S. Gagneux, Host-pathogen coevolution in human tuberculosis, Philos Trans R Soc Lond B Biol Sci, vol.367, pp.850-859, 2012.

J. L. Flynn and J. Chan, What's good for the host is good for the bug, Trends Microbiol, vol.13, pp.98-102, 2005.

I. Comas, J. Chakravartti, P. M. Small, J. Galagan, S. Niemann et al., Human T cell epitopes of Mycobacterium tuberculosis are evolutionarily hyperconserved, Nat Genet, vol.42, pp.498-503, 2010.

A. O. Moguche, M. Musvosvi, A. Penn-nicholson, C. R. Plumlee, H. Mearns et al., Antigen availability shapes T cell differentiation and function during tuberculosis, Cell Host Microbe, vol.21, pp.695-706, 2017.

S. H. Kaufmann and A. Dorhoi, Inflammation in tuberculosis: interactions, imba lances and interventions, Curr Opin Immunol, vol.25, issue.4, pp.441-450, 2013.

S. Shafiani, C. Dinh, J. M. Ertelt, A. O. Moguche, I. Siddiqui et al., Pathogen-specific Treg cells expand early during Mycobacterium tuberculosis infection but are later eliminated in response to interleukin-12, Immunity, vol.38, pp.1261-70, 2013.

R. P. Larson, S. Shafiani, and K. B. Urdahl, Foxp3(+) regulatory T cells in tuberculosis, Adv Exp Med Biol, vol.783, 2013.

A. Dorhoi and S. Kaufmann, Pathology and immune reactivity: understanding multidimensionality in pulmonary tuberculosis, Semin Immunopathol, vol.38, pp.153-66, 2016.

O. 'garra, A. Redford, P. S. Mcnab, F. W. Bloom, C. I. Wilkinson et al., The immune response in tuberculosis, Annu Rev Immunol, vol.31, pp.475-527, 2013.

P. K. Saha, P. K. Sharma, S. K. Sharma, A. Singh, and D. K. Mitra, Recruitment of Th1 effector cells in human tuberculosis: hierarchy of chemokine receptor(s) and their ligands, Cytokine, vol.63, pp.43-51, 2013.

L. Arlehamn, C. S. Gerasimova, A. Mele, F. Henderson, R. Swann et al., Memory T cells in latent Mycobacterium tuberculosis infection are directed against three antigenic islands and largely contained in a CXCR3+CCR6+ Th1 subset, PLoS Pathog, vol.9, 2013.

C. L. Fuller, J. L. Flynn, and T. A. Reinhart, In situ study of abundant expression of proinflammatory chemokines and cytokines in pulmonary granulomas that develop in cynomolgus macaques experimentally infected with Mycobacterium tuberculosis, Infect Immun, vol.71, pp.7023-7057, 2003.

C. Riou, P. Peixoto, B. Roberts, L. Ronacher, K. Walzl et al., Effect of standard tuberculosis treatment on plasma cytokine levels in patients with active pulmonary tuberculosis, PLoS One, vol.7, p.36886, 2012.

J. De-steenwinkel, G. J. De-knegt, T. Kate, M. T. Verbrugh, H. A. Ottenhoff et al., Dynamics of interferon-gamma release assay and cytokine profiles in blood and respiratory tract specimens from mice with tuberculosis and the effect of therapy, Eur J Clin Microbiol Infect Dis, issue.6, pp.1195-201, 2012.

A. Azzurri, O. Y. Sow, A. Amedei, B. Bah, S. Diallo et al., IFN-gammainducible protein 10 and pentraxin 3 plasma levels are tools for monitoring disease activity in Mycobacterium tuberculosis infection, Microbes Infect, vol.7, pp.1-8, 2005.

N. P. Juffermans, A. Verbon, S. J. Van-deventer, D. H. Van, J. T. Belisle et al., Elevated chemokine concentrations in sera of human immunodeficiency virus (HIV)-seropositive and HIV-seronegative patients with tuberculosis: a possible role for mycobacterial lipoarabinomannan, Infect Immun, vol.67, pp.4295-4302, 1999.

A. Cannas, L. Calvo, T. Chiacchio, G. Cuzzi, V. Vanini et al., IP-10 detection in urine is associated with lung diseases, BMC Infect Dis, vol.10, p.333, 2010.
DOI : 10.1186/1471-2334-10-333

URL : https://bmcinfectdis.biomedcentral.com/track/pdf/10.1186/1471-2334-10-333?site=bmcinfectdis.biomedcentral.com

D. Goletti, M. R. Lee, J. Y. Wang, N. Walter, and T. Ottenhoff, Update on tuberculosis biomarkers: from correlates of risk, to correlates of active disease and of cure from disease, Respirology, vol.23, pp.455-66, 2018.

M. Liu, S. Guo, J. M. Hibbert, V. Jain, N. Singh et al., CXCL10/IP-10 in infectious diseases pathogenesis and potential therapeutic implications, Cytokine Growth Factor Rev, vol.22, pp.121-151, 2011.
DOI : 10.1016/j.cytogfr.2011.06.001

URL : http://europepmc.org/articles/pmc3203691?pdf=render

M. L. Albert, A. Casrouge, S. Chevaliez, C. Hézode, I. Rosa et al., Interferon induced protein 10 remains a useful biomarker of treatment failure in patients stratified for the interleukin-28B rs12979860 haplotype, Hepatology, vol.53, pp.1410-1411, 2011.

A. Casrouge, J. Decalf, M. Ahloulay, C. Lababidi, H. Mansour et al., Evidence for an antagonist form of the chemokine CXCL10 in patients chronically infected with HCV, J Clin Invest, vol.121, pp.308-325, 2011.
URL : https://hal.archives-ouvertes.fr/pasteur-01402089

J. Decalf, K. V. Tarbell, A. Casrouge, J. D. Price, G. Linder et al., Inhibition of DPP4 activity in humans establishes its in vivo role in CXCL10 posttranslational modification: prospective placebo-controlled clinical studies, EMBO Mol Med, vol.8, pp.679-83, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-01380633

R. Barreira-da-silva, M. E. Laird, N. Yatim, L. Fiette, M. A. Ingersoll et al., Dipeptidylpeptidase 4 inhibition enhances lymphocyte trafficking, improving both naturally occurring tumor immunity and immunotherapy, Nat Immunol, vol.16, pp.850-858, 2015.
URL : https://hal.archives-ouvertes.fr/pasteur-01380810

C. Klemann, L. Wagner, M. Stephan, and S. Von-hörsten, Cut to the chase: a review of CD26/dipeptidyl peptidase-4's (DPP4) entanglement in the immune system, Clin Exp Immunol, vol.185, pp.1-21, 2016.

P. Proost, E. Schutyser, P. Menten, S. Struyf, A. Wuyts et al., Amino-terminal truncation of CXCR3 agonists impairs receptor signaling and lymphocyte chemotaxis, while preserving antiangiogenic properties, Blood, vol.98, pp.3554-61, 2001.
DOI : 10.1182/blood.v98.13.3554

URL : http://www.bloodjournal.org/content/98/13/3554.full.pdf

P. Proost, S. Struyf, J. Van-damme, P. Fiten, E. Ugarte-berzal et al., Chemokine isoforms and processing in inflammation and immunity, J Autoimmun, vol.85, pp.45-57, 2017.
DOI : 10.1016/j.jaut.2017.06.009

K. Tonby, M. Ruhwald, D. Kvale, and A. M. Dyrhol-riise, IP-10 measured by Dry Plasma Spots as biomarker for therapy responses in Mycobacterium tuberculosis infection, Sci Rep, vol.5, p.9223, 2015.
DOI : 10.1038/srep09223

URL : http://www.nature.com/articles/srep09223.pdf

E. G. Meissner, J. Decalf, A. Casrouge, H. Masur, S. Kottilil et al., Dynamic changes of post-translationally modified forms of CXCL10 and soluble DPP4 in HCV subjects receiving interferon-free therapy, PLoS One, vol.10, 2015.
URL : https://hal.archives-ouvertes.fr/pasteur-01380964

H. Wang, J. Yue, J. Yang, R. Gao, and J. Liu, Clinical diagnostic utility of adenosine deaminase, interferon-?, interferon-?-induced protein of 10 kDa, and dipeptidyl peptidase 4 levels in tuberculous pleural effusions, Heart Lung, vol.41, issue.1, pp.70-75, 2012.

D. Scheel-toellner, E. Richter, K. M. Toellner, N. Reiling, H. H. Wacker et al., CD26 expression in leprosy and other granulomatous diseases correlates with the production of interferon-gamma, Lab Invest, vol.73, pp.685-90, 1995.

G. Kaplan, A. D. Luster, G. Hancock, and Z. A. Cohn, The expression of a gamma interferon-induced protein (IP-10) in delayed immune responses in human skin, J Exp Med, vol.166, pp.1098-108, 1987.

O. J. Cordero, F. J. Salgado, and M. Nogueira, On the origin of serum CD26 and its altered concentration in cancer patients, Cancer Immunol Immunother, vol.58, pp.1723-1770, 2009.

O. J. Cordero, R. Varela-calviño, T. López-gonzález, C. Calviño-sampedro, J. E. Viñuela et al., CD26 expression on T helper populations and sCD26 serum levels in patients with rheumatoid arthritis, PLoS One, vol.10, 2015.
DOI : 10.1371/journal.pone.0131992

URL : https://doi.org/10.1371/journal.pone.0131992

R. S. Wallis and R. Hafner, Advancing host-directed therapy for tuberculosis, Nat Rev Immunol, vol.15, pp.255-63, 2015.
DOI : 10.1038/nri3813

S. Sakai, K. D. Mayer-barber, and D. L. Barber, Defining features of protective CD4 T cell responses to Mycobacterium tuberculosis, Curr Opin Immunol, vol.29, pp.137-179, 2014.
DOI : 10.1016/j.coi.2014.06.003

URL : http://europepmc.org/articles/pmc4122329?pdf=render

S. Sakai, K. D. Kauffman, M. A. Sallin, A. H. Sharpe, H. A. Young et al., CD4 T cell-derived IFN-? plays a minimal role in control of pulmonary Mycobacterium tuberculosis infection and must be actively repressed by PD-1 to prevent lethal disease, PLoS Pathog, vol.12, 2016.

J. S. Woodworth, S. B. Cohen, A. O. Moguche, C. R. Plumlee, E. M. Agger et al., Subunit vaccine H56/CAF01 induces a population of circulating CD4 T cells that traffic into the Mycobacterium tuberculosis-infected lung, Mucosal Immunol, vol.10, issue.2, pp.555-64, 2017.

P. Andersen and J. S. Woodworth, Tuberculosis vaccines-rethinking the current paradigm, Trends Immunol, vol.35, pp.387-95, 2014.

C. R. Horsburgh, C. E. Barry, and C. Lange, Treatment of tuberculosis, N Engl J Med, vol.373, pp.2149-60, 2015.

T. Karagiannis, P. Boura, and A. Tsapas, Safety of dipeptidyl peptidase 4 inhibitors: a perspective review, Ther Adv Drug Saf, vol.5, pp.138-184, 2014.

K. Gooßen and S. Gräber, Longer term safety of dipeptidyl peptidase-4 inhibitors in patients with type 2 diabetes mellitus: systematic review and meta-analysis, Diabetes Obes Metab, vol.14, pp.1061-72, 2012.

M. Monici, Cell and tissue autofluorescence research and diagnostic applications, Biotechnol Annu Rev, vol.11, pp.11007-11009, 2005.

W. Baschong, R. Suetterlin, and R. H. Laeng, Control of autofluorescence of archival formaldehyde-fixed, paraffin-embedded tissue in confocal laser scanning microscopy (CLSM), J Histochem Cytochem, vol.49, pp.1565-71, 2001.