Z. X. Zhang, L. Yang, K. J. Young, B. Dutemple, and L. Zhang, Identification of a previously unknown antigen-specific regulatory T cell and its mechanism of suppression, Nat Med, 2000.

K. Fischer, S. Voelkl, and J. Heymann, Isolation and characterization of human antigen-specific TCR alpha beta+ CD4(-)CD8-double-negative regulatory T cells, Blood, vol.105, issue.7, pp.2828-2835, 2005.

J. Morel, M. Berthelot, and L. Pratique, Club rhumatismes et inflammations, fiches immunoloqie, 20072.

C. Lambert, Lymphocytes T non conventionnels circulants détectés par cytométrie en flux. Immunologie, 2005.

D. A. Ferrick, D. P. King, and K. A. Jackson, Intraepithelial ?? T lymphocytes: sentinel cells at mucosal barriers, Springer Seminars in Immunopathology, vol.22, pp.283-296, 2000.

J. Chen, H. Niu, W. He, and D. Ba, Antitumor activity of expanded human tumor-infiltrating gammadelta T lymphocytes, Int Arch Allergy Immunol, vol.125, issue.3, pp.256-263, 2001.

Z. W. Chen and N. L. Letvin, Adaptive immune response of Vgamma2Vdelta2 T cells: a new paradigm, Trends Immunol, vol.24, issue.4, pp.213-219, 2003.

D. Rosa, S. C. Mitra, D. K. Watanabe, N. Herzenberg, L. A. Herzenberg et al., Vdelta1 and Vdelta2 gammadelta T cells express distinct surface markers and might be developmentally distinct lineages, J Leukoc Biol, vol.70, issue.4, pp.518-526, 2001.

, Vdelta1 and Vdelta2 gammadelta T cells express distinct surface markers and might be developmentally distinct lineages. De Rosa, J Leukoc Biol, 2001.

J. M. Jameson, G. Cauvi, D. A. Witherden, and W. L. Havran, A keratinocyte-responsive gamma delta TCR is necessary for dendritic epidermal T cell activation by damaged keratinocytes and maintenance in the epidermis, J Immunol, vol.172, issue.6, pp.3573-3579, 2004.

, Collège des Enseignants d'Immunologie. Immunologie fondamentale et immunopathologie, pp.2-3

. Médecine, 2 ème édition, 2018.

B. Choufi and K. Lassoued, The thymus in allogeneic haematopoietic stem cell transplantation, vol.18, 2012.

L. Deist, F. Rieux-laucat, F. Hivroz, and C. , Anomalies d'expression du complexe récepteur T de l'antigène/CD3 et déficits immunitaires, Med Sci, vol.23, issue.2, pp.161-166, 2007.

J. L. Aymeric, G. Lefranc, . Immunologie-humaine-de-boeck, and . Supérieur, , 2011.

A. K. Sewell, Why must T cells be cross-reactive?, Nat Rev Immunol, vol.12, issue.9, pp.669-677, 2012.

D. Brandt and C. M. Hedrich, TCR??+CD3+CD4-CD8-(double negative) T cells in autoimmunity, Autoimmun Rev, vol.17, issue.4, pp.422-430, 2018.

M. E. Call and K. W. Wucherpfennig, Molecular mechanisms for the assembly of the T cell receptor-CD3 complex, Mol Immunol, vol.40, issue.18, pp.1295-1305, 2004.

G. Werlen and E. Palmer, The T-cell receptor signalosome: a dynamic structure with expanding complexity, 18 CD3 complex and immunodeficiencies Le Deist Med Sci, vol.14, pp.299-305, 2002.

L. Deist, F. Rieux-laucat, F. Hivroz, and C. , Anomalies d'expression du complexe récepteur T de l'antigène/CD3 et déficits immunitaires, Med Sci, vol.23, issue.2, pp.161-166, 2007.

M. N. Martina, S. Noel, A. Saxena, H. Rabb, and A. R. Hamad, Double negative (DN) ?? T cells: misperception and overdue recognition, Immunol Cell Biol, vol.93, issue.3, pp.305-310, 2015.

M. L. Joachims, J. L. Chain, S. W. Hooker, C. J. Knott-craig, and L. F. Thompson, Human alpha beta and gamma delta thymocyte development: TCR gene rearrangements, intracellular TCR beta expression, and gamma delta developmental potential--differences between men and mice, J Immunol, vol.176, issue.3, pp.1543-1552, 2006.

J. C. Chapman, F. M. Chapman, and S. D. Michael, The production of alpha/beta and gamma/delta double negative (DN) Tcells and their role in the maintenance of pregnancy, Reprod Biol Endocrinol, vol.13, p.73, 2015.

. Dikwa, K. Pike-overzet, F. Weerkamp, D. De-ridder, E. De-haas et al., New insights on human T cell development by quantitative T cell receptor gene rearrangement studies and gene expression profiling, J Exp Med, vol.201, pp.1715-1738, 2005.

H. Spits, Development of alphabeta T cells in the human thymus, Nat Rev Immunol, vol.2, pp.760-72, 2002.

T. Kreslavsky, H. J. Kim, and S. B. Koralov, Negative selection, not receptor editing, is a physiological response of autoreactive thymocytes, J Exp Med, vol.210, issue.10, pp.1911-1918, 2013.

. Joachimsml, J. L. Chain, S. W. Hooker, C. J. Knott-craig, and L. F. Thompson, Human ?? and ?? thymocyte development: TCR gene rearrangements, intracellular TCR? expression and ?? developmental potential -differences between men and mice, J Immunol Baltim Md, vol.176, pp.1543-52, 1950.

S. M. Hayes, L. Li, and P. E. Love, TCR signal strength influences alphabeta/gammadelta lineage fate, Immunity, vol.22, pp.583-93, 2005.

S. Mayans, D. Stepniak, S. Palida, A. Larange, J. Dreux et al., ??T cell receptors expressed by CD4?CD8??? intraepithelial T cells drive their fate into a unique lineagewith unusualMHC reactivities, Immunity, vol.41, pp.207-225, 2014.

R. Wang, Y. Wang-zhu, and H. Grey, Interactions between double positive thymocytes and high affinity ligands presented by cortical epithelial cells generate double negative thymocytes with T cell regulatory activity, Proc Natl Acad Sci, vol.99, issue.4, pp.2181-2186, 2002.

T. A. Baldwin, M. M. Sandau, S. C. Jameson, and K. A. Hogquist, The timing of TCR alpha expression critically influences T cell development and selection, J Exp Med, vol.202, issue.1, pp.111-121, 2005.

J. J. Priatel, O. Utting, and H. S. Teh, TCR/self-antigen interactions drive double-negative T cell peripheral expansion and differentiation into suppressor cells, J Immunol Baltim Md, vol.167, pp.6188-94, 1950.

. Pe-love, . Shores, and . Johnson, T cell development in mice that lack the zeta chain of the T cell antigen receptor complex, Advancement of Science, vol.261, issue.5123, pp.918-921, 1993.

J. C. Crispín, M. Oukka, and G. Bayliss, Expanded double negative T cells in patients with systemic lupus erythematosus produce IL-17 and infiltrate the kidneys, J Immunol, vol.181, issue.12, pp.8761-8766, 2008.

J. A. Tarbox, M. P. Keppel, and N. Topcagic, Elevated double negative T cells in pediatric autoimmunity, J Clin Immunol, vol.34, issue.5, pp.594-599, 2014.

C. M. Hedrich, J. C. Crispin, and G. C. Tsokos, Epigenetic regulation of cytokine expression in systemic lupus erythematosus with special focus on T cells, Autoimmunity, vol.47, issue.4, pp.234-241, 2014.

L. T. Michelle, S. A. Nguyen, J. E. Jones, and . Prier, Transcriptional enhancers in the regulation of T cell differentiation, Front. Immunol, 2015.

L. J. Kieffer, L. Yan, J. H. Hanke, and P. B. Kavathas, Appropriate developmental expression of human CD8 beta in transgenic mice, J Immunol, vol.159, issue.10, pp.4907-4912, 1997.

X. L. Zhang, CD8 expression up to the double-positive CD3(low/intermediate) stage of thymic differentiation is sufficient for development of peripheral functional cytotoxic T lymphocytes, J Exp Med, 2001.

F. Lambolez, M. Kronenberg, and H. Cheroutre, Thymic differentiation of TCR alpha beta(+) CD8 alpha alpha(+) IELs, Immunol Rev, vol.215, pp.178-188, 2007.

I. Bilic, C. Koesters, and B. Unger, Negative regulation of CD8 expression via Cd8 enhancer-mediated recruitment of the zinc finger protein MAZR, Nat Immunol, vol.7, issue.4, pp.392-400, 2006.

W. Ellmeier, L. Haust, and R. Tschismarov, Transcriptional control of CD4 and CD8 coreceptor expression during T cell development, Cell Mol Life Sci, vol.70, issue.23, pp.4537-4553, 2013.

L. A. Pobezinsky, G. S. Angelov, and X. Tai, Clonal deletion and the fate of autoreactive thymocytes that survive negative selection, Nat Immunol, vol.13, issue.6, pp.569-578, 2012.

S. Mayans, D. Stepniak, and S. Palida, ??T cell receptors expressed by CD4(-)CD8??(-) intraepithelial T cells drive their fate into a unique lineage with unusual MHC reactivities, Immunity, vol.41, issue.2, pp.207-218, 2014.

D. Brandt and C. M. Hedrich, TCR??+CD3+CD4-CD8-(double negative) T cells in autoimmunity, Autoimmun Rev, vol.17, issue.4, pp.422-430, 2018.

M. S. Ford, Z. Zhang, W. Chen, and L. Zhang, Double-negative T regulatory cells can develop outside the thymus and do not mature from CD8+ T cell precursors, J Immunol Baltim Md, 2006.

D. Zhang, W. Yang, N. Degauque, Y. Tian, A. Mikita et al., New differentiation pathway for double-negative regulatory T cells that regulates the magnitude of immune responses, Blood, vol.109, issue.9, pp.4071-4079, 2007.

Z. X. Zhang, L. Yang, K. J. Young, B. Dutemple, and L. Zhang, Identification of a previously unknown antigen-specific regulatory T cell and its mechanism of suppression, Nat Med, vol.6, issue.7, pp.782-789, 2000.

M. S. Ford, K. J. Young, Z. Zhang, P. S. Ohashi, and L. Zhang, The immune regulatory function of lymphoproliferative double negative T cells in vitro and in vivo, J Exp Med, vol.196, issue.2, pp.261-267, 2002.

Y. Ma, K. M. He, B. Garcia, M. W. Jevnikar, A. Zhang et al., Adoptive transfer of double negative T regulatory cells induces Bcell death in vivo and alters rejection pattern of rat-to-mouse heart transplantation, Xenotransplantation, vol.15, issue.1, pp.56-63, 2008.

K. M. He, Y. Ma, and S. Wang, Donor double-negative Treg promote allogeneic mixed chimerism and tolerance

, He KM European journal of immunology, European Journal of Immunology, vol.37, p.12, 2007.

J. F. Gao, M. S. Mcintyre, and C. Stephen, Regulation of antigen-expressing dendritic cells by double negative regulatory T cells

J. F. Gao, European journal of immunology, vol.41, issue.9, 2011.

I. V. Grishkan, A. Ntranos, P. A. Calabresi, and A. R. Gocke, Helper T cells down-regulate CD4 expression upon chronic stimulation giving rise to double-negative T cells, Cell Immunol, vol.284, issue.1-2, pp.68-74, 2013.

Z. X. Zhang, Identification of a previously unknown antigen-specific regulatory T cell and its mechanism of suppression. Nature medicine, 2000.

D. Zhang, W. Yang, N. Degauque, Y. Tian, A. Mikita et al., New differentiation pathway for double-negative regulatory T cells that regulates the magnitude of immune responses, Blood, vol.109, issue.9, pp.4071-4079, 2007.

K. Fischer, S. Voelkl, and J. Heymann, Isolation and characterization of human antigen-specific TCR alpha beta+ CD4(-)CD8-double-negative regulatory T cells, Blood, vol.105, issue.7, pp.2828-2835, 2005.

A. J. Ligocki and J. Y. Niederkorn, Advances on Non-CD4 + Foxp3+ T Regulatory Cells: CD8+, Type 1, and Double Negative T Regulatory Cells in Organ Transplantation, Transplantation, vol.99, issue.8, pp.1553-1559, 2015.

S. Strober, S. Dejbachsh-jones, V. Vlasselaer, P. Duwe, G. Salimi et al., Cloned natural suppressor cell lines express the CD3+CD4-CD8-surface phenotype and the alpha, beta heterodimer of the T cell antigen receptor, J Immunol, vol.143, issue.4, pp.1118-1122, 1989.

C. M. Hedrich, CAMP-responsive element modulator ? (CREM?) trans-represses the transmembrane glycoprotein CD8 and contributes to the generation of CD3+CD4?CD8? T cells in health and disease

, J Biol Chem, 2013.

C. M. Hedrich, T. Rauen, and J. C. Crispin, cAMP-responsive element modulator ? (CREM?) trans-represses the transmembrane glycoprotein CD8 and contributes to the generation of CD3+CD4-CD8-T cells in health and disease, J Biol Chem, vol.288, issue.44, pp.31880-31887, 2013.

H. Hassan, S. Sakaguchi, and M. Tenno, Cd8 enhancer E8I and Runx factors regulate CD8? expression in activated CD8+ T cells, Proc Natl Acad Sci, vol.108, issue.45, pp.18330-18335, 2011.

Z. X. Zhang, K. Young, and L. Zhang, CD3+CD4-CD8-alphabeta-TCR+ T cell as immune regulatory cell, J Mol Med (Berl)

Z. X. Zhang, L. Yang, K. J. Young, B. Dutemple, and L. Zhang, Identification of a previously unknown antigen-specific regulatory T cell and its mechanism of suppression, Nat Med, vol.6, issue.7, pp.782-789, 2000.

Z. X. Zhang, K. Young, and L. Zhang, CD3+CD4-CD8-alphabeta-TCR+ T cell as immune regulatory cell, J Mol Med (Berl)

S. C. Juvet and L. Zhang, Double negative regulatory T cells in transplantation and autoimmunity: recent progress and future directions, J Mol Cell Biol, vol.4, issue.1, pp.48-58, 2012.

K. L. Harland, E. B. Day, S. H. Apte, B. E. Russ, P. C. Doherty et al., Epigenetic plasticity of Cd8a locus during CD8 + T-cell development and effector differentiation and reprogramming, Nature Communications, 2014.

A. Onodera, M. Yamashita, and Y. Endo, STAT6-mediated displacement of polycomb by trithorax complex establishes longterm maintenance of GATA3 expression in T helper type 2 cells, J Exp Med, vol.207, issue.11, pp.2493-2506, 2010.

C. M. Hedrich, J. C. Crispín, and T. Rauen, cAMP responsive element modulator (CREM) ? mediates chromatin remodeling of CD8 during the generation of CD3+ CD4-CD8-T cells, J Biol Chem, vol.289, issue.4, pp.2361-2370, 2014.

H. , Cd8 enhancer E8I and Runx factors regulate CD8? expression in activated CD8+ T cells, Proc Natl Acad Sci, 2011.

D. Balomenos, R. Rumold, and A. N. Theofilopoulos, The proliferative in vivo activities of lpr double-negative T cells and the primary role of p59fyn in their activation and expansion, J Immunol, vol.159, issue.5, pp.2265-2273, 1997.

. Sc and L. Zhang, Double negative regulatory T cells in transplantation and autoimmunity: recent progress and future directions, J Mol Cell Biol, vol.4, issue.1, pp.48-58, 2012.

Z. X. Zhang, Identification of a previously unknown antigen-specific regulatory T cell and its mechanism of suppression, Nat Med, 2000.

S. C. Juvet and L. Zhang, Double negative regulatory T cells in transplantation and autoimmunity: recent progress and future directions, J Mol Cell Biol, vol.4, issue.1, pp.48-58, 2012.

F. Mcintyre, M. S. Young, K. J. Gao, J. Joe, B. Zhang et al., Cutting edge: in vivo trogocytosis as a mechanism of double negative regulatory T cell-mediated antigen-specific suppression, J Immunol, vol.181, issue.4, pp.2271-2275, 2008.

F. Mcintyre, M. S. Young, K. J. Gao, J. Joe, B. Zhang et al., Cutting edge: in vivo trogocytosis as a mechanism of double negative regulatory T cell-mediated antigen-specific suppression, J Immunol, vol.181, issue.4, pp.2271-2275, 2008.

C. W. Thomson, B. P. Lee, and L. Zhang, Double-negative regulatory T cells: non-conventional regulators, Immunol Res, vol.35, issue.1-2, pp.163-178, 2006.

D. Zhang, W. Yang, N. Degauque, Y. Tian, A. Mikita et al., New differentiation pathway for double-negative regulatory T cells that regulates the magnitude of immune responses, Blood, vol.109, issue.9, pp.4071-4079, 2007.

, Adoptive transfer of double negative T regulatory cells induces B-cell death in vivo and alters rejection pattern of rat-to-mouse heart transplantation, Xenotransplantation, 2008.

K. J. Young, The nature and mechanisms of DN regulatory T-cell mediated suppression, Human immunology, 2002.

Z. X. Zhang, D. Lian, and X. Huang, Adoptive transfer of DNT cells induces long-term cardiac allograft survival and augments recipient CD4(+)Foxp3(+) Treg cell accumulation, Transpl Immunol, vol.24, issue.2, pp.119-126, 2011.

J. F. Gao, M. S. Mcintyre, and S. C. Juvet, Regulation of antigen-expressing dendritic cells by double negative regulatory T cells, Eur J Immunol, vol.41, issue.9, pp.2699-2708, 2011.

S. Voelkl, R. Gary, and A. Mackensen, Characterization of the immunoregulatory function of human TCR-??+ CD4-CD8-double-negative T cells, Eur J Immunol, vol.41, issue.3, pp.739-748, 2011.

Z. Mciver, B. Serio, and A. Dunbar, Double-negative regulatory T cells induce allotolerance when expanded after allogeneic haematopoietic stem cell transplantation, Br J Haematol, vol.141, issue.2, pp.170-178, 2008.

D. Zhang, W. Yang, N. Degauque, Y. Tian, A. Mikita et al., New differentiation pathway for double-negative regulatory T cells that regulates the magnitude of immune responses, Blood, vol.109, issue.9, pp.4071-4079, 2007.

. Hertl, Transplantation de cellules souches, 2018.

B. Martín-antonio, M. Granell, and A. Urbano-ispizua, Genomic polymorphisms of the innate immune system and allogeneic stem cell transplantation, Expert Rev Hematol, 2010.

Z. Mciver, B. Serio, and A. Dunbar, Double-negative regulatory T cells induce allotolerance when expanded after allogeneic haematopoietic stem cell transplantation, Br J Haematol, vol.141, issue.2, pp.170-178, 2008.

H. Ye, Y. Chang, X. Zhao, and X. Huang, Characterization of CD3+CD4-CD8-(double negative) T cells reconstitution in patients following hematopoietic stem-cell transplantation, Transpl Immunol, vol.25, issue.4, pp.180-186, 2011.

A. J. Ligocki and J. Y. Niederkorn, Advances on Non-CD4 + Foxp3+ T Regulatory Cells: CD8+, Type 1, and Double Negative T Regulatory Cells in Organ Transplantation, Transplantation, vol.99, issue.8, pp.1553-1559, 2015.

J. P. Moorman, D. Prayther, D. Mcvay, Y. S. Hahn, and C. S. Hahn, The C-terminal region of hepatitis C core protein is required for Fas-ligand independent apoptosis in Jurkat cells by facilitating Fas oligomerization, Virology, vol.312, issue.2, pp.320-329, 2003.

C. Schütz, K. Fischer, and S. Völkl, A new flow cytometric assay for the simultaneous analysis of antigen-specific elimination of T cells in heterogeneous T cell populations, J Immunol Methods, vol.344, issue.2, pp.98-108, 2009.

K. Nagase, R. Shirai, and T. Okawa, CD4/CD8 Double-negative Mycosis Fungoides Mimicking Erythema Gyratum Repens in a Patient with Underlying Lung Cancer, Nagase K, Acta Derm Venereol, 2014.

M. T. Fierro, M. Novelli, and P. Savoia, CD45RA+ immunophenotype in mycosis fungoides: clinical, histological and immunophenotypical features in 22 patients, J Cutan Pathol, vol.28, issue.7, pp.356-362, 2001.

A. Ito, K. Sugita, A. Ikeda, and O. Yamamoto, CD4/CD8 Double-negative Mycosis Fungoides: A Case Report and Literature Review, Yonago Acta Med, vol.62, issue.1, pp.153-158, 2019.

E. Hodak, M. David, L. Maron, A. Aviram, E. Kaganovsky et al., CD4/CD8 double-negative epidermotropic cutaneous T-cell lymphoma: an immunohistochemical variant of mycosis fungoides, J Am Acad Dermatol, vol.55, issue.2, pp.276-284, 2006.

C. Massone, G. Crisman, H. Kerl, and L. Cerroni, The prognosis of early mycosis fungoides is not influenced by phenotype and Tcell clonality, Br J Dermatol, vol.159, issue.4, pp.881-886, 2008.

C. B. Cooke, L. Krenacs, and M. Stetler-stevenson, Hepatosplenic T-cell lymphoma: a distinct clinicopathologic entity of cytotoxic gamma delta T-cell origin, Blood, vol.88, issue.11, pp.4265-4274, 1996.

A. C. Mackey, L. Green, L. C. Liang, P. Dinndorf, and M. Avigan, Hepatosplenic T cell lymphoma associated with infliximab use in young patients treated for inflammatory bowel disease, J Pediatr Gastroenterol Nutr, vol.44, issue.2, pp.265-267, 2007.

F. Suarez, I. Wlodarska, and F. Rigal-huguet, Hepatosplenic alphabeta T-cell lymphoma: an unusual case with clinical, histologic, and cytogenetic features of gammadelta hepatosplenic T-cell lymphoma, Am J Surg Pathol, vol.24, issue.7, pp.1027-1032, 2000.

F. I. Alsohaibani, M. A. Abdulla, and M. M. Fagih, Hepatosplenic T-cell lymphoma, Indian J Hematol Blood Transfus, vol.27, issue.1, pp.39-42, 2011.

A. Khader, M. Shaan, S. Balakrishnan, B. Ambooken, K. Muhammed et al., Multifaceted adult T-cell leukemia/lymphoma in India: a case series, Indian J Dermatol, vol.60, issue.1, p.103, 2015.

E. Matutes, Adult T-cell leukaemia/lymphoma, J Clin Pathol, vol.60, issue.12, pp.1373-1377, 2007.

T. H. Lin, H. C. Wu, Y. C. Hsieh, C. E. Tseng, R. Ichinohasama et al., CD4 and CD8 double-negative adult T-cell leukemia/lymphoma with monomorphic cells expressing CD99: a diagnostic challenge in a country non-endemic for human T-cell leukemia virus, Pathol Int, vol.63, issue.2, pp.132-137, 2013.

F. I. Alsohaibani, M. A. Abdulla, and M. M. Fagih, Hepatosplenic T-cell lymphoma, Indian J Hematol Blood Transfus, vol.27, issue.1, pp.39-42, 2011.

M. Ilic and I. Ilic, Epidemiology of pancreatic cancer, World J Gastroenterol, vol.22, issue.44, pp.9694-9705, 2016.

Y. Lu, P. Hu, and H. Zhou, Double-negative T Cells Inhibit Proliferation and Invasion of Human Pancreatic Cancer Cells in Co-culture, Anticancer Res, vol.39, issue.11, pp.5911-5918, 2019.

D. Movia, D. Bazou, and A. Prina-mello, ALI multilayered co-cultures mimic biochemical mechanisms of the cancer cellfibroblast cross-talk involved in NSCLC MultiDrug Resistance, BMC Cancer, vol.19, issue.1, p.854, 2019.

S. Merims, X. Li, and B. Joe, Anti-leukemia effect of ex vivo expanded DNT cells from AML patients: a potential novel autologous T-cell adoptive immunotherapy, Leukemia, vol.25, issue.9, pp.1415-1422, 2011.

A. I. Rojo, P. Rada, and M. Mendiola, The PTEN/NRF2 axis promotes human carcinogenesis, Antioxid Redox Signal, vol.21, issue.18, pp.2498-2514, 2014.

R. Ferraz, C. F. Cunha, and M. Pimentel, CD3 + CD4 neg CD8 neg (double negative) T lymphocytes and NKT cells as the main cytotoxic-related-CD107a + cells in lesions of cutaneous leishmaniasis caused by Leishmania (Viannia) braziliensis. Parasit Vectors, vol.10, p.219, 2017.

, Maladies-auto-immunes

D. Brandt and C. M. Hedrich, TCR ?? + CD3 + CD4 -CD8 -(double negative) T cells in autoimmunity, Autoimmun Rev, vol.17, issue.4, pp.422-430, 2018.

G. C. Tsokos, Systemic lupus erythematosus, N Engl J Med, vol.365, issue.22, pp.2110-2121, 2011.

J. C. Crispín, M. Oukka, and G. Bayliss, Expanded double negative T cells in patients with systemic lupus erythematosus produce IL-17 and infiltrate the kidneys, J Immunol, vol.181, issue.12, pp.8761-8766, 2008.

P. A. Sieling, S. A. Porcelli, and B. T. Duong, Human double-negative T cells in systemic lupus erythematosus provide help for IgG and are restricted by CD1c, J Immunol, vol.165, issue.9, pp.5338-5344, 2000.

Z. W. Lai, R. Borsuk, and A. Shadakshari, Mechanistic target of rapamycin activation triggers IL-4 production and necrotic death of double-negative T cells in patients with systemic lupus erythematosus, J Immunol, vol.191, issue.5, pp.2236-2246, 2013.

T. Koga, C. M. Hedrich, and M. Mizui, CaMK4-dependent activation of AKT/mTOR and CREM-? underlies autoimmunityassociated Th17 imbalance, J Clin Invest, vol.124, issue.5, pp.2234-2245, 2014.

Y. Gan, X. Zhao, and J. He, Increased Interleukin-17F is Associated with Elevated Autoantibody Levels and More Clinically Relevant Than Interleukin-17A in Primary Sjögren's Syndrome, J Immunol Res, vol.2017, p.4768408, 2017.

A. Alunno, O. Bistoni, and E. Bartoloni, IL-17-producing CD4-CD8-T cells are expanded in the peripheral blood, infiltrate salivary glands and are resistant to corticosteroids in patients with primary Sjogren's syndrome, Ann Rheum Dis, vol.72, issue.2, pp.286-292, 2013.

F. Mackay and P. Schneider, Cracking the BAFF code, Nat Rev Immunol, vol.9, issue.7, p.491, 2009.

M. Harigai, M. Kawamoto, M. Hara, T. Kubota, N. Kamatani et al., Excessive production of IFN-gamma in patients with systemic lupus erythematosus and its contribution to induction of B lymphocyte stimulator/B cell-activating factor/TNF ligand superfamily-13B, J Immunol, vol.181, issue.3, pp.2211-2219, 2008.

J. C. Crispín and G. C. Tsokos, Human TCR-alpha beta+ CD4-CD8-T cells can derive from CD8+ T cells and display an inflammatory effector phenotype, J Immunol, vol.183, issue.7, pp.4675-4681, 2009.

D. Brandt, M. Sergon, S. Abraham, K. Mäbert, and C. M. Hedrich, TCR + CD3 + CD4 -CD8 -effector T cells in psoriasis, Clin Immunol, vol.181, pp.51-59, 2017.

I. Grozdev, N. Korman, and N. Tsankov, Psoriasis as a systemic disease, Clin Dermatol, vol.32, issue.3, pp.343-350, 2014.

M. A. Lowes, M. Suárez-fariñas, and J. G. Krueger, Immunology of psoriasis, Annu Rev Immunol, vol.32, pp.227-255, 2014.

A. Ueyama, C. Imura, and Y. Fusamae, Potential role of IL-17-producing CD4/CD8 double negative ?? T cells in psoriatic skin inflammation in a TPA-induced STAT3C transgenic mouse model, PD-1/PD-L and autoimmunity:a growing relationship. Zamani MR, vol.85, pp.27-35, 2016.

F. Rieux-laucat, Le syndrome lymphoprolifératif avec auto-immunité: un défaut hérité ou acquis d'apoptose lymphocytaire

, Med Sci (Paris), vol.22, issue.6-7, pp.645-650, 2006.

M. C. Sneller, J. Wang, and J. K. Dale, Clincal, immunologic, and genetic features of an autoimmune lymphoproliferative syndrome associated with abnormal lymphocyte apoptosis, Blood, vol.89, issue.4, pp.1341-1348, 1997.

S. E. Straus, E. S. Jaffe, and J. M. Puck, The development of lymphomas in families with autoimmune lymphoproliferative syndrome with germline Fas mutations and defective lymphocyte apoptosis, Blood, vol.98, issue.1, pp.194-200, 2001.

S. E. Straus, E. S. Jaffe, and J. M. Puck, The development of lymphomas in families with autoimmune lymphoproliferative syndrome with germline Fas mutations and defective lymphocyte apoptosis, Blood, vol.98, issue.1, pp.194-200, 2001.

J. B. Oliveira, J. J. Bleesing, and U. Dianzani, Revised diagnostic criteria and classification for the autoimmune lymphoproliferative syndrome (ALPS): report from the 2009 NIH International Workshop, Blood, vol.116, issue.14, pp.35-40, 2010.

C. E. Jackson, R. E. Fischer, and A. P. Hsu, Autoimmune lymphoproliferative syndrome with defective Fas: genotype influences penetrance, Am J Hum Genet, vol.64, issue.4, pp.1002-1014, 1999.

, Normalfunction gène FAS génetic home ref

A. Martin-villalba, I. Herr, and J. I. , CD95 ligand (Fas-L/APO-1L) and tumor necrosis factor-related apoptosisinducing ligand mediate ischemia-induced apoptosis in neurons, J Neurosci, vol.19, issue.10, pp.3809-3817, 1999.

K. Fischer, S. Voelkl, and J. Heymann, Isolation and characterization of human antigen-specific TCR alpha beta+ CD4(-)CD8-double-negative regulatory T cells, Blood, vol.105, issue.7, pp.2828-2835, 2005.

R. M. Siegel, J. K. Frederiksen, and D. A. Zacharias, Fas preassociation required for apoptosis signaling and dominant inhibition by pathogenic mutations, Science, vol.288, issue.5475, pp.2354-2357, 2000.

H. S. Kuehn, I. Caminha, and J. E. Niemela, FAS haploinsufficiency is a common disease mechanism in the human autoimmune lymphoproliferative syndrome, J Immunol, vol.186, issue.10, pp.6035-6043, 2011.

M. Van-der-burg, R. De-groot, and W. M. Comans-bitter, Autoimmune lymphoproliferative syndrome (ALPS) in a child from consanguineous parents: a dominant or recessive disease, Pediatr Res, vol.47, issue.3, pp.336-343, 2000.

Z. Y. Kucuk, L. M. Charbonnier, R. L. Mcmasters, T. Chatila, and J. J. Bleesing, CTLA-4 haploinsufficiency in a patient with an autoimmune lymphoproliferative disorder, J Allergy Clin Immunol, vol.140, issue.3, pp.862-864, 2017.

L. L. Bi, G. Pan, and T. P. Atkinson, Dominant inhibition of Fas ligand-mediated apoptosis due to a heterozygous mutation associated with autoimmune lymphoproliferative syndrome (ALPS) Type Ib, BMC Med Genet, vol.8, p.41, 2007.

D. Sanséau, Atlas of Genetics and Cytogenetics in Oncology and Haematology, 2014.

D. R. Matson and D. T. Yang, Autoimmune Lymphoproliferative Syndrome: An Overview, Arch Pathol Lab Med, vol.144, issue.2, pp.245-251, 2020.

R. Strich, Programmed Cell Death Initiation and Execution in Budding Yeast, Genetics, vol.200, issue.4, pp.1003-1014, 2015.

S. Poppema, E. Maggio, and A. Van-den-berg, Development of lymphoma in Autoimmune Lymphoproliferative Syndrome (ALPS) and its relationship to Fas gene mutations, Leuk Lymphoma, vol.45, issue.3, pp.423-431, 2004.

A. Pépin, Régulation de l'apoptose des lymphocytes T par les protéines de la famille TSC22D. Sciences agricoles, 2011.

K. Bride and D. Teachey, Autoimmune lymphoproliferative syndrome: more than a FAScinating disease, vol.6, 1000.

F. Rieux-laucat, Le syndrome lymphoprolifératif avec auto-immunité : un défaut hérité ou acquis d'apoptose lymphocytaire

, Med Sci (Paris), vol.22, issue.6-7, pp.645-650, 2006.

J. B. Oliveira, J. J. Bleesing, and U. Dianzani, Revised diagnostic criteria and classification for the autoimmune lymphoproliferative syndrome (ALPS): report from the 2009 NIH International Workshop, Blood, vol.116, issue.14, pp.35-40, 2010.

I. Caminha, T. A. Fleisher, and R. L. Hornung, Using biomarkers to predict the presence of FAS mutations in patients with features of the autoimmune lymphoproliferative syndrome, J Allergy Clin Immunol, vol.125, issue.4, pp.946-949, 2010.

A. E. Seif, C. S. Manno, C. Sheen, S. A. Grupp, and D. T. Teachey, Identifying autoimmune lymphoproliferative syndrome in children with Evans syndrome: a multi-institutional study, Blood, vol.115, issue.11, pp.2142-2145, 2010.

S. Sava?an, I. Warrier, S. Buck, J. Kaplan, and Y. Ravindranath, Increased lymphocyte Fas expression and high incidence of common variable immunodeficiency disorder in childhood Evans' syndrome, Clin Immunol, vol.125, issue.3, pp.224-229, 2007.

J. B. Oliveira, J. J. Bleesing, and U. Dianzani, Revised diagnostic criteria and classification for the autoimmune lymphoproliferative syndrome (ALPS): report from the 2009 NIH International Workshop, Blood, vol.116, issue.14, pp.35-40, 2010.

A. Magerus-chatinet, M. C. Stolzenberg, and M. S. Loffredo, FAS-L, IL-10, and double-negative CD4-CD8-TCR alpha/beta+ T cells are reliable markers of autoimmune lymphoproliferative syndrome (ALPS) associated with FAS loss of function, Blood, vol.113, issue.13, pp.3027-3030, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00397943

C. Speckmann, K. Lehmberg, and M. H. Albert, X-linked inhibitor of apoptosis (XIAP) deficiency: the spectrum of presenting manifestations beyond hemophagocytic lymphohistiocytosis, Clin Immunol, vol.149, issue.1, pp.133-141, 2013.

H. S. Kuehn, I. Caminha, and J. E. Niemela, FAS haploinsufficiency is a common disease mechanism in the human autoimmune lymphoproliferative syndrome, J Immunol, vol.186, issue.10, pp.6035-6043, 2011.

S. Poppema, Development of lymphoma in Autoimmune Lymphoproliferative Syndrome (ALPS) and its relationship to Fas gene mutations, Leukemia & lymphoma, 2004.

A. Chadburn, J. Said, and D. Gratzinger, HHV8/KSHV-Positive Lymphoproliferative Disorders and the Spectrum of Plasmablastic and Plasma Cell Neoplasms: 2015 SH/EAHP Workshop Report-Part, vol.3, pp.171-187, 2017.

D. C. Fajgenbaum, F. Van-rhee, and C. S. Nabel, HHV-8-negative, idiopathic multicentric Castleman disease: novel insights into biology, pathogenesis, and therapy, Blood, vol.123, pp.2924-2933, 2014.

C. S. Nabel, S. Sameroff, and D. Shilling, Virome capture sequencing does not identify active viral infection in unicentric and idiopathic multicentric Castleman disease, PLoS One, vol.14, issue.6, p.218660, 2019.

Z. Sbihi, A. Dossier, and D. Boutboul, iNKT and memory B-cell alterations in HHV-8 multicentric Castleman disease, Blood, vol.129, issue.7, pp.855-865, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01515291

Z. Li, X. Lan, and C. Li, Recurrent PDGFRB mutations in unicentric Castleman disease, Leukemia, vol.33, issue.4, pp.1035-1038, 2019.

D. R. Matson and P. Md, Autoimmune Lymphoproliferative Syndrome: An Overview, Arch Pathol Lab Med, vol.144, issue.2, pp.245-251, 2020.

E. Boggio, N. Clemente, and A. Mondino, IL-17 protects T cells from apoptosis and contributes to development of ALPSlike phenotypes, Blood, vol.123, issue.8, pp.1178-1186, 2014.

B. Neven, J. Bruneau, and M. C. Stolzenberg, Defective anti-polysaccharide response and splenic marginal zone disorganization in ALPS patients, Blood, vol.124, issue.10, pp.1597-1609, 2014.

J. B. Oliveira, N. Bidère, and J. E. Niemela, NRAS mutation causes a human autoimmune lymphoproliferative syndrome, Proc Natl Acad Sci, vol.104, issue.21, pp.8953-8958, 2007.

H. J. Chun, L. Zheng, and M. Ahmad, Pleiotropic defects in lymphocyte activation caused by caspase-8 mutations lead to human immunodeficiency, Nature, vol.419, issue.6905, pp.395-399, 2002.

. Ta-fleisher, JB Oliveira Autoimmune lymphoproliferative syndrome.. Primary Immunodeficiency Diseases books, 2013.

D. S. Thommen, J. Schreiner, and P. Müller, Progression of Lung Cancer Is Associated with Increased Dysfunction of T Cells Defined by Coexpression of Multiple Inhibitory Receptors, Cancer Immunol Res, vol.3, issue.12, pp.1344-1355, 2015.

B. Piqueras, C. Lavenu-bombled, and L. Galicier, Common variable immunodeficiency patient classification based on impaired B cell memory differentiation correlates with clinical aspects, J Clin Immunol, vol.23, issue.5, pp.385-400, 2003.

W. I. Lee, J. L. Huang, and K. W. Yeh, Clinical features and genetic analysis of Taiwanese patients with the hyper IgM syndrome phenotype, Pediatr Infect Dis J, vol.32, issue.9, pp.1010-1016, 2013.

J. C. Latourelle, M. Sun, and M. F. Lew, The Gly2019Ser mutation in LRRK2 is not fully penetrant in familial Parkinson's disease: the GenePD study, BMC Med, vol.6, p.32, 2008.

J. D. Milner, T. P. Vogel, and L. Forbes, Early-onset lymphoproliferation and autoimmunity caused by germline STAT3 gainof-function mutations, Blood, vol.125, issue.4, pp.591-599, 2015.

V. K. Rao and J. B. Oliveira, How I treat autoimmune lymphoproliferative syndrome, Blood, vol.118, issue.22, pp.5741-5751, 2011.

K. L. Bride, T. Vincent, and K. Smith-whitley, Sirolimus is effective in relapsed/refractory autoimmune cytopenias: results of a prospective multi-institutional trial, Blood, vol.127, issue.1, pp.17-28, 2016.

K. C. Dowdell, J. E. Niemela, and S. Price, Somatic FAS mutations are common in patients with genetically undefined autoimmune lymphoproliferative syndrome, Blood, vol.115, issue.25, pp.5164-5169, 2010.

F. Rieux-laucat, What's up in the ALPS, Curr Opin Immunol, vol.49, pp.79-86, 2017.

C. Tourrel-cuzin and B. Gausseres, Principe de la cytométrie en flux et du tri cellulaire, Unité de Biologie fonctionnelle et adaptative -CNRS UMR 8251, 2020.