R. I. Lechler, M. Sykes, A. W. Thomson, and L. A. Turka, Organ transplantationhow much of the promise has been realized, Nat Med, vol.11, pp.605-618, 2005.

L. A. Turka and R. I. Lechler, Towards the identification of biomarkers of transplantation tolerance, Nat Rev Immunol, vol.9, pp.521-527, 2009.

C. M. Lin and R. G. Gill, Direct and indirect allograft recognition: pathways dictating graft rejection mechanisms, Curr Opin Organ Transplant, vol.21, pp.40-44, 2016.

W. Liu and X. C. Li, An overview on non-T cell pathways in transplant rejection and tolerance, Curr Opin Organ Transplant, vol.15, pp.422-428, 2010.

D. Scozzi, M. Ibrahim, C. Menna, A. S. Krupnick, D. Kreisel et al., The role of neutrophils in transplanted organs, Am J Transplant, vol.17, pp.328-363, 2017.

M. Goldman, L. Moine, A. Braun, M. Flamand, V. Abramowicz et al., A role for eosinophils in transplant rejection, Trends Immunol, vol.22, pp.247-51, 2001.

A. S. Chong and M. L. Alegre, The impact of infection and tissue damage in solid-organ transplantation, Nat Rev Immunol, vol.12, pp.459-71, 2012.

N. D. Jones, S. E. Turvey, A. Van-maurik, M. Hara, C. I. Kingsley et al., Differential susceptibility of heart, skin, and islet allografts to T cell-mediated rejection, J Immunol, vol.166, pp.2824-2854, 2001.

G. Benichou, Y. Yamada, S. H. Yun, C. Lin, M. Fray et al., Immune recognition and rejection of allogeneic skin grafts, Immunotherapy, vol.3, pp.757-70, 2011.

T. Hautz, D. Wolfram, J. Grahammer, R. Starzl, C. Krapf et al., Mechanisms and mediators of inflammation: potential models for skin rejection and targeted therapy in vascularized composite allotransplantation, Clin Dev Immunol, p.757310, 2012.

M. Metz, F. Siebenhaar, and M. Maurer, Mast cell functions in the innate skin immune system, Immunobiology, vol.213, pp.251-60, 2008.

S. N. Abraham and A. L. St-john, Mast cell-orchestrated immunity to pathogens, Nat Rev Immunol, vol.10, pp.440-52, 2010.

F. A. Redegeld, Y. Yu, S. Kumari, N. Charles, and U. Blank, Non-IgE mediated mast cell activation, Immunol Rev, vol.282, pp.87-113, 2018.

K. Mukai, M. Tsai, P. Starkl, T. Marichal, and S. J. Galli, IgE and mast cells in host defense against parasites and venoms, Semin Immunopathol, vol.38, pp.581-603, 2016.

J. Jahanyar, M. M. Koerner, M. Loebe, K. A. Youker, G. Torre-amione et al., The role of mast cells after solid organ transplantation, Transplantation, vol.85, pp.1365-71, 2008.

L. F. Lu, E. F. Lind, D. C. Gondek, K. A. Bennett, M. W. Gleeson et al., Mast cells are essential intermediaries in regulatory T-cell tolerance, Nature, vol.442, pp.997-1002, 2006.

V. C. De-vries, K. Pino-lagos, E. C. Nowak, K. A. Bennett, C. Oliva et al., Mast cells condition dendritic cells to mediate allograft tolerance, Immunity, vol.35, pp.550-61, 2011.

E. C. Nowak, V. C. De-vries, A. Wasiuk, C. Ahonen, K. A. Bennett et al., Tryptophan hydroxylase-1 regulates immune tolerance and inflammation, J Exp Med, vol.209, pp.2127-2162, 2012.

M. Boerma, W. P. Fiser, G. Hoyt, G. J. Berry, L. Joseph et al., Influence of mast cells on outcome after heterotopic cardiac transplantation in rats

, Transpl Int, vol.20, pp.256-65, 2007.

S. Itoh, S. Nakae, J. B. Velotta, H. Kosuge, A. Connolly et al., The role of recipient mast cells in acute and chronic cardiac allograft rejection in C57BL/6-KitW-sh/W-sh mice, J Heart Lung Transplant, vol.29, pp.401-410, 2010.

M. Q. Yang, Y. Y. Ma, J. Ding, and J. Y. Li, The role of mast cells in ischemia and reperfusion injury, Inflamm Res, vol.63, pp.899-905, 2014.

U. Blank, I. K. Madera-salcedo, L. Danelli, J. Claver, N. Tiwari et al., Vesicular trafficking and signaling for cytokine and chemokine secretion in mast cells, Front Immunol, vol.5, p.453, 2014.

S. Wernersson and G. Pejler, Mast cell secretory granules: armed for battle, Nat Rev Immunol, vol.14, pp.478-94, 2014.

J. A. Boyce, Mast cells and eicosanoid mediators: a system of reciprocal paracrine and autocrine regulation, Immunol Rev, vol.217, pp.168-85, 2007.

K. Mukai, M. Tsai, H. Saito, and S. J. Galli, Mast cells as sources of cytokines, chemokines, and growth factors, Immunol Rev, vol.282, pp.121-50, 2018.

E. Simpson, D. Scott, and P. Chandler, The male-specific histocompatibility antigen, H-Y: a history of transplantation, immune response genes, sex determination and expression cloning, Annu Rev Immunol, vol.15, pp.39-61, 1997.

S. Celli, M. L. Albert, and P. Bousso, Visualizing the innate and adaptive immune responses underlying allograft rejection by two-photon microscopy, Nat Med, vol.17, pp.744-753, 2011.
URL : https://hal.archives-ouvertes.fr/pasteur-01402079

D. Zelenika, E. Adams, A. Mellor, E. Simpson, P. Chandler et al., Rejection of H-Y disparate skin grafts by monospecific CD4+ Th1 and Th2 cells: no requirement for CD8+ T cells or B cells, J Immunol, vol.161, pp.1868-74, 1998.

A. Dahdah, G. Gautier, T. Attout, F. Fiore, E. Lebourdais et al., Mast cells aggravate sepsis by inhibiting peritoneal macrophage phagocytosis, J Clin Invest, vol.124, pp.4577-89, 2014.

E. Tchougounova, G. Pejler, and M. Abrink, The chymase, mouse mast cell protease 4, constitutes the major chymotrypsin-like activity in peritoneum and ear tissue. A role for mouse mast cell protease 4 in thrombin regulation and fibronectin turnover, J Exp Med, vol.198, pp.423-454, 2003.

M. Pons, L. Ali, W. Beghdadi, L. Danelli, A. M. Madjene et al., Mast cells and MCPT4 chymase promote renal impairment after partial ureteral obstruction. Front Immunol, vol.8, p.450, 2017.

M. Carpentier, P. Chappert, C. Kuhn, M. Lalfer, H. Flament et al., Extrathymic induction of Foxp3(+) regulatory T cells declines with age in a T-cell intrinsic manner, Eur J Immunol, vol.43, pp.2598-604, 2013.
URL : https://hal.archives-ouvertes.fr/pasteur-00912507

A. Porcherie, C. Mathieu, R. Peronet, E. Schneider, J. Claver et al., Critical role of the neutrophil-associated high-affinity receptor for IgE in the pathogenesis of experimental cerebral malaria, J Exp Med, vol.208, pp.2225-2261, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00629378

B. Habibi-asl, H. Vaez, T. Imankhah, and S. Hamidi, Impact of caffeine on weight changes due to ketotifen administration, Adv Pharm Bull, vol.4, pp.83-92, 2014.

Y. Muto, Z. Wang, M. Vanderberghe, A. Two, R. L. Gallo et al., Mast cells are key mediators of cathelicidin-initiated skin inflammation in rosacea, J Invest Dermatol, vol.134, pp.2728-2764, 2014.

E. Rossato, B. Mkaddem, S. Kanamaru, Y. Hurtado-nedelec, M. Hayem et al., Reversal of arthritis by human monomeric IgA through the receptor-mediated SH2 domain-containing phosphatase 1 inhibitory pathway, Arthritis Rheumatol, vol.67, pp.1766-77, 2015.

W. Beghdadi, L. C. Madjene, J. Claver, G. Pejler, L. Beaudoin et al., Mast cell chymase protects against renal fibrosis in murine unilateral ureteral obstruction, Kidney Int, vol.84, pp.317-343, 2013.

M. Metz, M. A. Grimbaldeston, S. Nakae, A. M. Piliponsky, M. Tsai et al., Mast cells in the promotion and limitation of chronic inflammation, Immunol Rev, vol.217, pp.304-332, 2007.

H. Shen and D. R. Goldstein, IL-6 and TNF-alpha synergistically inhibit allograft acceptance, J Am Soc Nephrol, vol.20, pp.1032-1072, 2009.

M. S. Chae, J. W. Kim, H. S. Chung, C. S. Park, J. Lee et al., The impact of serum cytokines in the development of early allograft dysfunction in living donor liver transplantation, Medicine, vol.97, p.400, 2018.

P. Pribylova-hribova, K. Kotsch, A. Lodererova, O. Viklicky, S. Vitko et al., TGF-beta1 mRNA upregulation influences chronic renal allograft dysfunction, Kidney Int, vol.69, pp.1872-1881, 2006.

R. Saluja, A. Zoltowska, M. E. Ketelaar, and G. Nilsson, IL-33 and Thymic Stromal Lymphopoietin in mast cell functions, Eur J Pharmacol, vol.778, pp.68-76, 2016.

Y. Kobayashi, Neutrophil infiltration and chemokines, Crit Rev Immunol, vol.26, pp.307-323, 2006.

V. C. De-vries, A. Wasiuk, K. A. Bennett, M. J. Benson, R. Elgueta et al., Mast cell degranulation breaks peripheral tolerance, Am J Transplant, vol.9, pp.2270-80, 2009.

S. He and A. F. Walls, Human mast cell chymase induces the accumulation of neutrophils, eosinophils and other inflammatory cells in vivo, Br J Pharmacol, vol.125, pp.1491-500, 1998.

K. Tani, F. Ogushi, H. Kido, T. Kawano, Y. Kunori et al., Chymase is a potent chemoattractant for human monocytes and neutrophils, J Leukoc Biol, vol.67, pp.585-594, 2000.

K. J. Wood and R. Goto, Mechanisms of rejection: current perspectives, Transplantation, vol.93, pp.1-10, 2012.

M. H. Oberbarnscheidt, Q. Zeng, Q. Li, H. Dai, A. L. Williams et al., Non-self recognition by monocytes initiates allograft rejection, J Clin Invest, vol.124, pp.3579-89, 2014.

A. Mason, P. L. Mottram, A. Mirisklavos, G. J. Clurne, T. E. Mandel et al., A comparison of cyclosporine, donor-specific transfusion, and antilymphocyte serum suppression of skin, heart, and fetal pancreatic islet allograft rejection in mice, Transplantation, vol.45, pp.285-294, 1988.

I. L. Bernstein, Cromolyn sodium in the treatment of asthma: coming of age in the United States, J Allergy Clin Immunol, vol.76, pp.381-389, 1985.

M. Zweifel, H. Hirsiger, K. Matozan, M. Welle, T. Schaffner et al., Mast cells in ongoing acute rejection: increase in number and expression of a different phenotype in rat heart transplants, Transplantation, vol.73, pp.1707-1723, 2002.

M. Yamada, M. Ueda, T. Naruko, S. Tanabe, Y. S. Han et al., Mast cell chymase expression and mast cell phenotypes in human rejected kidneys, Kidney Int, vol.59, pp.1374-81, 2001.

C. Arikan, D. Nart, M. Kilic, H. A. Yuksekkaya, and S. Aydogdu, Association of mast cells and liver allograft rejection, Pediatr Transplant, vol.12, pp.347-52, 2008.

U. Blank, M. Essig, L. Scandiuzzi, M. Benhamou, and Y. Kanamaru, Mast cells and inflammatory kidney disease, Immunol Rev, vol.217, pp.79-95, 2007.

J. Wang, M. Hossain, A. Thanabalasuriar, M. Gunzer, C. Meininger et al., Visualizing the function and fate of neutrophils in sterile injury and repair, Science, vol.358, pp.111-117, 2017.

A. Y. Hershko, R. Suzuki, N. Charles, D. Alvarez-errico, J. L. Sargent et al., Mast cell interleukin-2 production contributes to suppression of chronic allergic dermatitis, Immunity, vol.35, pp.562-71, 2011.

B. K. Wershil, Z. S. Wang, J. R. Gordon, and S. J. Galli, Recruitment of neutrophils during IgE-dependent cutaneous late phase reactions in the mouse is mast celldependent. Partial inhibition of the reaction with antiserum against tumor necrosis factor-alpha, J Clin Invest, vol.87, pp.446-53, 1991.

M. W. Greaves and R. A. Sabroe, Histamine: the quintessential mediator, J Dermatol, vol.23, pp.735-775, 1996.

E. M. Renkin and F. E. Curry, Endothelial permeability: pathways and modulations, Ann N Y Acad Sci, vol.401, pp.248-59, 1982.

S. J. Compton, J. A. Cairns, S. T. Holgate, and A. F. Walls, Human mast cell tryptase stimulates the release of an IL-8-dependent neutrophil chemotactic activity from human umbilical vein endothelial cells (HUVEC), Clin Exp Immunol, vol.121, pp.31-37, 2000.

T. Oka, J. Kalesnikoff, P. Starkl, M. Tsai, and S. J. Galli, Evidence questioning cromolyn's effectiveness and selectivity as a 'mast cell stabilizer' in mice, Lab Invest, vol.92, pp.1472-82, 2012.

H. Jones, L. Hargrove, L. Kennedy, F. Meng, A. Graf-eaton et al., Inhibition of mast cell-secreted histamine decreases biliary proliferation and fibrosis in primary sclerosing cholangitis Mdr2(-/-) mice, Hepatology, vol.64, pp.1202-1218, 2016.

A. Tellechea, E. C. Leal, A. Kafanas, M. E. Auster, S. Kuchibhotla et al., Mast cells regulate wound healing in diabetes, Diabetes, vol.65, pp.2006-2025, 2016.

P. Y. Gan, K. M. O'sullivan, J. D. Ooi, M. A. Alikhan, D. Odobasic et al., Mast cell stabilization ameliorates autoimmune antimyeloperoxidase glomerulonephritis, J Am Soc Nephrol, vol.27, pp.1321-1354, 2016.

Z. Q. Hei, X. L. Gan, P. J. Huang, J. Wei, N. Shen et al., Influence of ketotifen, cromolyn sodium, and compound 48/80 on the survival rates after intestinal ischemia reperfusion injury in rats, BMC Gastroenterol, vol.8, p.42, 2008.