L. Herman, D. Gerbert, L. Larson, M. Leger, and R. Mcnellis, Vaccines, thimerosal, and neurodevelopmental outcomes, Journal of the American Academy of Physician Assistants, vol.19, issue.1, pp.16-18, 2006.
DOI : 10.1097/01720610-200601000-00004

D. Geier, L. Sykes, and M. Geier, A Review of Thimerosal (Merthiolate) and its Ethylmercury Breakdown Product: Specific Historical Considerations Regarding Safety and Effectiveness, Journal of Toxicology and Environmental Health, Part B, vol.23, issue.8, pp.575-596, 2007.
DOI : 10.1002/ajim.4700050308

A. Chatterjee, O. Keefe, and C. , Current controversies in the USA regarding vaccine safety, Expert Review of Vaccines, vol.9, issue.5, pp.497-502, 2010.
DOI : 10.1586/erv.10.36

L. Ball, R. Ball, and R. Pratt, An Assessment of Thimerosal Use in Childhood Vaccines, PEDIATRICS, vol.107, issue.5, pp.1147-1154, 2001.
DOI : 10.1542/peds.107.5.1147

J. Barile, G. Kuperminc, E. Weintraub, J. Mink, and W. Thompson, Thimerosal Exposure in Early Life and Neuropsychological Outcomes 7-10 Years Later, Journal of Pediatric Psychology, vol.37, issue.1, pp.106-118, 2012.
DOI : 10.1093/jpepsy/jsr048

J. Dorea, M. Farina, and J. Rocha, Toxicity of ethylmercury (and Thimerosal): a comparison with methylmercury, Journal of Applied Toxicology, vol.120, issue.9347, pp.700-711, 2013.
DOI : 10.1002/jat.2855

L. Yel, L. Brown, K. Su, S. Gollapudi, and S. Gupta, Thimerosal induces neuronal cell apoptosis by causing cytochrome c and apoptosis-inducing factor release from mitochondria, International Journal of Molecular Medicine, vol.16, pp.971-977, 2005.
DOI : 10.3892/ijmm.16.6.971

D. Baskin, H. Ngo, and V. Didenko, Thimerosal Induces DNA Breaks, Caspase-3 Activation, Membrane Damage, and Cell Death in Cultured Human Neurons and Fibroblasts, Toxicological Sciences, vol.74, issue.2, pp.361-368, 2003.
DOI : 10.1093/toxsci/kfg126

M. Humphrey, M. Cole, J. Pendergrass, and K. Kiningham, Mitochondrial Mediated Thimerosal-Induced Apoptosis in a Human Neuroblastoma Cell Line (SK-N-SH), NeuroToxicology, vol.26, issue.3, pp.407-416, 2005.
DOI : 10.1016/j.neuro.2005.03.008

S. Lee, M. Mian, H. Lee, C. Kang, and J. Kim, Thimerosal induces oxidative stress in HeLa S epithelial cells, Environmental Toxicology and Pharmacology, vol.22, issue.2, pp.194-199, 2006.
DOI : 10.1016/j.etap.2006.03.003

W. Li, S. Chen, L. Chen, G. Yang, and J. Li, Thimerosal-Induced Apoptosis in Mouse C2C12 Myoblast Cells Occurs through Suppression of the PI3K/Akt/Survivin Pathway, PLoS ONE, vol.8, issue.11, p.49064, 2012.
DOI : 10.1371/journal.pone.0049064.g008

S. Trompezinski, C. Migdal, M. Tailhardat, L. Varlet, B. Courtellemont et al., Characterization of early events involved in human dendritic cell maturation induced by sensitizers: Cross talk between MAPK signalling pathways, Toxicology and Applied Pharmacology, vol.230, issue.3, pp.397-406, 2008.
DOI : 10.1016/j.taap.2008.03.012

A. Agrawal, P. Kaushal, S. Agrawal, S. Gollapudi, and S. Gupta, Thimerosal induces TH2 responses via influencing cytokine secretion by human dendritic cells, Journal of Leukocyte Biology, vol.81, issue.2, pp.474-482, 2007.
DOI : 10.1189/jlb.0706467

S. Makani, S. Gollapudi, L. Yel, S. Chiplunkar, and S. Gupta, Biochemical and molecular basis of thimerosal-induced apoptosis in T cells: a major role of mitochondrial pathway, Genes and Immunity, vol.3, issue.5, pp.270-278, 2002.
DOI : 10.1038/sj.gene.6363854

G. Guzzi, P. Pigatto, F. Spadari, L. Porta, and C. , Effect of thimerosal, methylmercury, and mercuric chloride in Jurkat T Cell Line, Interdisciplinary Toxicology, vol.5, issue.3, pp.159-161, 2012.
DOI : 10.2478/v10102-012-0026-1

L. Buanec, H. Gougeon, M. Mathian, A. Lebon, P. Dupont et al., IFN-?? and CD46 stimulation are associated with active lupus and skew natural T regulatory cell differentiation to type 1 regulatory T (Tr1) cells, Proceedings of the National Academy of Sciences, vol.108, issue.47, pp.18995-19000, 2011.
DOI : 10.1073/pnas.1113301108

H. Lecoeur, E. Ledru, M. Prevost, and M. Gougeon, Strategies for phenotyping apoptotic peripheral human lymphocytes comparing ISNT, annexin-V and 7-AAD cytofluorometric staining methods, Journal of Immunological Methods, vol.209, issue.2, pp.111-123, 1997.
DOI : 10.1016/S0022-1759(97)00138-5

H. Lecoeur, M. Melki, H. Saidi, and M. Gougeon, Chapter Three Analysis of Apoptotic Pathways by Multiparametric Flow Cytometry: Application to HIV Infection, Methods in enzymology, vol.442, pp.51-82, 2008.
DOI : 10.1016/S0076-6879(08)01403-1

P. Petit, H. Lecoeur, E. Zorn, C. Dauguet, and B. Mignotte, Alterations in mitochondrial structure and function are early events of dexamethasone-induced thymocyte apoptosis, The Journal of Cell Biology, vol.130, issue.1, pp.157-167, 1995.
DOI : 10.1083/jcb.130.1.157

G. Rothe and G. Valet, Flow cytometric analysis of respiratory burst activity in phagocytes with hydroethidine and 29,79-dichlorofluorescin, Journal of leukocyte biology, vol.47, pp.440-448, 1990.

J. Torres-roca, H. Lecoeur, C. Amatore, and M. Gougeon, The early intracellular production of a reactive oxygen intermediate mediates apoptosis in dexamethasone-treated thymocytes, Cell death and differentiation, vol.2, pp.309-319, 1995.

A. Cossarizza, R. Ferraresi, L. Troiano, E. Roat, and L. Gibellini, Simultaneous analysis of reactive oxygen species and reduced glutathione content in living cells by polychromatic flow cytometry, Nature Protocols, vol.10, issue.12, pp.1790-1797, 2009.
DOI : 10.1097/INF.0b013e31814689be

C. Seror, M. Melki, F. Subra, S. Raza, and M. Bras, Extracellular ATP acts on P2Y2 purinergic receptors to facilitate HIV-1 infection, The Journal of Experimental Medicine, vol.50, issue.9, pp.1823-1834, 2011.
DOI : 10.1128/CVI.00166-07

L. Chen, Mitochondrial Membrane Potential in Living Cells, Annual Review of Cell Biology, vol.4, issue.1, pp.155-181, 1988.
DOI : 10.1146/annurev.cb.04.110188.001103

C. Ferraro-peyret, L. Quemeneur, M. Flacher, J. Revillard, and L. Genestier, Caspase-Independent Phosphatidylserine Exposure During Apoptosis of Primary T Lymphocytes, The Journal of Immunology, vol.169, issue.9, pp.4805-4810, 2002.
DOI : 10.4049/jimmunol.169.9.4805

M. Tafani, D. Minchenko, A. Serroni, and J. Farber, Induction of the mitochondrial permeability transition mediates the killing of HeLa cells by staurosporine, Cancer research, vol.61, pp.2459-2466, 2001.

T. Caserta, A. Smith, A. Gultice, M. Reedy, and T. Brown, Q-VD-OPh, a broad spectrum caspase inhibitor with potent antiapoptotic properties, APOPTOSIS, vol.8, issue.4, pp.345-352, 2003.
DOI : 10.1023/A:1024116916932

J. Cai and D. Jones, Superoxide in Apoptosis. MITOCHONDRIAL GENERATION TRIGGERED BY CYTOCHROME c LOSS, Journal of Biological Chemistry, vol.273, issue.19, pp.11401-11404, 1998.
DOI : 10.1074/jbc.273.19.11401

H. Dussmann, D. Kogel, M. Rehm, and J. Prehn, Mitochondrial Membrane Permeabilization and Superoxide Production during Apoptosis: A SINGLE-CELL ANALYSIS, Journal of Biological Chemistry, vol.278, issue.15, pp.12645-12649, 2003.
DOI : 10.1074/jbc.M210826200

A. Wells, H. Gudmundsdottir, and L. Turka, Following the fate of individual T cells throughout activation and clonal expansion. Signals from T cell receptor and CD28 differentially regulate the induction and duration of a proliferative response., Journal of Clinical Investigation, vol.100, issue.12, pp.3173-3183, 1997.
DOI : 10.1172/JCI119873

S. Masters, L. Mielke, A. Cornish, C. Sutton, O. Donnell et al., Regulation of interleukin-1?? by interferon-?? is species specific, limited by suppressor of cytokine signalling 1 and influences interleukin-17 production, EMBO reports, vol.144, issue.8, pp.640-646, 2010.
DOI : 10.4049/jimmunol.0803227

A. Chauvat, N. Benhamouda, E. Loison, M. Gougeon, and A. Gey, Pitfalls in anti-influenza T cell detection by Elispot using thimerosal containing pandemic H1N1 vaccine as antigen, Journal of Immunological Methods, vol.378, issue.1-2, pp.81-87, 2012.
DOI : 10.1016/j.jim.2012.02.008

S. Liu, C. Huang, C. Huang, B. Wang, and P. Chang, Thimerosal-Induced Apoptosis in Human SCM1 Gastric Cancer Cells: Activation of p38 MAP Kinase and Caspase-3 Pathways without Involvement of [Ca2+]i Elevation, Toxicological Sciences, vol.100, issue.1, pp.109-117, 2007.
DOI : 10.1093/toxsci/kfm205

M. Yole, M. Wickstrom, and B. Blakley, Cell death and cytotoxic effects in YAC-1 lymphoma cells following exposure to various forms of mercury, Toxicology, vol.231, issue.1, pp.40-57, 2007.
DOI : 10.1016/j.tox.2006.11.062

K. Woo, T. Lee, J. Bae, B. Jang, and D. Song, Thimerosal induces apoptosis and G2/M phase arrest in human leukemia cells, Molecular Carcinogenesis, vol.21, issue.9, pp.657-666, 2006.
DOI : 10.1002/mc.20202

G. Kroemer and J. Reed, Mitochondrial control of cell death, Nature Medicine, vol.6, issue.5, pp.513-519, 2000.
DOI : 10.1038/74994

S. Plotkin, Vaccines: correlates of vaccine-induced immunity. Clinical infectious diseases: an official publication of the Infectious Diseases Society of, America, vol.47, pp.401-409, 2008.

J. Mcelhaney, D. Xie, W. Hager, M. Barry, and Y. Wang, T Cell Responses Are Better Correlates of Vaccine Protection in the Elderly, The Journal of Immunology, vol.176, issue.10, pp.6333-6339, 2006.
DOI : 10.4049/jimmunol.176.10.6333

T. Wilkinson, C. Li, C. Chui, A. Huang, and M. Perkins, Preexisting influenza-specific CD4+ T cells correlate with disease protection against influenza challenge in humans, Nature Medicine, vol.94, issue.2, pp.274-280, 2012.
DOI : 10.1128/JVI.79.9.5684-5694.2005

S. Sridhar, S. Begom, A. Bermingham, K. Hoschler, and W. Adamson, Cellular immune correlates of protection against symptomatic pandemic influenza, Nature Medicine, vol.5, issue.10, pp.1305-1312, 2013.
DOI : 10.1038/nm.3350

J. Schoenborn and C. Wilson, Regulation of Interferon????? During Innate and Adaptive Immune Responses, Advances in immunology, vol.96, pp.41-101, 2007.
DOI : 10.1016/S0065-2776(07)96002-2