jackson, Reactive species mechanisms of cellular hypoxia-reoxygenation injury, Am J Physiol Cell Physiol, vol.282, issue.2, pp.227-268, 2002.

D. N. Granger and P. R. Kvietys, Reperfusion injury and reactive oxygen species: The evolution of a concept, Redox Biol, vol.6, pp.524-51, 2015.

D. Patschan, S. Patschan, and G. A. Müller, Inflammation and microvasculopathy in renal ischemia reperfusion injury, J Transplant, p.764154, 2012.

C. Y. Lu, J. Hartono, M. Senitko, and J. Chen, The inflammatory response to ischemic acute kidney injury: a result of the 'right stuff' in the 'wrong place, Curr Opin Nephrol Hypertens, 2007.

L. Gu, Y. Tao, C. Chen, Y. Ye, X. Xiong et al., Initiation of the inflammatory response after renal ischemia/reperfusion injury during renal transplantation, Int Urol Nephrol, 2018.

A. Kezi?, N. Stajic, and F. Thaiss, Innate Immune Response in Kidney Ischemia/Reperfusion Injury: Potential Target for Therapy, J Immunol Res, p.6305439, 2017.

H. Zhao, J. S. Perez, K. Lu, A. J. George, and D. Ma, Role of Toll-like receptor-4 in renal graft ischemiareperfusion injury, Am J Physiol Renal Physiol, vol.306, issue.8, pp.801-812, 2014.

B. J. Nankivell and D. R. Kuypers, Diagnosis and prevention of chronic kidney allograft loss, Lancet, vol.378, issue.9800, pp.1428-1465, 2011.

H. Zhao, A. Alam, A. P. Soo, A. George, and D. Ma, Ischemia-Reperfusion Injury Reduces Long Term Renal Graft Survival: Mechanism and Beyond, EBioMedicine, vol.28, pp.31-42, 2018.

A. Mella, M. Messina, A. Lavacca, and L. Biancone, Complement cascade and kidney transplantation: The rediscovery of an ancient enemy, World J Transplant, vol.4, issue.3, pp.168-75, 2014.

J. S. Danobeitia, A. Djamali, and L. A. Fernandez, The role of complement in the pathogenesis of renal ischemia-reperfusion injury and fibrosis. Fibrogenesis Tissue Repair, vol.7, p.16, 2014.

P. Cravedi and P. S. Heeger, Complement as a multifaceted modulator of kidney transplant injury, J Clin Invest, vol.124, issue.6, pp.2348-54, 2014.

D. N. Grigoryev, D. I. Cheranova, D. P. Heruth, P. Huang, L. Q. Zhang et al., Meta-analysis of molecular response of kidney to ischemia reperfusion injury for the identification of new candidate genes, BMC Nephrol, vol.14, p.231, 2013.

D. K. De-vries, L. G. Wijermars, M. E. Reinders, J. H. Lindeman, and A. F. Schaapherder, Donor pre-treatment in clinical kidney transplantation: a critical appraisal, Clin Transplant, vol.27, issue.6, pp.799-808, 2013.

B. Schröppel and C. Legendre, Delayed kidney graft function: from mechanism to translation, Kidney Int, vol.86, issue.2, pp.251-259, 2014.

N. Perico, D. Cattaneo, M. H. Sayegh, and G. Remuzzi, Delayed graft function in kidney transplantation, Lancet, vol.364, issue.9447, pp.1814-1841, 2004.

D. Rosa, S. Antonelli, M. Ronco, and C. , Hypothermia and kidney: a focus on ischaemia-reperfusion injury, Nephrol Dial Transplant, vol.32, issue.2, pp.241-247, 2017.

B. Barrou, N. Chatauret, T. Hauet, R. Thuret, F. Kleinclauss et al.,

L. Badet, Ischemia-reperfusion. Preservation solution and hypothermic machine perfusion
URL : https://hal.archives-ouvertes.fr/inserm-01057473

, Prog Urol, vol.26, issue.15, pp.964-976, 2016.

J. P. Faure, T. Hauet, Z. Han, J. M. Goujon, I. Petit et al.,

V. , Polyethylene glycol reduces early and long-term cold ischemia-reperfusion and renal medulla injury, J Pharmacol Exp Ther, vol.302, issue.3, pp.861-70, 2002.

S. Giraud, R. Codas, T. Hauet, M. Eugene, and L. Badet, Polyethylene glycols and organ protection against I/R injury, Prog Urol, vol.24, issue.1, pp.37-43, 2014.
DOI : 10.1016/s1166-7087(14)70062-4

URL : https://hal.archives-ouvertes.fr/hal-01930332

F. Catena, F. Coccolini, G. Montori, C. Vallicelli, A. Amaduzzi et al., Kidney preservation: review of present and future perspective, Transplant Proc, vol.45, issue.9, pp.3170-3177, 2013.

X. Wang, M. Xu, J. Jia, Z. Zhang, J. P. Gaut et al., CD47 blockade reduces ischemia/reperfusion injury in donation after cardiac death rat kidney transplantation, Am J Transplant, vol.18, issue.4, pp.843-854, 2018.
DOI : 10.1111/ajt.14523

URL : https://onlinelibrary.wiley.com/doi/pdf/10.1111/ajt.14523

N. M. Jager, F. Poppelaars, M. R. Daha, and M. A. Seelen, Complement in renal transplantation: The road to translation, Mol Immunol, vol.89, pp.22-35, 2017.

T. Saint-yves, P. O. Delpech, G. S. Thuillier, R. Hauet, and T. , Additives to preservation solutions
URL : https://hal.archives-ouvertes.fr/hal-01930329

, Prog Urol, vol.24, issue.1, pp.31-37, 2014.

Z. X. Yu, S. Qi, M. A. Lasaro, K. Bouchard, C. Dow et al., Targeting Complement Pathways During Cold Ischemia and Reperfusion Prevents Delayed Graft Function, Am J Transplant, vol.16, issue.9, pp.2589-97, 2016.

P. O. Delpech, R. Thuillier, T. Saintyves, J. Danion, L. Pape et al., Inhibition of complement improves graft outcome in a pig model of kidney autotransplantation, J Transl Med, vol.14, issue.1, p.277, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01928671

J. Cau, F. Favreau, K. Zhang, G. Febrer, G. R. De-la-motte et al., FR167653 improves renal recovery and decreases inflammation and fibrosis after renal ischemia reperfusion injury, J Vasc Surg, vol.49, issue.3, pp.728-768, 2009.
DOI : 10.1016/j.jvs.2008.09.056

URL : https://doi.org/10.1016/j.jvs.2008.09.056

J. T. Powell, D. S. Tsapepas, S. T. Martin, M. A. Hardy, and L. E. Ratner, Managing renal transplant ischemia reperfusion injury: novel therapies in the pipeline, Clin Transplant, vol.27, issue.4, pp.484-91, 2013.

S. O'neill, J. A. Ross, S. J. Wigmore, and E. M. Harrison, The role of heat shock protein 90 in modulating ischemia-reperfusion injury in the kidney, Expert Opin Investig Drugs, vol.21, issue.10, pp.1535-1583, 2012.

C. A. Farrar, B. Keogh, W. Mccormack, O. Shaughnessy, A. Parker et al., Inhibition of TLR2 promotes graft function in a murine model of renal transplant ischemia-reperfusion injury, FASEB J, vol.26, issue.2, pp.799-807, 2012.

E. K. Van-den-akker, F. J. Dor, J. N. Ijzermans, and R. W. De-bruin, MicroRNAs in Kidney Transplantation: Living up to Their Expectations?, J Transplant, p.354826, 2015.

W. Wystrychowski, G. Wystrychowski, E. Zukowska-szczechowska, E. Obuchowicz, W. Grzeszczak et al., Nephroprotective effect of pentoxifylline in renal ischemiareperfusion in rat depends on the timing of its administration, Transplant Proc, vol.46, issue.8, pp.2555-2562, 2014.

Y. Lin, P. T. Manning, J. Jia, J. P. Gaut, Z. Xiao et al., CD47 blockade reduces ischemia-reperfusion injury and improves outcomes in a rat kidney transplant model, Transplantation, vol.98, issue.4, pp.394-401, 2014.
DOI : 10.1097/tp.0000000000000252

URL : http://europepmc.org/articles/pmc4887281?pdf=render

X. Qiao, R. S. Li, H. Li, G. Z. Zhu, X. G. Huang et al., Intermedin protects against renal ischemia-reperfusion injury by inhibition of oxidative stress, Am J Physiol Renal Physiol, vol.304, issue.1, pp.112-121, 2013.
DOI : 10.1152/ajprenal.00054.2012

N. Ambrosi, D. Guerrieri, F. Caro, F. Sanchez, G. Haeublein et al.,

, Alpha Lipoic Acid: A Therapeutic Strategy that Tend to Limit the Action of Free Radicals in Transplantation, Int J Mol Sci, vol.19, issue.1, p.102, 2018.

C. Zou, H. Hu, X. Xi, Z. Shi, G. Wang et al., Pioglitazone protects against renal ischemiareperfusion injury by enhancing antioxidant capacity, J Surg Res, vol.184, issue.2, pp.1092-1097, 2013.

S. A. Jin, S. K. Kim, H. J. Seo, J. Y. Jeong, K. T. Ahn et al., Beneficial Effects of Necrosis Modulator, Indole Derivative NecroX-7, on Renal Ischemia-Reperfusion Injury in Rats, Transplant Proc, vol.48, issue.1, pp.199-204, 2016.

A. Mehrabi, M. Zha, M. Sadeghi, B. M. Schmied, S. A. Müller et al.,

, Thymoglobulin and ischemia reperfusion injury in kidney and liver transplantation, Nephrol Dial Transplant, issue.8, pp.54-60, 2007.

S. Tillet, S. Giraud, P. O. Delpech, R. Thuillier, V. Ameteau et al.,

T. Hauet and G. Mauco, Kidney graft outcome using an anti-Xa therapeutic strategy in an experimental model of severe ischaemia-reperfusion injury, Br J Surg, vol.102, issue.1, pp.132-174, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01928106

P. H. Wang, T. T. Schwindt, G. F. Barnabé, F. L. Motta, P. Semedo et al., Administration of neural precursor cells ameliorates renal ischemia-reperfusion injury, Nephron Exp Nephrol, vol.112, issue.1, pp.20-28, 2009.

A. Bagul, J. H. Frost, and M. Drage, Stem cells and their role in renal ischaemia reperfusion injury, Am J Nephrol, vol.37, issue.1, pp.16-29, 2013.

P. Rowart, P. Erpicum, O. Detry, L. Weekers, C. Grégoire et al., Mesenchymal Stromal Cell Therapy in Ischemia/Reperfusion Injury, J Immunol Res, p.602597, 2015.

J. M. Sierra-parraga, M. Eijken, J. Hunter, C. Moers, H. Leuvenink et al., Mesenchymal Stromal Cells as Anti-Inflammatory and Regenerative Mediators for Donor Kidneys During Normothermic Machine Perfusion. Stem Cells Dev, vol.26, pp.1162-1170, 2017.

S. A. Hosgood, E. Van-heurn, and M. L. Nicholson, Normothermic machine perfusion of the kidney: better conditioning and repair? Transpl Int, vol.28, pp.657-64, 2015.

R. Thuillier, D. Dutheil, M. T. Trieu, V. Mallet, G. Allain et al.,

F. and H. T. , Supplementation with a new therapeutic oxygen carrier reduces chronic fibrosis and organ dysfunction in kidney static preservation, Am J Transplant, vol.11, issue.9, pp.1845-60, 2011.

G. J. Dugbartey, H. R. Bouma, M. N. Saha, I. Lobb, R. H. Henning et al., A Hibernation-Like State for Transplantable Organs: Is Hydrogen Sulfide Therapy the Future of Organ Preservation?, Antioxid Redox Signal, vol.28, issue.16, pp.1503-1515, 2018.

I. Lobb, A. Mok, Z. Lan, W. Liu, B. Garcia et al., Supplemental hydrogen sulphide protects transplant kidney function and prolongs recipient survival after prolonged cold ischaemia-reperfusion injury by mitigating renal graft apoptosis and inflammation, BJU Int, vol.110, issue.11, pp.1187-95, 2012.

J. X. Zhu, M. Kalbfleisch, Y. X. Yang, R. Bihari, I. Lobb et al., Detrimental effects of prolonged warm renal ischaemia-reperfusion injury are abrogated by supplemental hydrogen sulphide: an analysis using real-time intravital microscopy and polymerase chain reaction, BJU Int, vol.110, issue.11, pp.1218-1245, 2012.

A. C. Van-erp, L. Van-dullemen, R. J. Ploeg, and H. Leuvenink, Systematic review on the treatment of deceased organ donors. Transplant Rev (Orlando), p.955, 2018.

K. Stokfisz, A. Ledakowicz-polak, M. Zagorski, and M. Zielinska, Ischaemic preconditioning -Current knowledge and potential future applications after 30 years of experience, Adv Med Sci, 2017.

S. J. Jonker, T. P. Menting, M. C. Warlé, M. Ritskes-hoitinga, and K. E. Wever, Preclinical Evidence for the Efficacy of Ischemic Postconditioning against Renal Ischemia-Reperfusion Injury, a Systematic Review and Meta-Analysis, PLoS One, vol.11, issue.3, p.150863, 2016.

R. Macallister, T. Clayton, R. Knight, S. Robertson, J. Nicholas et al., REmote preconditioning for Protection Against Ischaemia-Reperfusion in renal transplantation (REPAIR): a multicentre, multinational, double-blind, factorial designed randomised controlled trial, 2015.

R. Thuret, Y. T. Saint, X. Tillou, N. Chatauret, R. Thuillier et al., Ischemic preand post-conditioning: current clinical applications, Prog Urol, vol.24, issue.1, pp.56-61, 2014.

R. P. Jia, J. J. Xie, F. Y. Luo, and J. G. Zhu, Ischemic preconditioning improves rat kidney allograft function after ischemia/reperfusion injury: the role of tumor necrosis factor-alpha, Transplant Proc, vol.40, issue.10, pp.3316-3336, 2008.

N. V. Krogstrup, M. Oltean, B. M. Bibby, G. J. Nieuwenhuijs-moeke, F. J. Dor et al.,

, Remote ischaemic conditioning on recipients of deceased renal transplants, effect on immediate and extended kidney graft function: a multicentre, randomised controlled trial protocol (CONTEXT), BMJ Open, vol.5, issue.8, p.7941, 2015.

N. V. Krogstrup, M. Oltean, G. J. Nieuwenhuijs-moeke, F. J. Dor, U. Møldrup et al., Remote Ischemic Conditioning on Recipients of Deceased Renal Transplants Does Not Improve Early Graft Function: A Multicenter Randomized, Controlled Clinical Trial, Am J Transplant, vol.17, issue.4, pp.1042-1049, 2017.

M. Z. Akhtar, A. I. Sutherland, H. Huang, R. J. Ploeg, and C. W. Pugh, The role of hypoxia-inducible factors in organ donation and transplantation: the current perspective and future opportunities, Am J Transplant, vol.14, issue.7, pp.1481-1488, 2014.

F. J. Burgos-revilla, V. Hevia, V. Diez, D. Carracedo, A. Gomis et al.,

, Machine perfusion: initial results in an expanded criteria donor kidney transplant program, Transplant Proc, vol.47, issue.1, pp.19-22, 2015.

S. A. Hosgood, E. Thompson, T. Moore, C. H. Wilson, and M. L. Nicholson, Normothermic machine perfusion for the assessment and transplantation of declined human kidneys from donation after circulatory death donors, Br J Surg, vol.105, issue.4, pp.388-394, 2018.

I. Jochmans, J. M. O'callaghan, J. Pirenne, and R. J. Ploeg, Hypothermic machine perfusion of kidneys retrieved from standard and high-risk donors, Transpl Int, vol.28, issue.6, pp.665-76, 2015.

O. N. Reznik, S. F. Bagnenko, I. V. Loginov, V. A. Iljina, A. N. Ananyev et al.,

, Machine perfusion as a tool to select kidneys recovered from uncontrolled donors after cardiac death, Transplant Proc, vol.40, issue.4, pp.1023-1029, 2008.

E. E. De-vries, E. R. Hoogland, B. Winkens, M. G. Snoeijs, and L. W. Van-heurn, Renovascular resistance of machine-perfused DCD kidneys is associated with primary nonfunction, Am J Transplant, 2011.

J. M. Kaths, A. Paul, L. A. Robinson, and M. Selzner, Ex vivo machine perfusion for renal graft preservation, Transplant Rev (Orlando), vol.32, issue.1, pp.1-9, 2018.

A. M. Hameed, H. C. Pleass, G. Wong, and W. J. Hawthorne, Maximizing kidneys for transplantation using machine perfusion: from the past to the future: A comprehensive systematic review and metaanalysis. Medicine (Baltimore), vol.95, p.5083, 2016.

L. Martínez-arcos, F. Alcañiz, J. J. , G. Santos, V. et al., Functional Results of Renal Preservation in Hypothermic Pulsatile Machine Perfusion Versus Cold Preservation: Systematic Review and Meta-Analysis of Clinical Trials, Transplant Proc, vol.50, issue.1, pp.24-32, 2018.

R. M. Cannon, G. N. Brock, R. N. Garrison, J. W. Smith, M. R. Marvin et al., To pump or not to pump: a comparison of machine perfusion vs cold storage for deceased donor kidney transplantation, J Am Coll Surg, vol.216, issue.4, pp.625-658, 2013.

W. Wang, D. Xie, X. Hu, H. Yin, H. Liu et al., Effect of Hypothermic Machine Perfusion on the Preservation of Kidneys Donated After Cardiac Death: A Single-Center, Randomized, Controlled Trial, Artif Organs, vol.41, issue.8, pp.753-758, 2017.

C. Moers, J. M. Smits, M. H. Maathuis, J. Treckmann, F. Van-gelder et al.,

M. Kutz, J. J. Van-der-heide, J. P. Squifflet, E. Van-heurn, G. R. Kirste et al., Machine perfusion or cold storage in deceased-donor kidney transplantation, N Engl J Med, vol.360, issue.1, pp.7-19, 2009.

J. C. Forde, W. P. Shields, M. Azhar, P. J. Daly, J. A. Zimmermann et al.,

P. Mohan, D. P. Hickey, and D. M. Little, Single centre experience of hypothermic machine perfusion of kidneys from extended criteria deceased heart-beating donors: a comparative study, Ir J Med Sci, 2016.

B. Jiao, S. Liu, H. Liu, D. Cheng, Y. Cheng et al., Hypothermic machine perfusion reduces delayed graft function and improves one-year graft survival of kidneys from expanded criteria donors: a meta-analysis, PLoS One, vol.8, issue.12, p.81826, 2013.

Z. Zhong, J. Lan, S. Ye, Z. Liu, L. Fan et al.,

, Outcome Improvement for Hypothermic Machine Perfusion Versus Cold Storage for Kidneys From Cardiac Death Donors. Artif Organs, vol.41, pp.647-653, 2017.

L. Yao, H. Zhou, Y. Wang, G. Wang, W. Wang et al., Hypothermic Machine Perfusion in DCD Kidney Transplantation: A Single Center Experience, Urol Int, vol.96, issue.2, pp.148-51, 2016.

X. Matillon, F. Danjou, P. Petruzzo, O. Thaunat, T. Rimmele et al., Hypothermic pulsatile preservation of kidneys from uncontrolled deceased donors after cardiac arrest -a retrospective study, Transpl Int, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01911338

D. P. Hoyer, A. Gallinat, S. Swoboda, J. Wohlschläger, U. Rauen et al.,

, Transpl Int, vol.27, issue.10, pp.1097-106, 2014.

M. F. Blum, Q. Liu, B. Soliman, P. Dreher, T. Okamoto et al., Comparison of normothermic and hypothermic perfusion in porcine kidneys donated after cardiac death, J Surg Res, vol.216, pp.35-45, 2017.

M. L. Nicholson and S. A. Hosgood, Renal transplantation after ex vivo normothermic perfusion: the first clinical study, Am J Transplant, vol.13, issue.5, pp.1246-52, 2013.

X. Tillou, R. Thuret, A. Doerfler, and . Ctafu, Ischemia/reperfusion during normothermic perfusion
DOI : 10.1016/s1166-7087(14)70064-8

, Prog Urol, vol.24, issue.1, pp.51-56, 2014.

H. Parissis, S. Mbarushimana, B. C. Ramesh, M. Parissis, S. Lampridis et al., The impact of off-pump surgery in end-organ function: practical end-points, J Cardiothorac Surg, 2015.

, Nov, vol.10, p.159

D. Somer and F. , Optimization of the perfusion circuit and its possible impact on the inflammatory response, J Extra Corpor Technol, vol.39, issue.4, pp.285-293, 2007.

S. G. Raja and G. A. Berg, Impact of off-pump coronary artery bypass surgery on systemic inflammation: current best available evidence, J Card Surg, vol.22, issue.5, pp.445-55, 2007.

K. G. Shann, D. S. Likosky, J. M. Murkin, R. A. Baker, Y. R. Baribeau et al., An evidence-based review of the practice of cardiopulmonary bypass in adults: a focus on neurologic injury, glycemic control, hemodilution, and the inflammatory response, J Thorac Cardiovasc Surg, 2006.

O. J. Warren, A. J. Smith, C. Alexiou, P. L. Rogers, N. Jawad et al., The inflammatory response to cardiopulmonary bypass: part 1--mechanisms of pathogenesis, J Cardiothorac Vasc Anesth, vol.23, issue.2, pp.223-254, 2009.

T. Gourlay, Biomaterial development for cardiopulmonary bypass, Perfusion, 2001.
DOI : 10.1177/026765910101600508

G. J. Peek and R. K. Firmin, The inflammatory and coagulative response to prolonged extracorporeal membrane oxygenation, ASAIO J, vol.45, issue.4, pp.250-63, 1999.

R. Ben-abraham, A. A. Weinbroum, B. Dekel, and G. Paret, Chemokines and the inflammatory response following cardiopulmonary bypass--a new target for therapeutic intervention?--A review

, Paediatr Anaesth, vol.13, issue.8, pp.655-61, 2003.

J. E. Millar, J. P. Fanning, C. I. Mcdonald, D. F. Mcauley, and J. F. Fraser, The inflammatory response to extracorporeal membrane oxygenation (ECMO): a review of the pathophysiology, Crit Care, 2016.

, Nov, vol.28, issue.1, p.387

A. B. Kumar and M. Suneja, Cardiopulmonary bypass-associated acute kidney injury, Anesthesiology, vol.114, issue.4, pp.964-70, 2011.

N. G. Baikoussis, N. A. Papakonstantinou, and E. Apostolakis, The "benefits" of the mini-extracorporeal circulation in the minimal invasive cardiac surgery era, J Cardiol, vol.63, issue.6, pp.391-397, 2014.

C. I. Mcdonald, J. F. Fraser, J. S. Coombes, and Y. L. Fung, Oxidative stress during extracorporeal circulation, Eur J Cardiothorac Surg, vol.46, issue.6, pp.937-980, 2014.

S. Boodram and E. Evans, Use of leukocyte-depleting filters during cardiac surgery with cardiopulmonary bypass: a review, J Extra Corpor Technol, vol.40, issue.1, pp.27-42, 2008.

C. S. Ng and S. Wan, Limiting inflammatory response to cardiopulmonary bypass: pharmaceutical strategies, Curr Opin Pharmacol, vol.12, issue.2, pp.155-164, 2012.
DOI : 10.1016/j.coph.2012.01.007

A. Salameh and S. Dhein, Strategies for Pharmacological Organoprotection during Extracorporeal Circulation Targeting Ischemia-Reperfusion Injury. Front Pharmacol, vol.6, p.296, 2015.
DOI : 10.3389/fphar.2015.00296

URL : https://www.frontiersin.org/articles/10.3389/fphar.2015.00296/pdf

R. C. Landis, J. R. Brown, D. Fitzgerald, D. S. Likosky, L. Shore-lesserson et al.,

, Attenuating the Systemic Inflammatory Response to Adult Cardiopulmonary Bypass: A Critical Review of the Evidence Base, J Extra Corpor Technol, vol.46, issue.3, pp.197-211, 2014.

O. A. Jarral, S. Saso, L. Harling, H. Ashrafian, H. Naase et al., Organ dysfunction in patients with left ventricular impairment: what is the effect of cardiopulmonary bypass? Heart Lung Circ, vol.23, pp.852-62, 2014.

D. Fudulu, U. Benedetto, G. G. Pecchinenda, P. Chivasso, V. D. Bruno et al., Current outcomes of off-pump versus on-pump coronary artery bypass grafting: evidence from randomized controlled trials, J Thorac Dis, vol.8, issue.10, pp.758-771, 2016.

Y. C. Chen, F. C. Tsai, J. T. Fang, and C. W. Yang, Acute kidney injury in adults receiving extracorporeal membrane oxygenation, J Formos Med Assoc, vol.113, issue.11, pp.778-85, 2014.
DOI : 10.1016/j.jfma.2014.04.006

URL : https://doi.org/10.1016/j.jfma.2014.04.006

D. J. Askenazi, D. T. Selewski, M. L. Paden, D. S. Cooper, B. C. Bridges et al., Renal replacement therapy in critically ill patients receiving extracorporeal membrane oxygenation

, Clin J Am Soc Nephrol, vol.7, issue.8, pp.1328-1364, 2012.

M. Ranucci, Perioperative renal failure: hypoperfusion during cardiopulmonary bypass? Semin Cardiothorac Vasc Anesth, vol.11, pp.265-273, 2007.
DOI : 10.1177/1089253207311141

H. Yimin, Y. Wenkui, S. Jialiang, C. Qiyi, S. Juanhong et al.,

L. Jieshou, Effects of continuous renal replacement therapy on renal inflammatory cytokines during extracorporeal membrane oxygenation in a porcine model, J Cardiothorac Surg, vol.8, p.113, 2013.

G. Villa, N. Katz, and C. Ronco, Extracorporeal Membrane Oxygenation and the Kidney, Cardiorenal Med, vol.6, issue.1, pp.50-60, 2015.

D. J. Kilburn, K. Shekar, and J. F. Fraser, The Complex Relationship of Extracorporeal Membrane Oxygenation and Acute Kidney Injury: Causation or Association?, Biomed Res Int, p.1094296, 2016.

D. Tomasso, N. Monaco, F. Landoni, and G. , Hepatic and renal effects of cardiopulmonary bypass

, Best Pract Res Clin Anaesthesiol, vol.29, issue.2, pp.151-61, 2015.

J. B. O'neal, A. D. Shaw, F. T. Billings, and . 4th, Acute kidney injury following cardiac surgery: current understanding and future directions, Crit Care, vol.20, issue.1, p.187, 2016.

Y. Abu-omar and C. Ratnatunga, Cardiopulmonary bypass and renal injury, Perfusion, 2006.
DOI : 10.1191/0267659106pf870oa

S. Steen, Q. Liao, L. Pierre, A. Paskevicius, and T. Sjöberg, Evaluation of LUCAS, a new device for automatic mechanical compression and active decompression resuscitation, Resuscitation, 2002.

J. K. Lunney, C. Ho, M. Wysocki, and D. M. Smith, Molecular genetics of the swine major histocompatibility complex, the SLA complex, Dev Comp Immunol, vol.33, pp.362-374, 2009.

. Thuret-rodolphe, Intérêt de la prise en charge des donneurs de reins décédés par arrêt cardiaque : développement des méthodes de Gillot et de CRN, 2013.

P. Chardon, C. Renard, C. R. Gaillard, and M. Vaiman, The porcine major histocompatibility complex and related paralogous regions: a review, Genet Sel Evol, vol.32, issue.2, pp.109-137, 2000.
DOI : 10.1186/1297-9686-32-2-109

URL : https://hal.archives-ouvertes.fr/hal-00894302

S. L. Lindell, H. Muir, J. Brassil, and M. J. Mangino, Hypothermic Machine Perfusion Preservation of the DCD Kidney: Machine Effects, J Transplant, p.802618, 2013.
DOI : 10.1155/2013/802618

URL : http://downloads.hindawi.com/journals/jtrans/2013/802618.pdf

I. Jochmans, M. Z. Akhtar, D. Nasralla, P. Kocabayoglu, C. Boffa et al., Past, Present, and Future of Dynamic Kidney and Liver Preservation and Resuscitation, Am J Transplant, vol.16, issue.9, pp.2545-55, 2016.

R. Deng, G. Gu, D. Wang, Q. Tai, L. Wu et al., Machine perfusion versus cold storage of kidneys derived from donation after cardiac death: a meta-analysis, PLoS One, vol.8, issue.3, p.56368, 2013.

J. C. Bains, R. M. Sandford, N. R. Brook, S. A. Hosgood, G. R. Lewis et al., Comparison of renal allograft fibrosis after transplantation from heart-beating and non-heart-beating donors, Br J Surg, vol.92, issue.1, pp.113-121, 2005.

G. Kootstra and E. Van-heurn, Non-heartbeating donation of kidneys for transplantation, Nat Clin Pract Nephrol, vol.3, issue.3, pp.154-63, 2007.

H. Peters-sengers, J. J. Homan-van-der-heide, M. Heemskerk, T. Berge, I. Ultee et al., Similar 5-Year Estimated Glomerular Filtration Rate Between Kidney Transplants From Uncontrolled and Controlled Donors After Circulatory Death-A Dutch Cohort Study, Transplantation, vol.101, issue.6, pp.1144-1151, 2017.

N. R. Brook, J. R. Waller, and M. L. Nicholson, Nonheart-beating kidney donation: current practice and future developments, Kidney Int, vol.63, issue.4, pp.1516-1545, 2003.
DOI : 10.1046/j.1523-1755.2003.00854.x

URL : https://doi.org/10.1046/j.1523-1755.2003.00854.x

E. R. Hoogland, M. G. Snoeijs, B. Winkens, M. H. Christaans, and L. W. Van-heurn, Kidney transplantation from donors after cardiac death: uncontrolled versus controlled donation, Am J Transplant, 2011.
DOI : 10.1111/j.1600-6143.2011.03562.x

M. A. Gentil, P. Castro-de-la-nuez, C. Gonzalez-corvillo, M. C. De-gracia, and M. Cabello,

. Ma, A. Rodriguez-benot, L. Ballesteros, A. Osuna, and M. Alonso, Non-Heart-Beating Donor Kidney Transplantation Survival Is Similar to Donation After Brain Death: Comparative Study With Controls in a Regional Program, Transplant Proc, vol.48, issue.9, pp.2867-2870, 2016.

A. Sánchez-fructuoso, P. Sánchez, D. , M. Vidas, M. et al.,

A. Guzmán, Non-heart beating donors, Nephrol Dial Transplant, issue.3, pp.26-31, 2004.

S. N. Heyman, M. Khamaisi, S. Rosen, and C. Rosenberger, Renal parenchymal hypoxia, hypoxia response and the progression of chronic kidney disease, Am J Nephrol, vol.28, issue.6, pp.998-1006, 2008.

W. M. Bernhardt, C. Warnecke, C. Willam, T. Tanaka, M. S. Wiesener et al., Organ protection by hypoxia and hypoxia-inducible factors, Methods Enzymol, vol.435, pp.221-266, 2007.

B. Jiao, S. Liu, H. Liu, D. Cheng, Y. Cheng et al., Hypothermic machine perfusion reduces delayed graft function and improves one-year graft survival of kidneys from expanded criteria donors: a meta-analysis, PLoS One, vol.8, issue.12, p.81826, 2013.

M. Matignon, F. Bonnefoy, P. Lang, and P. Grimbert,

, Transfus Clin Biol, vol.18, issue.2, pp.70-78, 2011.

D. A. Munguía-canales, C. De-la-cruz, J. L. Crowley-carrasco, S. Lazo-pérez, D. Macías-sotuela et al., Lung transplantation with uncontrolled non-heart-beating donor, Review. Cir Cir, vol.80, issue.1, pp.86-91, 2012.

P. E. Morrissey and A. P. Monaco, Donation after circulatory death: current practices, ongoing challenges, and potential improvements, Transplantation, vol.97, issue.3, pp.258-64, 2014.

M. E. Erasmus, D. Van-raemdonck, M. Z. Akhtar, A. Neyrinck, D. G. De-antonio et al.,

, DCD lung donation: donor criteria, procedural criteria, pulmonary graft function validation, and preservation, Transpl Int, vol.29, issue.7, pp.790-797, 2016.

D. S. Demos, A. Iyengar, B. S. Bryner, B. W. Gray, H. R. Hoffman et al.,

. De, R. H. Bartlett, J. D. Punch, and A. Rojas-peña, Successful Porcine Renal Transplantation After 60 Minutes of Donor Warm Ischemia: Extracorporeal Perfusion and Thrombolytics, ASAIO J, vol.61, issue.4, pp.474-483, 2015.

O. Reznik, A. Skvortsov, I. Loginov, A. Ananyev, S. Bagnenko et al., Kidney from uncontrolled donors after cardiac death with one hour warm ischemic time: resuscitation by extracorporal normothermic abdominal perfusion "in situ" by leukocytes-free oxygenated blood, Clin Transplant, vol.25, issue.4, pp.511-517, 2011.

O. N. Reznik, A. E. Skvortsov, A. O. Reznik, A. N. Ananyev, A. P. Tutin et al., Uncontrolled donors with controlled reperfusion after sixty minutes of asystole: a novel reliable resource for kidney transplantation, PLoS One, vol.8, issue.5, p.64209, 2013.

S. David, Mise au point et évaluation d'un modèle porcin de donneur rénal DDAC-LATA ou Maastricht 3, 2018.

C. Van-de-wauwer, E. A. Verschuuren, W. Van-der-bij, G. D. Nossent, and M. E. Erasmus, The use of non-heart-beating lung donors category III can increase the donor pool, Eur J Cardiothorac Surg, 2011.

I. Inci, S. Hillinger, D. Schneiter, I. Opitz, M. Schuurmans et al., Lung Transplantation with Controlled Donation after Circulatory Death Donors, Ann Thorac Cardiovasc Surg, 2018.

S. Osaki, M. R. Locher, E. B. Lushaj, S. A. Akhter, and T. Kohmoto, Functional evaluation of human donation after cardiac death donor hearts using a continuous isolated myocardial perfusion technique: Potential for expansion of the cardiac donor population, J Thorac Cardiovasc Surg, 2014.

K. K. Dhital, A. Iyer, M. Connellan, H. C. Chew, L. Gao et al.,

C. , D. A. Cartwright, B. Nair, P. Granger, E. Jansz et al.,

R. Graham, P. Spratt, and P. Macdonald, Adult heart transplantation with distant procurement and ex-vivo preservation of donor hearts after circulatory death: a case series, Lancet, vol.385, issue.9987, pp.2585-91, 2015.

S. Messer, A. Page, R. Axell, M. Berman, J. Hernández-sánchez et al.,

C. Lewis, A. Kydd, D. Jenkins, C. J. Watson, C. Sudarshan et al., Outcome after heart transplantation from donation after circulatory-determined death donors, J Heart Lung Transplant, vol.36, issue.12, pp.1311-1318, 2017.

S. Tsui and G. C. Oniscu, Extending normothermic regional perfusion to the thorax in donors after circulatory death. Curr Opin Organ Transplant, vol.22, pp.245-250, 2017.

C. W. White, S. J. Messer, S. R. Large, J. Conway, D. H. Kim et al., Transplantation of Hearts Donated after Circulatory Death. Front Cardiovasc Med, vol.5, p.8, 2018.