S. Lee, T. Miyagi, and C. Biron, Keeping NK cells in highly regulated antiviral warfare, Trends in Immunology, vol.28, issue.6, pp.252-261, 2007.
DOI : 10.1016/j.it.2007.04.001

M. Smyth, Y. Hayakawa, K. Takeda, and H. Yagita, New aspects of natural-killer-cell surveillance and therapy of cancer, Nature Reviews Cancer, vol.419, issue.11, pp.850-6110, 1038.
DOI : 10.1038/nature01112

H. Ljunggren and K. Karre, In search of the ???missing self???: MHC molecules and NK cell recognition, Immunology Today, vol.11, issue.790, pp.237-281, 1990.
DOI : 10.1016/0167-5699(90)90097-S

V. Renard, A. Cambiaggi, F. Vely, M. Blery, L. Olcese et al., Transduction of cytotoxic signals in natural killer cells: a general model of fine tuning between activatory and inhibitory pathways in lymphocytes, Immunological Reviews, vol.5, issue.111, pp.205-226, 1997.
DOI : 10.1084/jem.180.2.545

C. Heidecke, J. Araujo, J. Kupiec-weglinski, M. Abbud-filho, D. Araneda et al., Lack of evidence for an active role for natural killer cells in acute rejection of organ allografts, Transplantation, vol.40, issue.4, pp.441-445, 1985.

P. Markus, M. Van-den-brink, X. Cai, J. Harnaha, L. Palomba et al., Effect of selective depletion of natural killer cells on allograft rejection, Transplant Proc, vol.231, issue.1, pp.178-187, 1991.

W. Kitchens, S. Uehara, C. Chase, R. Colvin, P. Russell et al., The Changing Role of Natural Killer Cells in Solid Organ Rejection and Tolerance, Transplantation, vol.81, issue.6, pp.811-818, 2006.
DOI : 10.1097/01.tp.0000202844.33794.0e

W. Van-der-touw and J. Bromberg, Natural Killer Cells and the Immune Response in Solid Organ Transplantation, American Journal of Transplantation, vol.202, issue.3 Suppl, pp.1354-1362, 2010.
DOI : 10.1111/j.1365-2249.2008.03601.x

R. Rajalingam, The impact of HLA class I-specific killer cell immunoglobulinlike receptors on antibody-dependent natural killer cell-mediated cytotoxicity and organ allograft rejection, Front Immunol, 2016.

U. Hadad, O. Martinez, and S. Krams, NK cells after transplantation: friend or foe, Immunologic Research, vol.302, issue.5646, pp.259-67, 2014.
DOI : 10.1126/science.1087061

J. Kummer, P. Wever, A. Kamp, I. Ten-berge, C. Hack et al., Expression of granzyme A and B proteins by cytotoxic lymphocytes involved in acute renal allograft rejection, Kidney International, vol.47, issue.1, pp.70-77, 1995.
DOI : 10.1038/ki.1995.8

E. Vivier, D. Raulet, A. Moretta, M. Caligiuri, L. Zitvogel et al., Innate or Adaptive Immunity? The Example of Natural Killer Cells, Science, vol.5, issue.5, pp.44-53, 2011.
DOI : 10.1038/nri1603

URL : https://hal.archives-ouvertes.fr/hal-00611585

E. Narni-mancinelli, B. Jaeger, C. Bernat, A. Fenis, S. Kung et al., Tuning of Natural Killer Cell Reactivity by NKp46 and Helios Calibrates T Cell Responses, Science, vol.2, issue.1, pp.344-352, 2012.
DOI : 10.1186/1479-0556-2-13

URL : https://hal.archives-ouvertes.fr/hal-00685893

T. Hirohashi, C. Chase, D. Pelle, P. Sebastian, D. Alessandrini et al., A Novel Pathway of Chronic Allograft Rejection Mediated by NK Cells and Alloantibody, American Journal of Transplantation, vol.27, issue.2, pp.313-334, 2012.
DOI : 10.1016/j.healun.2007.11.571

D. Zecher, Q. Li, M. Oberbarnscheidt, A. Demetris, W. Shlomchik et al., NK Cells Delay Allograft Rejection in Lymphopenic Hosts by Downregulating the Homeostatic Proliferation of CD8+ T Cells, The Journal of Immunology, vol.184, issue.12, pp.6649-57, 2010.
DOI : 10.4049/jimmunol.0903729

J. Beilke, N. Kuhl, L. Van-kaer, and R. Gill, NK cells promote islet allograft tolerance via a perforin-dependent mechanism, Nature Medicine, vol.100, issue.10, pp.1059-65, 2005.
DOI : 10.1073/pnas.1332805100

G. Yu, X. Xu, M. Vu, E. Kilpatrick, and X. Li, NK cells promote transplant tolerance by killing donor antigen-presenting cells, The Journal of Experimental Medicine, vol.203, issue.8, pp.1851-1859, 2006.
DOI : 10.4049/jimmunol.173.1.214

URL : http://jem.rupress.org/content/jem/203/8/1851.full.pdf

S. Laffont, C. Seillet, J. Ortaldo, J. Coudert, and J. Guery, Natural killer cells recruited into lymph nodes inhibit alloreactive T-cell activation through perforin-mediated killing of donor allogeneic dendritic cells, Blood, vol.112, issue.3, pp.661-71, 2008.
DOI : 10.1182/blood-2007-10-120089

URL : https://hal.archives-ouvertes.fr/inserm-00306011

R. Owen, H. Wood, A. Foord, P. Sturgeon, and L. Baldwin, EVIDENCE FOR ACTIVELY ACQUIRED TOLERANCE TO Rh ANTIGENS, Proceedings of the National Academy of Sciences, vol.40, issue.6, pp.420-424, 1954.
DOI : 10.1073/pnas.40.6.420

R. Billingham, L. Brent, and P. Medawar, ???Actively Acquired Tolerance??? of Foreign Cells, Nature, vol.12, issue.4379, pp.603-609, 1953.
DOI : 10.3181/00379727-61-15294P

M. Sykes and D. Sachs, Mixed allogeneic chimerism as an approach to transplantation tolerance, Immunology Today, vol.9, issue.1, pp.23-30, 1988.
DOI : 10.1016/0167-5699(88)91352-7

J. Scandling, S. Busque, S. Dejbakhsh-jones, C. Benike, M. Millan et al., Tolerance and Chimerism after Renal and Hematopoietic-Cell Transplantation, New England Journal of Medicine, vol.358, issue.4, pp.362-370, 2008.
DOI : 10.1056/NEJMoa074191

T. Kawai, A. Cosimi, T. Spitzer, N. Tolkoff-rubin, M. Suthanthiran et al., HLA-Mismatched Renal Transplantation without Maintenance Immunosuppression, New England Journal of Medicine, vol.358, issue.4, pp.353-61, 2008.
DOI : 10.1056/NEJMoa071074

URL : https://dash.harvard.edu/bitstream/handle/1/27528053/nejmoa071074.pdf?sequence=1

A. Massart, A. Pallier, J. Pascual, O. Viklicky, K. Budde et al., The DESCARTES-Nantes survey of kidney transplant recipients displaying clinical operational tolerance identifies 35 new tolerant patients and 34 almost tolerant patients, Nephrology Dialysis Transplantation, vol.31, issue.6, pp.311002-311015, 2016.
DOI : 10.1038/172603a0

G. Roussey-kesler, M. Giral, A. Moreau, J. Subra, C. Legendre et al., Clinical Operational Tolerance after Kidney Transplantation, American Journal of Transplantation, vol.74, issue.Suppl 1, pp.736-782, 2006.
DOI : 10.1097/00007890-200209270-00014

URL : http://onlinelibrary.wiley.com/doi/10.1111/j.1600-6143.2006.01280.x/pdf

C. Ballet, G. Roussey-kesler, J. Aubin, S. Brouard, M. Giral et al., Humoral and Cellular Responses to Influenza Vaccination in Human Recipients Naturally Tolerant to a Kidney Allograft, American Journal of Transplantation, vol.42, issue.11, pp.2796-801, 2006.
DOI : 10.1111/j.1365-2141.2004.05204.x

S. Brouard, A. Pallier, K. Renaudin, Y. Foucher, R. Danger et al., The Natural History of Clinical Operational Tolerance After Kidney Transplantation Through Twenty-Seven Cases, American Journal of Transplantation, vol.21, issue.12 Suppl, pp.3296-307, 2012.
DOI : 10.1681/ASN.2009121296

R. Calne, R. Sells, J. Pena, D. Davis, P. Millard et al., Induction of Immunological Tolerance by Porcine Liver Allografts, Nature, vol.4, issue.5205, pp.472-478, 1969.
DOI : 10.1084/jem.126.4.655

S. Qian, A. Demetris, N. Murase, A. Rao, J. Fung et al., Murine liver allograft transplantation: Tolerance and donor cell chimerism, Hepatology, vol.136, issue.4, pp.916-940, 1994.
DOI : 10.1016/S0171-2985(87)80003-7

N. Kamada and T. Shinomiya, Clonal deletion as the mechanism of abrogation of immunological memory following liver grafting in rats, Immunology, vol.55, issue.1, pp.85-90, 1985.

M. Martínez-llordella, I. Puig-pey, G. Orlando, M. Ramoni, G. Tisone et al., Multiparameter Immune Profiling of Operational Tolerance in Liver Transplantation, American Journal of Transplantation, vol.4, issue.2, pp.309-328, 2007.
DOI : 10.1016/0168-8278(95)80226-6

J. Lozano, A. Pallier, M. Martinez-llordella, R. Danger, M. López et al., Comparison of Transcriptional and Blood Cell-Phenotypic Markers Between Operationally Tolerant Liver and Kidney Recipients, American Journal of Transplantation, vol.3, issue.9, pp.1916-1942, 2011.
DOI : 10.1038/nri981

K. Newell, A. Asare, A. Kirk, T. Gisler, K. Bourcier et al., Identification of a B cell signature associated with renal transplant tolerance in humans, Journal of Clinical Investigation, vol.120, issue.6, pp.1836-1883, 2010.
DOI : 10.1172/JCI39933DS1

P. Sagoo, E. Perucha, B. Sawitzki, S. Tomiuk, D. Stephens et al., Development of a cross-platform biomarker signature to detect renal transplant tolerance in humans, Journal of Clinical Investigation, vol.120, issue.6, pp.1848-61, 2010.
DOI : 10.1172/JCI39922DS1

D. Baron, G. Ramstein, M. Chesneau, Y. Echasseriau, A. Pallier et al., A common gene signature across multiple studies relate biomarkers and functional regulation in tolerance to renal allograft, Kidney International, vol.87, issue.5, pp.984-95, 2015.
DOI : 10.1038/ki.2014.395

URL : https://hal.archives-ouvertes.fr/hal-01119557

A. Pallier, S. Hillion, R. Danger, M. Giral, M. Racapé et al., Patients with drug-free long-term graft function display increased numbers of peripheral B cells with a memory and inhibitory phenotype, Kidney International, vol.78, issue.5, pp.503-516, 2010.
DOI : 10.1038/ki.2010.162

S. Miller, D. Dykes, and H. Polesky, A simple salting out procedure for extracting DNA from human nucleated cells, Nucleic Acids Research, vol.16, issue.3, 1988.
DOI : 10.1093/nar/16.3.1215

J. Sun, L. Gaidulis, M. Miller, R. Goto, R. Rodriguez et al., Development of a multiplex PCR-SSP method for killer-cell immunoglobulinlike receptor genotyping, Tissue Antigens, vol.64, issue.4, 2004.

G. David, M. Morvan, K. Gagne, N. Kerdudou, C. Willem et al., Discrimination between the main activating and inhibitory killer cell immunoglobulin-like receptor positive natural killer cell subsets using newly characterized monoclonal antibodies, Immunology, vol.26, issue.2, pp.172-84, 2009.
DOI : 10.1111/j.1365-2249.2007.03424.x

M. Lee, S. Villanueva, J. Sumegi, J. Zhang, K. Kogawa et al., Characterisation of diverse PRF1 mutations leading to decreased natural killer cell activity in North American families with haemophagocytic lymphohistiocytosis, J Med Genet, vol.41, issue.2, pp.137-44011528, 2003.

I. Voskoboinik and J. Trapani, Perforinopathy: A Spectrum of Human Immune Disease Caused by Defective Perforin Delivery or Function, Frontiers in Immunology, vol.4, 2013.
DOI : 10.3389/fimmu.2013.00441

W. Oboshi, T. Watanabe, Y. Matsuyama, A. Kobara, N. Yukimasa et al., The influence of NK cell-mediated ADCC: Structure and expression of the CD16 molecule differ among Fc??RIIIa-V158F genotypes in healthy Japanese subjects, Human Immunology, vol.77, issue.2, pp.165-71, 2016.
DOI : 10.1016/j.humimm.2015.11.001

S. Uehara, C. Chase, R. Colvin, P. Russell, and J. Madsen, Further evidence that NK cells may contribute to the development of cardiac allograft vasculopathy, Transplantation Proceedings, vol.37, issue.1, pp.70-71, 2005.
DOI : 10.1016/j.transproceed.2005.01.052

S. Uehara, C. Chase, W. Kitchens, H. Rose, R. Colvin et al., NK Cells Can Trigger Allograft Vasculopathy: The Role of Hybrid Resistance in Solid Organ Allografts, The Journal of Immunology, vol.175, issue.5, pp.3424-3454, 2005.
DOI : 10.4049/jimmunol.175.5.3424

L. Hidalgo, J. Sellares, B. Sis, M. Mengel, J. Chang et al., Interpreting NK Cell Transcripts Versus T Cell Transcripts in Renal Transplant Biopsies, American Journal of Transplantation, vol.7, issue.10, pp.1180-91, 2012.
DOI : 10.1038/ni1402

L. Hidalgo, B. Sis, J. Sellares, P. Campbell, M. Mengel et al., NK Cell Transcripts and NK Cells in Kidney Biopsies from Patients with Donor-Specific Antibodies: Evidence for NK Cell Involvement in Antibody-Mediated Rejection, American Journal of Transplantation, vol.6, issue.8, pp.1812-1834, 2010.
DOI : 10.4049/jimmunol.166.10.6404

T. Legris, C. Picard, D. Todorova, L. Lyonnet, C. Laporte et al., Antibody-Dependent NK Cell Activation Is Associated with Late Kidney Allograft Dysfunction and the Complement-Independent Alloreactive Potential of Donor-Specific Antibodies, Frontiers in Immunology, vol.24, issue.11, 2016.
DOI : 10.1093/intimm/dxs048

URL : https://hal.archives-ouvertes.fr/hal-01462978

W. Murphy, V. Kumar, and M. Bennett, Acute rejection of murine bone marrow allografts by natural killer cells and T cells. Differences in kinetics and target antigens recognized, Journal of Experimental Medicine, vol.166, issue.5, pp.1499-509, 1987.
DOI : 10.1084/jem.166.5.1499

C. Neudoerfl, B. Mueller, C. Blume, K. Daemen, M. Stevanovic-meyer et al., The Peripheral NK Cell Repertoire after Kidney Transplantation is Modulated by Different Immunosuppressive Drugs, Frontiers in Immunology, vol.4, 2013.
DOI : 10.3389/fimmu.2013.00046

A. Meehan, N. Mifsud, T. Nguyen, B. Levvey, G. Snell et al., Impact of Commonly Used Transplant Immunosuppressive Drugs on Human NK Cell Function Is Dependent upon Stimulation Condition, PLoS ONE, vol.132, issue.3, 2013.
DOI : 10.1371/journal.pone.0060144.t001

U. Hoffmann, C. Neudorfl, K. Daemen, J. Keil, M. Stevanovic-meyer et al., NK Cells of Kidney Transplant Recipients Display an Activated Phenotype that Is Influenced by Immunosuppression and Pathological Staging, PLOS ONE, vol.13, issue.7, 2015.
DOI : 10.1371/journal.pone.0132484.s008

N. Bjorkstrom, P. Riese, F. Heuts, S. Andersson, C. Fauriat et al., Expression patterns of NKG2A, KIR, and CD57 define a process of CD56dim NK-cell differentiation uncoupled from NK-cell education, Blood, vol.116, issue.19, pp.3853-64, 2010.
DOI : 10.1182/blood-2010-04-281675

A. Freud, A. Yokohama, B. Becknell, M. Lee, H. Mao et al., Evidence for discrete stages of human natural killer cell differentiation in vivo, The Journal of Experimental Medicine, vol.161, issue.4, pp.1033-1076, 2006.
DOI : 10.1182/blood.V97.10.3146

V. Beziat, B. Descours, C. Parizot, P. Debre, and V. Vieillard, NK Cell Terminal Differentiation: Correlated Stepwise Decrease of NKG2A and Acquisition of KIRs, PLoS ONE, vol.4, issue.8, 2010.
DOI : 10.1371/journal.pone.0011966.s003

D. Chiesa, M. Romeo, E. Falco, M. Balsamo, M. Augugliaro et al., Evidence that the KIR2DS5 gene codes for a surface receptor triggering natural killer cell function, European Journal of Immunology, vol.182, issue.8, pp.2284-2293, 2008.
DOI : 10.4049/jimmunol.169.9.5118

A. Pessino, S. Sivori, C. Bottino, A. Malaspina, L. Morelli et al., Molecular Cloning of NKp46: A Novel Member of the Immunoglobulin Superfamily Involved in Triggering of Natural Cytotoxicity, The Journal of Experimental Medicine, vol.159, issue.5, pp.953-60, 1998.
DOI : 10.1016/S0960-9822(06)00263-6

S. Sivori, D. Pende, C. Bottino, E. Marcenaro, A. Pessino et al., NKp46 is the major triggering receptor involved in the natural cytotoxicity of fresh or cultured human NK cells. Correlation between surface density of NKp46 and natural cytotoxicity against autologous, allogeneic or xenogeneic target cells, 05<1656::AID-IMMU1656>3.0.CO, pp.1656-6610, 1999.
DOI : 10.1006/bbrc.1994.2482

S. Sivori, M. Vitale, L. Morelli, L. Sanseverino, R. Augugliaro et al., p46, a Novel Natural Killer Cell???specific Surface Molecule That Mediates Cell Activation, The Journal of Experimental Medicine, vol.7, issue.7, pp.1129-1165, 1997.
DOI : 10.1016/0167-5699(87)90173-3

D. Pende, S. Parolini, A. Pessino, S. Sivori, R. Augugliaro et al., Identification and Molecular Characterization of NKp30, a Novel Triggering Receptor Involved in Natural Cytotoxicity Mediated by Human Natural Killer Cells, Journal of Experimental Medicine, vol.190, issue.10, pp.1505-1521, 1999.
DOI : 10.1084/jem.190.10.1505

T. Lakshmikanth, S. Burke, T. Ali, S. Kimpfler, F. Ursini et al., NCRs and DNAM-1 mediate NK cell recognition and lysis of human and mouse melanoma cell lines in vitro and in vivo, Journal of Clinical Investigation, vol.119, issue.5, pp.1251-6310, 1172.
DOI : 10.1172/JCI36022DS1

A. Shibuya, D. Campbell, C. Hannum, H. Yssel, K. Franz-bacon et al., DNAM-1, A Novel Adhesion Molecule Involved in the Cytolytic Function of T Lymphocytes, Immunity, vol.4, issue.6, pp.573-81, 1996.
DOI : 10.1016/S1074-7613(00)70060-4

Z. Zhang, N. Wu, Y. Lu, D. Davidson, M. Colonna et al., DNAM-1 controls NK cell activation via an ITT-like motif, The Journal of Experimental Medicine, vol.161, issue.12, pp.2165-82, 2015.
DOI : 10.1128/MCB.21.18.6102-6112.2001

URL : http://jem.rupress.org/content/jem/212/12/2165.full.pdf

B. Sanchez-correa, C. Campos, A. Pera, J. Bergua, M. Arcos et al., Natural killer cell immunosenescence in acute myeloid leukaemia patients: new targets for immunotherapeutic strategies?, Cancer Immunology, Immunotherapy, vol.173, issue.4, pp.453-63, 2016.
DOI : 10.1111/cei.12095

S. Kulkarni, M. Martin, and M. Carrington, The Yin and Yang of HLA and KIR in human disease, Seminars in Immunology, vol.20, issue.6, pp.343-52, 2008.
DOI : 10.1016/j.smim.2008.06.003

M. Cavalcanti, A. Jewett, and B. Bonavida, Irreversible cancer cell-induced functional anergy and apoptosis in resting and activated NK cells., International Journal of Oncology, vol.14, issue.2, pp.361-367, 1999.
DOI : 10.3892/ijo.14.2.361

Q. Zhou, A. Gil-krzewska, G. Peruzzi, and F. Borrego, Matrix metalloproteinases inhibition promotes the polyfunctionality of human natural killer cells in therapeutic antibody-based anti-tumour immunotherapy, Clinical & Experimental Immunology, vol.7, issue.1, pp.131-140, 2013.
DOI : 10.1371/journal.pone.0038580

S. Jost, J. Reardon, E. Peterson, D. Poole, R. Bosch et al., Expansion of 2B4+ natural killer (NK) cells and decrease in NKp46+ NK cells in response to influenza, Immunology, vol.15, issue.4, pp.516-542, 2011.
DOI : 10.1128/CVI.00357-07

R. Arriga, S. Caratelli, A. Coppola, G. Spagnoli, A. Venditti et al., Enhancement of anti-leukemia activity of NK cells <i>in vitro</i> and <i>in vivo</i> by inhibition of leukemia cell-induced NK cell damage, Oncotarget, vol.7, issue.2, pp.2070-2079, 2016.
DOI : 10.18632/oncotarget.6529

R. Costello, S. Sivori, E. Marcenaro, M. Lafage-pochitaloff, M. Mozziconacci et al., Defective expression and function of natural killer cell-triggering receptors in patients with acute myeloid leukemia, Blood, vol.99, issue.10, pp.3661-3668, 2002.
DOI : 10.1182/blood.V99.10.3661

T. Garcia-iglesias, D. Toro-arreola, A. Albarran-somoza, B. , D. Toro-arreola et al., Low NKp30, NKp46 and NKG2D expression and reduced cytotoxic activity on NK cells in cervical cancer and precursor lesions, BMC Cancer, vol.3, issue.5, pp.186-196, 2009.
DOI : 10.1371/journal.pone.0002150

M. Parsons, C. Tang, S. Jegaskanda, R. Center, A. Brooks et al., Anti-HIV Antibody-Dependent Activation of NK Cells Impairs NKp46 Expression, The Journal of Immunology, vol.192, issue.1, pp.308-323, 2014.
DOI : 10.4049/jimmunol.1301247

J. Nattermann, G. Feldmann, G. Ahlenstiel, B. Langhans, T. Sauerbruch et al., Surface expression and cytolytic function of natural killer cell receptors is altered in chronic hepatitis C, Gut, vol.55, issue.6, pp.869-77, 2005.
DOI : 10.1136/gut.2005.076463

D. Chiesa, M. Pesce, S. Muccio, L. Carlomagno, S. Sivori et al., Features of memory-like and PD-1(+) human NK cell subsets, Front Immunol, 2016.

A. Beldi-ferchiou, M. Lambert, S. Dogniaux, F. Vely, E. Vivier et al., PD-1 mediates functional exhaustion of activated NK cells in patients with Kaposi sarcoma, Oncotarget, vol.7, issue.45, pp.72961-77, 2016.
DOI : 10.18632/oncotarget.12150

URL : https://hal.archives-ouvertes.fr/hal-01438166

S. Wiesmayr, S. Webber, C. Macedo, I. Popescu, L. Smith et al., Decreased NKp46 and NKG2D and elevated PD-1 are associated with altered NK-cell function in pediatric transplant patients with PTLD, European Journal of Immunology, vol.179, issue.2, pp.541-50, 2012.
DOI : 10.4049/jimmunol.179.5.3362

S. Durr and V. Kindler, Implication of indolamine 2,3 dioxygenase in the tolerance toward fetuses, tumors, and allografts, Journal of Leukocyte Biology, vol.209, issue.5, pp.681-688, 2013.
DOI : 10.1084/jem.20120408

G. Brandacher, R. Margreiter, and D. Fuchs, Clinical relevance of indoleamine 2,3-dioxygenase for alloimmunity and transplantation, Current Opinion in Organ Transplantation, vol.13, issue.1, pp.10-15, 2008.
DOI : 10.1097/MOT.0b013e3282f3df26

D. Chiesa, M. Carlomagno, S. Frumento, G. Balsamo, M. Cantoni et al., The tryptophan catabolite L-kynurenine inhibits the surface expression of NKp46- and NKG2D-activating receptors and regulates NK-cell function, Blood, vol.108, issue.13, pp.4118-4143, 2006.
DOI : 10.1182/blood-2006-03-006700

A. Kroemer, X. Xiao, N. Degauque, K. Edtinger, H. Wei et al., The Innate NK Cells, Allograft Rejection, and a Key Role for IL-15, The Journal of Immunology, vol.180, issue.12, pp.7818-7844, 2008.
DOI : 10.4049/jimmunol.180.12.7818

G. Deniz, G. Erten, U. Kücüksezer, D. Kocacik, C. Karagiannidis et al., Regulatory NK Cells Suppress Antigen-Specific T Cell Responses, The Journal of Immunology, vol.180, issue.2, pp.850-857, 2008.
DOI : 10.4049/jimmunol.180.2.850

URL : http://www.jimmunol.org/content/jimmunol/180/2/850.full.pdf

A. Maroof, L. Beattie, S. Zubairi, M. Svensson, S. Stager et al., Posttranscriptional Regulation of Il10 Gene Expression Allows Natural Killer Cells to Express Immunoregulatory Function, Immunity, vol.29, issue.2, pp.295-305, 2008.
DOI : 10.1016/j.immuni.2008.06.012

K. Garrod, F. Liu, L. Forrest, I. Parker, S. Kang et al., NK Cell Patrolling and Elimination of Donor-Derived Dendritic Cells Favor Indirect Alloreactivity, The Journal of Immunology, vol.184, issue.5, pp.2329-2365, 2010.
DOI : 10.4049/jimmunol.0902748

E. Serti, J. Werner, M. Chattergoon, A. Cox, V. Lohmann et al., Monocytes Activate Natural Killer Cells via Inflammasome-Induced Interleukin 18 in Response to Hepatitis C Virus Replication, Gastroenterology, vol.147, issue.1, 2014.
DOI : 10.1053/j.gastro.2014.03.046

L. Rovis, T. , K. Brlic, P. Kaynan, N. et al., Inflammatory monocytes and NK cells play a crucial role in DNAM-1???dependent control of cytomegalovirus infection, The Journal of Experimental Medicine, vol.73, issue.9, pp.1835-50, 2016.
DOI : 10.1093/intimm/dxm119

I. Blanca, E. Bere, H. Young, and J. Ortaldo, Human B Cell Activation by Autologous NK Cells Is Regulated by CD40-CD40 Ligand Interaction: Role of Memory B Cells and CD5+ B Cells, The Journal of Immunology, vol.167, issue.11, pp.6132-6141, 2001.
DOI : 10.4049/jimmunol.167.11.6132

F. Ghiringhelli, C. Ménard, M. Terme, C. Flament, J. Taieb et al., regulatory T cells inhibit natural killer cell functions in a transforming growth factor????????dependent manner, The Journal of Experimental Medicine, vol.9, issue.8, pp.1075-85, 2005.
DOI : 10.4049/jimmunol.172.10.5957

URL : https://hal.archives-ouvertes.fr/pasteur-00020140

J. Zimmer, E. Andres, and F. Hentges, NK cells and Treg cells: A fascinating dance cheek to cheek, European Journal of Immunology, vol.6, issue.646, pp.2942-2947, 2008.
DOI : 10.4049/jimmunol.170.7.3572

E. Vivier, E. Tomasello, M. Baratin, T. Walzer, and S. Ugolini, Functions of natural killer cells, Nature Immunology, vol.178, issue.5, pp.503-513, 2008.
DOI : 10.1073/pnas.050588297

URL : https://hal.archives-ouvertes.fr/hal-00294184

R. Liu, L. Van-kaer, L. Cava, A. Price, M. Campagnolo et al., Autoreactive T Cells Mediate NK Cell Degeneration in Autoimmune Disease, The Journal of Immunology, vol.176, issue.9, pp.5247-54, 2006.
DOI : 10.4049/jimmunol.176.9.5247

P. Vacca, C. Cantoni, M. Vitale, C. Prato, F. Canegallo et al., Crosstalk between decidual NK and CD14+ myelomonocytic cells results in induction of Tregs and immunosuppression, Proceedings of the National Academy of Sciences, vol.273, issue.24, pp.11918-11941, 2010.
DOI : 10.1001/jama.1995.03520480053039

I. Pedroza-pacheco, A. Madrigal, and A. Saudemont, Interaction between natural killer cells and regulatory T cells: perspectives for immunotherapy, Cellular & Molecular Immunology, vol.179, issue.3, pp.222-231, 2013.
DOI : 10.1126/scitranslmed.3001302

F. Regateiro, D. Howie, S. Cobbold, and H. Waldmann, TGF-?? in transplantation tolerance, Current Opinion in Immunology, vol.23, issue.5, pp.660-669, 2011.
DOI : 10.1016/j.coi.2011.07.003

F. Braza, E. Dugast, I. Panov, C. Paul, K. Vogt et al., Central Role of CD45RA- Foxp3hi Memory Regulatory T Cells in Clinical Kidney Transplantation Tolerance, Journal of the American Society of Nephrology, vol.26, issue.8, pp.1795-805, 2015.
DOI : 10.1681/ASN.2014050480

M. Chesneau, L. Michel, E. Dugast, A. Chenouard, D. Baron et al., Tolerant Kidney Transplant Patients Produce B Cells with Regulatory Properties, Journal of the American Society of Nephrology, vol.26, issue.10, pp.2588-98, 2015.
DOI : 10.1681/ASN.2014040404