F. Caligaris-cappio, The nature of the B lymphocyte in B-chronic lymphocytic leukemia, Blood Cells, vol.19, pp.601-613, 1993.

S. Kitada, I. M. Pedersen, A. D. Schimmer, and J. C. Reed, Dysregulation of apoptosis genes in hematopoietic malignancies, Oncogene, vol.21, pp.3459-3474, 2002.

D. Rossi, Integrated mutational and cytogenetic analysis identifies new prognostic subgroups in chronic lymphocytic leukemia, Blood, vol.121, pp.1403-1412, 2013.

A. Abbaci, Neurotensin receptor type 2 protects B-cell chronic lymphocytic leukemia cells from apoptosis, Oncogene, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01803034

M. Barragán, Involvement of protein kinase C and phosphatidylinositol 3-kinase pathways in the survival of B-cell chronic lymphocytic leukemia cells, Blood, vol.99, pp.2969-2976, 2002.

S. Cuní, A sustained activation of PI3K/NF-kappaB pathway is critical for the survival of chronic lymphocytic leukemia B cells, Leukemia, vol.18, pp.1391-1400, 2004.

J. A. Burger, The CLL Cell Microenvironment, Advances in Chronic Lymphocytic Leukemia, pp.25-45, 2013.

P. E. Patten, CD38 expression in chronic lymphocytic leukemia is regulated by the tumor microenvironment, Blood, vol.111, pp.5173-5181, 2008.

P. Giannoni, An interaction between hepatocyte growth factor and its receptor (c-MET) prolongs the survival of chronic lymphocytic leukemic cells through STAT3 phosphorylation: a potential role of mesenchymal cells in the disease, Haematologica, vol.96, pp.1015-1023, 2011.

J. A. Burger, Blood-derived nurse-like cells protect chronic lymphocytic leukemia B cells from spontaneous apoptosis through stromal cell-derived factor-1, Blood, vol.96, pp.2655-2663, 2000.

F. Boissard, J. Fournié, C. Laurent, M. Poupot, and L. Ysebaert, Nurse like cells: chronic lymphocytic leukemia associated macrophages, Leukemia & Lymphoma, vol.56, pp.1570-1572, 2015.

L. Jia, Extracellular HMGB1 promotes differentiation of nurse-like cells in chronic lymphocytic leukemia, Blood, vol.123, pp.1709-1719, 2014.

A. Polk, Colony-Stimulating Factor-1 Receptor Is Required for Nurse-like, Cell Survival in Chronic Lymphocytic Leukemia. Clin. Cancer Res, vol.22, pp.6118-6128, 2016.

M. Burger, Small peptide inhibitors of the CXCR4 chemokine receptor (CD184) antagonize the activation, migration, and antiapoptotic responses of CXCL12 in chronic lymphocytic leukemia B cells, Blood, vol.106, pp.1824-1830, 2005.

M. Nishio, Nurselike cells express BAFF and APRIL, which can promote survival of chronic lymphocytic leukemia cells via a paracrine pathway distinct from that of SDF-1?, Blood, vol.106, pp.1012-1020, 2005.

J. A. Burger, High-level expression of the T-cell chemokines CCL3 and CCL4 by chronic lymphocytic leukemia B cells in nurselike cell cocultures and after BCR stimulation, Blood, vol.113, pp.3050-3058, 2009.

F. Boissard, Nurse-like cells promote CLL survival through LFA-3/CD2 interactions, Oncotarget, vol.8, pp.52225-52236, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02348622

S. Saada, Differential Expression of Neurotensin and Specific Receptors, NTSR1 and NTSR2, in Normal and Malignant Human B Lymphocytes, The Journal of Immunology, vol.189, pp.5293-5303, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01819265

L. Dubanet, Anti-apoptotic role and clinical relevance of neurotrophins in diffuse large B-cell lymphomas, Br. J. Cancer, vol.113, pp.934-944, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01500853

M. Tosolini, Human Monocyte Recognition of Adenosine-Based Cyclic Dinucleotides Unveils the A2a G?s Protein-Coupled Receptor Tonic Inhibition of Mitochondrially Induced Cell Death, Mol Cell Biol, vol.35, pp.479-495, 2015.

A. Fauchais, Role of Endogenous Brain-Derived Neurotrophic Factor and Sortilin in B Cell Survival, The Journal of Immunology, vol.181, pp.3027-3038, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00453223

A. Nykjaer and T. E. Willnow, Sortilin: a receptor to regulate neuronal viability and function, Trends Neurosci, vol.35, pp.261-270, 2012.

P. G. Longo, The Akt/Mcl-1 pathway plays a prominent role in mediating antiapoptotic signals downstream of the B-cell receptor in chronic lymphocytic leukemia B cells, Blood, vol.111, pp.846-855, 2008.

C. C. Licence and . By-nc-nd, , p.24

P. G. Longo, The Akt signaling pathway determines the different proliferative capacity of chronic lymphocytic leukemia B-cells from patients with progressive and stable disease, Leukemia, vol.21, pp.110-120, 2007.

S. Kitada, Expression of apoptosis-regulating proteins in chronic lymphocytic leukemia: correlations with In vitro and In vivo chemoresponses, Blood, vol.91, pp.3379-3389, 1998.

J. A. Woyach, A. J. Johnson, and J. C. Byrd, The B-cell receptor signaling pathway as a therapeutic target in CLL, Blood, vol.120, pp.1175-1184, 2012.

J. A. Woyach, Resistance Mechanisms for the Bruton's Tyrosine Kinase Inhibitor Ibrutinib, New England Journal of Medicine, vol.370, pp.2286-2294, 2014.

J. A. Woyach, BTKC481S-Mediated Resistance to Ibrutinib in Chronic Lymphocytic Leukemia, J. Clin. Oncol, vol.35, pp.1437-1443, 2017.

F. Arruga and S. Deaglio, Mechanisms of Resistance to Targeted Therapies in Chronic Lymphocytic Leukemia, Handb Exp Pharmacol, 2017.

I. E. Ahn, Clonal evolution leading to ibrutinib resistance in chronic lymphocytic leukemia, Blood, vol.129, pp.1469-1479, 2017.

M. Y. Choi, M. K. Kashyap, and D. Kumar, The chronic lymphocytic leukemia microenvironment: Beyond the B-cell receptor, Best Pract Res Clin Haematol, vol.29, pp.40-53, 2016.

J. A. Burger, M. Burger, and T. J. Kipps, Chronic lymphocytic leukemia B cells express functional CXCR4 chemokine receptors that mediate spontaneous migration beneath bone marrow stromal cells, Blood, vol.94, pp.3658-3667, 1999.

B. T. Messmer, In vivo measurements document the dynamic cellular kinetics of chronic lymphocytic leukemia B cells, J. Clin. Invest, vol.115, pp.755-764, 2005.

M. Nishio, Nurselike cells express BAFF and APRIL, which can promote survival of chronic lymphocytic leukemia cells via a paracrine pathway distinct from that of SDF-1?, Blood, vol.106, pp.1012-1020, 2005.

D. O. Croci, Nurse-like cells control the activity of chronic lymphocytic leukemia B cells via galectin-1, Leukemia, vol.27, pp.1413-1416, 2013.

Y. Herishanu, The lymph node microenvironment promotes B-cell receptor signaling, NF-kappaB activation, and tumor proliferation in chronic lymphocytic leukemia, Blood, vol.117, pp.563-574, 2011.

F. Cattaneo, Cell-surface receptors transactivation mediated by g protein-coupled receptors, Int J Mol Sci, vol.15, pp.19700-19728, 2014.

N. Delcourt, J. Bockaert, and P. Marin, GPCR-jacking: from a new route in RTK signalling to a new concept in GPCR activation, Trends Pharmacol. Sci, vol.28, pp.602-607, 2007.

C. B. Vaegter, Sortilin associates with Trk receptors to enhance anterograde transport and neurotrophin signaling, Nat. Neurosci, vol.14, pp.54-61, 2011.

B. A. Teicher, S. P. Fricker, and . Cxcl12, SDF-1)/CXCR4 Pathway in Cancer, Clin Cancer Res, vol.16, pp.2927-2931, 2010.

E. Joo, Cell Survival by Stromal Cell-Derived Factor

, /CXCL12 Involves Activation of CREB and Induction of Mcl-1 and c-Fos in Factor-Dependent Human Cell Line MO7e, Stem Cells and Development, vol.13, pp.563-570, 2004.

M. Cojoc, Emerging targets in cancer management: role of the CXCL12/CXCR4 axis, Onco Targets Ther, vol.6, pp.1347-1361, 2013.

G. P. Zapata and J. , Mouse Models of Chronic Lymphocytic Leukemia

, Chronic Lymphocytic Leukemia, 2012.

J. Wang, The Antiapoptotic Gene mcl-1 Is Up-Regulated by the Phosphatidylinositol 3-Kinase/Akt Signaling Pathway through a Transcription Factor Complex Containing CREB, Molecular and Cellular Biology, vol.19, pp.6195-6206, 1999.

, Licence CC BY

D. R. Boggs, S. C. Chen, Z. N. Zhang, and A. Zhang, Chronic lymphocytic leukemia in China, Am. J. Hematol, vol.25, pp.349-354, 1987.

F. Boissard, J. Fournié, A. Quillet-mary, L. Ysebaert, and M. Poupot, Nurselike cells mediate ibrutinib resistance in chronic lymphocytic leukemia patients, Blood Cancer Journal, vol.5, pp.355-355, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02348685

F. Boissard, J. Fournié, C. Laurent, M. Poupot, Y. et al., Nurse like cells: chronic lymphocytic leukemia associated macrophages, Leukemia & Lymphoma, vol.56, pp.1570-1572, 2015.

F. Boissard, M. Tosolini, L. Ligat, A. Quillet-mary, F. Lopez et al., Nurse-like cells promote CLL survival through LFA-3/CD2 interactions, Oncotarget, vol.8, pp.52225-52236, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02348622

T. P. Bonnert, K. E. Garka, P. Parnet, G. Sonoda, J. R. Testa et al., The cloning and characterization of human MyD88: a member of an IL-1 receptor related family, 1997.

, FEBS Letters, vol.402, pp.81-84

A. Bordron, C. Bagacean, A. Mohr, A. Tempescul, B. Bendaoud et al., Resistance to complement activation, cell membrane hypersialylation and relapses in chronic lymphocytic leukemia patients treated with rituximab and chemotherapy, Oncotarget, vol.9, pp.31590-31605, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01854920

N. Bossard, M. Velten, L. Remontet, A. Belot, N. Maarouf et al., Survival of cancer patients in France: a population-based study from The Association of the French Cancer Registries (FRANCIM), 2007.
URL : https://hal.archives-ouvertes.fr/inserm-00346768

, Eur. J. Cancer, vol.43, pp.149-160

J. M. Botto, J. Chabry, P. Sarret, J. P. Vincent, and J. Mazella, Stable expression of the mouse levocabastine-sensitive neurotensin receptor in HEK 293 cell line: binding properties, photoaffinity labeling, and internalization mechanism, Biochem. Biophys. Res. Commun, vol.243, pp.585-590, 1998.

M. Boules, Z. Li, K. Smith, P. Fredrickson, R. et al., Diverse roles of neurotensin agonists in the central nervous system, Front Endocrinol (Lausanne), vol.4, p.36, 2013.

L. A. Boven, J. Middel, P. Portegies, J. Verhoef, G. H. Jansen et al., , 1999.

, Overexpression of nerve growth factor and basic fibroblast growth factor in AIDS dementia complex, J. Neuroimmunol, vol.97, pp.154-162

M. Bozzola, A. N. Thome, E. Giraldi, A. M. Lhiaubet, and R. M. Schimpff, Plasma neurotensin levels in prepubertal children and adults: possible involvement in the regulation of growth hormone secretion, J. Pediatr. Endocrinol. Metab, vol.11, pp.615-621, 1998.

L. Breuza, M. Garcia, M. Delgrossi, L. Bivic, and A. , Role of the membraneproximal O-glycosylation site in sorting of the human receptor for neurotrophins to the apical membrane of MDCK cells, Exp. Cell Res, vol.273, pp.178-186, 2002.
URL : https://hal.archives-ouvertes.fr/hal-00306518

G. M. Brodeur, J. E. Minturn, R. Ho, A. M. Simpson, R. Iyer et al., Trk receptor expression and inhibition in neuroblastomas, Clin. Cancer Res, vol.15, pp.3244-3250, 2009.

G. M. Brodeur, J. E. Minturn, R. Ho, A. M. Simpson, R. Iyer et al.,

V. Kolla and A. E. Evans, Trk receptor expression and inhibition in neuroblastomas, Clin. Cancer Res, vol.15, pp.3244-3250, 2009.

C. Brodie and E. W. Gelfand, Functional nerve growth factor receptors on human B lymphocytes. Interaction with IL-2, The Journal of Immunology, vol.148, pp.3492-3497, 1992.

C. Brodie, A. Oshiba, H. Renz, K. Bradley, and E. W. Gelfand, Nerve growth-factor and anti-CD40 provide opposite signals for the production of IgE in interleukin-4-treated lymphocytes, Eur. J. Immunol, vol.26, pp.171-178, 1996.

J. R. Brown, Inherited susceptibility to chronic lymphocytic leukemia: evidence and prospects for the future, Ther Adv Hematol, vol.4, pp.298-308, 2013.

J. R. Brown, D. Neuberg, K. Phillips, H. Reynolds, J. Silverstein et al., Prevalence of familial malignancy in a prospectively screened cohort of patients with lymphoproliferative disorders, Br. J. Haematol, vol.143, pp.361-368, 2008.

J. R. Brown, J. C. Byrd, S. E. Coutre, D. M. Benson, I. W. Flinn et al., Idelalisib, an inhibitor of phosphatidylinositol 3-kinase p110?, for relapsed/refractory chronic lymphocytic leukemia, Blood, vol.123, pp.3390-3397, 2014.

L. M. Brown, A. Blair, R. Gibson, G. D. Everett, K. P. Cantor et al., Pesticide exposures and other agricultural risk factors for leukemia among men in Iowa and Minnesota, Cancer Res, vol.50, pp.6585-6591, 1990.

M. Bruchard, G. Mignot, V. Derangère, F. Chalmin, A. Chevriaux et al., Chemotherapy-triggered cathepsin B release in myeloid-derived suppressor cells activates the Nlrp3 inflammasome and promotes tumor growth, Nat. Med, vol.19, pp.57-64, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00919371

A. Brunet, A. Bonni, M. J. Zigmond, M. Z. Lin, P. Juo et al., Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor, Cell, vol.96, pp.857-868, 1999.

C. Buechele, T. Baessler, B. J. Schmiedel, C. E. Schumacher, L. Grosse-hovest et al., 4-1BB ligand modulates direct and Rituximab-induced NK-cell reactivity in chronic lymphocytic leukemia, European Journal of Immunology, vol.42, pp.737-748, 2012.

A. G. Buggins and C. J. Pepper, The role of Bcl-2 family proteins in chronic lymphocytic leukaemia, Leuk. Res, vol.34, pp.837-842, 2010.

J. A. Burger, Nurture versus Nature: The Microenvironment in Chronic Lymphocytic Leukemia, Hematology, pp.96-103, 2011.

J. A. Burger, The CLL Cell Microenvironment, Advances in Chronic Lymphocytic Leukemia, pp.25-45, 2013.

J. A. Burger and J. J. Buggy, Bruton tyrosine kinase inhibitor ibrutinib, p.32765, 2013.

, Leuk. Lymphoma, vol.54, pp.2385-2391

J. A. Burger and N. Chiorazzi, B cell receptor signaling in chronic lymphocytic leukemia, Trends in Immunology, vol.34, pp.592-601, 2013.

, Licence CC BY

J. A. Burger and S. Brien, Evolution of CLL treatment -from chemoimmunotherapy to targeted and individualized therapy, Nat Rev Clin Oncol, vol.15, pp.510-527, 2018.

J. A. Burger, M. Burger, and T. J. Kipps, Chronic lymphocytic leukemia B cells express functional CXCR4 chemokine receptors that mediate spontaneous migration beneath bone marrow stromal cells, Blood, vol.94, pp.3658-3667, 1999.

J. A. Burger, N. Tsukada, M. Burger, N. J. Zvaifler, M. Dell'aquila et al., , 2000.

, Blood-derived nurse-like cells protect chronic lymphocytic leukemia B cells from spontaneous apoptosis through stromal cell-derived factor-1, Blood, vol.96, pp.2655-2663

J. A. Burger, M. P. Quiroga, E. Hartmann, A. Bürkle, W. G. Wierda et al., High-level expression of the T-cell chemokines CCL3 and CCL4 by chronic lymphocytic leukemia B cells in nurselike cell cocultures and after BCR stimulation, Blood, vol.113, pp.3050-3058, 2009.

J. A. Burger, A. Tedeschi, P. M. Barr, T. Robak, C. Owen et al., Ibrutinib as Initial Therapy for Patients with Chronic Lymphocytic Leukemia, New England Journal of Medicine, vol.373, pp.2425-2437, 2015.

J. A. Burger, D. A. Landau, A. Taylor-weiner, I. Bozic, H. Zhang et al., Clonal evolution in patients with chronic lymphocytic leukaemia developing resistance to BTK inhibition, Nat Commun, vol.7, p.11589, 2016.

J. A. Burger, K. W. Li, M. J. Keating, M. Sivina, A. M. Amer et al., Leukemia cell proliferation and death in chronic lymphocytic leukemia patients on therapy with the BTK inhibitor ibrutinib, JCI Insight, vol.2, 2017.

A. Bürkle, M. Niedermeier, A. Schmitt-gräff, W. G. Wierda, M. J. Keating et al., Overexpression of the CXCR5 chemokine receptor, and its ligand, CXCL13 in B-cell chronic lymphocytic leukemia, Blood, vol.110, pp.3316-3325, 2007.

J. D. Burton, C. H. Weitz, and N. E. Kay, Malignant chronic lymphocytic leukemia B cells elaborate soluble factors that down-regulate T cell and NK function, American Journal of Hematology, vol.30, pp.61-67, 1989.

J. C. Byrd, R. R. Furman, S. E. Coutre, I. W. Flinn, J. A. Burger et al., Targeting BTK with Ibrutinib in Relapsed Chronic Lymphocytic Leukemia, 2013.

R. S. Bystry, V. Aluvihare, K. A. Welch, M. Kallikourdis, and A. G. Betz, B cells and professional APCs recruit regulatory T cells via CCL4, Nat. Immunol, vol.2, pp.1126-1132, 2001.

N. Cahill and R. Rosenquist, Uncovering the DNA methylome in chronic lymphocytic leukemia, Epigenetics, vol.8, pp.138-148, 2013.

F. Caligaris-cappio, Role of the microenvironment in chronic lymphocytic leukaemia, Br. J. Haematol, vol.123, pp.380-388, 2003.

F. Caligaris-cappio, Chronic Lymphocytic Leukemia: "Cinderella" Is Becoming a Star, Mol Med, vol.15, pp.67-69, 2009.

G. A. Calin, C. D. Dumitru, M. Shimizu, R. Bichi, S. Zupo et al.,

M. Keating and K. Rai, Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia, PNAS, vol.99, pp.15524-15529, 2002.

G. A. Calin, M. Ferracin, A. Cimmino, G. Di-leva, M. Shimizu et al., A MicroRNA signature associated with prognosis and progression in chronic lymphocytic leukemia, N. Engl. J. Med, vol.353, pp.1793-1801, 2005.

C. Calissano, R. N. Damle, G. Hayes, E. J. Murphy, M. K. Hellerstein et al., In vivo intraclonal and interclonal kinetic heterogeneity in B-cell chronic lymphocytic leukemia, Blood, vol.114, pp.4832-4842, 2009.

C. Calissano, R. N. Damle, X. J. Yan, W. Li, S. Marsilio et al., Multi-Parameter Phenotypic Analysis of Members of Chronic Lymphocytic Leukemia Clones Identifies Distinct Proliferative and Resting/Re-Entry Compartments with Discrete Gene Expression Profiles, Blood, vol.114, pp.668-668, 2009.

C. Calissano, R. N. Damle, S. Marsilio, X. Yan, S. Yancopoulos et al., Intraclonal complexity in chronic lymphocytic leukemia: fractions enriched in recently born/divided and older/quiescent cells, Mol. Med, vol.17, pp.1374-1382, 2011.

P. V. Campregher and N. Hamerschlak, Novel Prognostic Gene Mutations Identified in Chronic Lymphocytic Leukemia and Their Impact on Clinical Practice, Clinical Lymphoma, Myeloma and Leukemia, vol.14, pp.271-276, 2014.

S. Capalbo, P. Trerotoli, A. Ciancio, C. Battista, G. Serio et al., Increased risk of lymphoproliferative disorders in relatives of patients with B-cell chronic lymphocytic leukemia: relevance of the degree of familial linkage, Eur. J. Haematol, vol.65, pp.114-117, 2000.

P. Carmeliet and R. K. Jain, Molecular mechanisms and clinical applications of angiogenesis, Nature, vol.473, pp.298-307, 2011.

R. Carraway and S. E. Leeman, The isolation of a new hypotensive peptide, neurotensin, from bovine hypothalami, J. Biol. Chem, vol.248, pp.6854-6861, 1973.

R. Carraway and S. E. Leeman, The amino acid sequence of a hypothalamic peptide, neurotensin, J. Biol. Chem, vol.250, pp.1907-1911, 1975.

R. Carraway and S. E. Leeman, Characterization of radioimmunoassayable neurotensin in the rat. Its differential distribution in the central nervous system, small intestine, and stomach, J. Biol. Chem, vol.251, pp.7045-7052, 1976.

R. E. Carraway and S. P. Mitra, Binding and biologic activity of neurotensin in guinea pig ileum, Peptides, vol.15, pp.1451-1459, 1994.

R. E. Carraway and A. M. Plona, Involvement of neurotensin in cancer growth: Evidence, mechanisms and development of diagnostic tools, Peptides, vol.27, pp.2445-2460, 2006.

D. Cassiman, C. Denef, V. J. Desmet, and T. Roskams, Human and rat hepatic stellate cells express neurotrophins and neurotrophin receptors, Hepatology, vol.33, pp.148-158, 2001.

I. Castagliuolo, C. C. Wang, L. Valenick, A. Pasha, S. Nikulasson et al., Neurotensin is a proinflammatory neuropeptide in colonic Licence CC BY-NC-ND 3.0 inflammation, J. Clin. Invest, vol.103, pp.843-849, 1999.

R. A. Castillo-rodríguez, M. L. Arango-rodríguez, L. Escobedo, D. Hernandez-baltazar, A. Gompel et al., Suicide HSVtk Gene Delivery by Neurotensin-Polyplex Nanoparticles via the Bloodstream and GCV Treatment Specifically Inhibit the Growth of Human MDA-MB-231 Triple Negative Breast Cancer Tumors Xenografted in Athymic Mice, PLoS One, vol.9, 2014.

R. Castriconi, C. Cantoni, M. Della-chiesa, M. Vitale, E. Marcenaro et al., Transforming growth factor beta 1 inhibits expression of NKp30 and NKG2D receptors: consequences for the NK-mediated killing of dendritic cells, Proc. Natl. Acad. Sci. U.S.A, vol.100, pp.4120-4125, 2003.

D. Catovsky, J. Fooks, and S. Richards, Prognostic factors in chronic lymphocytic leukaemia: the importance of age, sex and response to treatment in survival. A report from the MRC CLL 1 trial. MRC Working Party on Leukaemia in Adults, Br. J. Haematol, vol.72, pp.141-149, 1989.

F. Cattaneo, G. Guerra, M. Parisi, M. De-marinis, D. Tafuri et al., Cell-surface receptors transactivation mediated by g protein-coupled receptors, Int J Mol Sci, vol.15, pp.19700-19728, 2014.

F. Cattaneo, G. Guerra, M. Parisi, M. De-marinis, D. Tafuri et al., Cell-Surface Receptors Transactivation Mediated by G Protein-Coupled Receptors, Int J Mol Sci, vol.15, pp.19700-19728, 2014.

C. Caux, R. N. Ramos, G. C. Prendergast, N. Bendriss-vermare, and C. Ménétrier-caux,

, A Milestone Review on How Macrophages Affect Tumor Growth, Cancer Res, vol.76, pp.6439-6442

R. Chakravarthy, K. Mnich, and A. M. Gorman, Nerve growth factor (NGF)-mediated regulation of p75(NTR) expression contributes to chemotherapeutic resistance in triple negative breast cancer cells, Biochem. Biophys. Res. Commun, vol.478, pp.1541-1547, 2016.

A. Chanan-khan and C. W. Porter, Immunomodulating drugs for chronic lymphocytic leukaemia, The Lancet Oncology, vol.7, pp.480-488, 2006.

W. Chanput, J. J. Mes, and H. J. Wichers, THP-1 cell line: An in vitro cell model for immune modulation approach, International Immunopharmacology, vol.23, pp.37-45, 2014.

S. Chantepie, S. Krieger, and M. Leporrier, Méthylation dans les hémopathies malignes : techniques et perspectives. Hématologie, vol.18, pp.124-132, 2012.

M. V. Chao, Neurotrophins and their receptors: a convergence point for many signalling pathways, Nat. Rev. Neurosci, vol.4, pp.299-309, 2003.

M. V. Chao and M. Bothwell, Neurotrophins: to cleave or not to cleave, Neuron, vol.33, pp.9-12, 2002.

E. Chapiro, I. Radford-weiss, C. Bastard, I. Luquet, C. Lefebvre et al., The most frequent t(14;19)(q32;q13)-positive B-cell malignancy corresponds to an aggressive subgroup of atypical chronic lymphocytic leukemia, Leukemia, vol.22, pp.2123-2127, 2008.

E. Chapiro, N. Leporrier, I. Radford-weiss, C. Bastard, H. Mossafa et al.,

I. De-braekeleer, M. Terré, C. Brizard, and F. , Gain of the short arm of chromosome 2 (2p) is a frequent recurring chromosome aberration in untreated chronic lymphocytic leukemia (CLL) at advanced stages, Leuk. Res, vol.34, pp.63-68, 2010.

F. Checler, J. P. Vincent, and P. Kitabgi, Purification and characterization of a novel neurotensin-degrading peptidase from rat brain synaptic membranes, J. Biol. Chem, vol.261, pp.11274-11281, 1986.

J. Chen, J. Chen, and R. C. Harris, Angiotensin II induces epithelial-tomesenchymal transition in renal epithelial cells through reactive oxygen species/Src/caveolinmediated activation of an epidermal growth factor receptor-extracellular signal-regulated kinase signaling pathway, Mol. Cell. Biol, vol.32, pp.981-991, 2012.

L. Chen, G. Widhopf, L. Huynh, L. Rassenti, K. R. Rai et al., Expression of ZAP-70 is associated with increased B-cell receptor signaling in chronic lymphocytic leukemia, Blood, vol.100, pp.4609-4614, 2002.

L. Chen, L. Huynh, J. Apgar, L. Tang, L. Rassenti et al., , 2008.

, ZAP-70 enhances IgM signaling independent of its kinase activity in chronic lymphocytic leukemia, Blood, vol.111, pp.2685-2692

Z. Chen, A. Ieraci, H. Teng, H. Dall, C. Meng et al., Sortilin controls intracellular sorting of brain-derived neurotrophic factor to the regulated secretory pathway, J. Neurosci, vol.25, pp.6156-6166, 2005.

S. Chiaretti, M. Marinelli, I. D. Giudice, S. Bonina, A. Piciocchi et al., NOTCH1, SF3B1, BIRC3 and TP53 mutations in patients with chronic lymphocytic leukemia undergoing first-line treatment: correlation with biological parameters and response to treatment, Leukemia & Lymphoma, vol.55, pp.2785-2792, 2014.

N. Chiorazzi and M. Ferrarini, B Cell Chronic Lymphocytic Leukemia: Lessons Learned from Studies of the B Cell Antigen Receptor, Annu. Rev. Immunol, vol.21, pp.841-894, 2003.

N. Chiorazzi and M. Ferrarini, Cellular origin(s) of chronic lymphocytic leukemia: cautionary notes and additional considerations and possibilities, Blood, vol.117, pp.1781-1791, 2011.

N. Chiorazzi, K. R. Rai, and M. Ferrarini, Chronic lymphocytic leukemia, N. Engl. J. Med, vol.352, pp.804-815, 2005.
URL : https://hal.archives-ouvertes.fr/hal-01375537

J. E. Chipuk, T. Kuwana, L. Bouchier-hayes, N. M. Droin, D. D. Newmeyer et al., Direct activation of Bax by p53 mediates mitochondrial membrane permeabilization and apoptosis, Science, vol.303, pp.1010-1014, 2004.

M. Y. Choi, M. K. Kashyap, and D. Kumar, The chronic lymphocytic leukemia microenvironment: Beyond the B-cell receptor, Best Pract Res Clin Haematol, vol.29, pp.40-53, 2016.

A. Cimmino, G. A. Calin, M. Fabbri, M. V. Iorio, M. Ferracin et al., miR-15 and miR-16 induce apoptosis by targeting BCL2, Proc. Natl. Acad. Sci. U.S.A, vol.102, pp.13944-13949, 2005.

C. A. Clarke, S. L. Glaser, S. L. Gomez, S. S. Wang, T. H. Keegan et al., Lymphoid malignancies in US Asians: incidence rate differences by birthplace and acculturation, Cancer Epidemiol Biomarkers Prev, vol.20, pp.1064-1077, 2011.

, Licence CC BY

D. E. Cochrane, R. E. Carraway, K. Harrington, M. Laudano, S. Rawlings et al., HMC-1 human mast cells synthesize neurotensin (NT) precursor, secrete bioactive NT-like peptide(s) and express NT receptor NTS1, Inflamm. Res, vol.60, pp.1139-1151, 2011.

V. Coelho, S. Krysov, A. Steele, M. Sanchez-hidalgo, P. W. Johnson et al., Identification in CLL of circulating intraclonal subgroups with varying B-cell receptor expression and function, Blood, vol.122, pp.2664-2672, 2013.

S. B. Coffelt, R. Hughes, and C. E. Lewis, Tumor-associated macrophages: effectors of angiogenesis and tumor progression, Biochim. Biophys. Acta, vol.1796, pp.11-18, 2009.

S. B. Coffelt, A. O. Tal, A. Scholz, M. De-palma, S. Patel et al., Angiopoietin-2 regulates gene expression in TIE2-expressing monocytes and augments their inherent proangiogenic functions, Cancer Res, vol.70, pp.5270-5280, 2010.

S. Cohen, R. Levi-montalcini, and V. Hamburger, A NERVE GROWTH-STIMULATING FACTOR ISOLATED FROM SARCOM AS 37 AND 180*, Proc Natl Acad Sci U S A, vol.40, pp.1014-1018, 1954.

M. Cojoc, C. Peitzsch, F. Trautmann, L. Polishchuk, G. D. Telegeev et al., Emerging targets in cancer management: role of the CXCL12/CXCR4 axis, Onco Targets Ther, vol.6, pp.1347-1361, 2013.

R. J. Collins, L. A. Verschuer, B. V. Harmon, R. L. Prentice, J. H. Pope et al., Spontaneous programmed death (apoptosis) of B-chronic lymphocytic leukaemia cells following their culture in vitro, Br. J. Haematol, vol.71, pp.343-350, 1989.

F. Colotta, P. Allavena, A. Sica, C. Garlanda, and A. Mantovani, Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability, Carcinogenesis, vol.30, pp.1073-1081, 2009.

M. Cols, C. M. Barra, B. He, I. Puga, W. Xu et al., Stromal endothelial cells establish a bidirectional crosstalk with chronic lymphocytic leukemia cells through the TNF-related factors BAFF, APRIL, and CD40L, J. Immunol, vol.188, pp.6071-6083, 2012.

A. Contri, A. M. Brunati, L. Trentin, A. Cabrelle, M. Miorin et al., Chronic lymphocytic leukemia B cells contain anomalous Lyn tyrosine kinase, a putative contribution to defective apoptosis, J Clin Invest, vol.115, pp.369-378, 2005.

P. E. Cooper, M. H. Fernstrom, O. P. Rorstad, S. E. Leeman, and J. B. Martin, The regional distribution of somatostatin, substance P and neurotensin in human brain, Brain Res, vol.218, pp.219-232, 1981.

K. C. Corbit, D. A. Foster, and M. R. Rosner, Protein kinase Cdelta mediates neurogenic but not mitogenic activation of mitogen-activated protein kinase in neuronal cells, Mol. Cell. Biol, vol.19, pp.4209-4218, 1999.

A. Cosson, E. Chapiro, N. Bougacha, J. Lambert, L. Herbi et al., Gain in the short arm of chromosome 2 (2p+) induces gene overexpression and drug resistance in chronic lymphocytic leukemia: analysis of Licence CC BY-NC-ND 3.0 the central role of XPO1, Leukemia, vol.31, pp.1625-1629, 2017.

P. Cramer and M. Hallek, Prognostic factors in chronic lymphocytic leukemia-what do we need to know?, Nat Rev Clin Oncol, vol.8, pp.38-47, 2011.

P. Cramer, P. Langerbeins, B. Eichhorst, and M. Hallek, Advances in first-line treatment of chronic lymphocytic leukemia: current recommendations on management and first-line treatment by the German CLL Study Group (GCLLSG), Eur. J. Haematol, vol.96, pp.9-18, 2016.

M. Crespo, F. Bosch, N. Villamor, B. Bellosillo, D. Colomer et al., ZAP-70 expression as a surrogate for immunoglobulin-variable-region mutations in chronic lymphocytic leukemia, N. Engl. J. Med, vol.348, pp.1764-1775, 2003.

D. O. Croci, P. E. Morande, S. Dergan-dylon, M. Borge, M. A. Toscano et al., Nurse-like cells control the activity of chronic lymphocytic leukemia B cells via galectin-1, Leukemia, vol.27, pp.1413-1416, 2013.

J. L. Croucher, R. Iyer, N. Li, V. Molteni, J. Loren et al., TrkB Inhibition by GNF-4256 Slows Growth and Enhances Chemotherapeutic Efficacy in Neuroblastoma Xenografts, Cancer Chemother Pharmacol, vol.75, pp.131-141, 2015.

D. Crowther-swanepoel, P. Broderick, M. C. Di-bernardo, S. E. Dobbins, M. Torres et al., Common variants at 2q37.3, 8q24.21, 15q21.3 and 16q24.1 influence chronic lymphocytic leukemia risk, Nat. Genet, vol.42, pp.132-136, 2010.

J. M. Dal-porto, S. B. Gauld, K. T. Merrell, D. Mills, A. E. Pugh-bernard et al.,

, B cell antigen receptor signaling 101, Mol. Immunol, vol.41, pp.599-613

R. N. Damle, T. Wasil, F. Fais, F. Ghiotto, A. Valetto et al., Ig V gene mutation status and CD38 expression as novel prognostic indicators in chronic lymphocytic leukemia, Blood, vol.94, pp.1840-1847, 1999.

R. N. Damle, S. Temburni, C. Calissano, S. Yancopoulos, T. Banapour et al., CD38 expression labels an activated subset within chronic lymphocytic leukemia clones enriched in proliferating B cells, Blood, vol.110, pp.3352-3359, 2007.

G. D'arena and P. Musto, Monoclonal B-Cell Lymphocytosis, Transl Med UniSa, vol.8, pp.75-79, 2014.

G. D'arena, P. Musto, N. Cascavilla, M. Dell'olio, N. Renzo et al., CD38 Expression Correlates with Adverse Biological Features and Predicts Poor Clinical Outcome in B-Cell Chronic Lymphocytic Leukemia, Leukemia & Lymphoma, vol.42, pp.109-114, 2001.

S. R. Datta, H. Dudek, X. Tao, S. Masters, H. Fu et al., , 1997.

, Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery, Cell, vol.91, pp.231-241

M. J. Davis, T. M. Tsang, Y. Qiu, J. K. Dayrit, J. B. Freij et al.,

M. A. , Macrophage M1/M2 polarization dynamically adapts to changes in cytokine microenvironments in Cryptococcus neoformans infection, MBio, vol.4, pp.264-00213, 2013.

M. De-la-fuente, J. J. Garrido, R. M. Arahuetes, and A. Hernanz, Stimulation of phagocytic function in mouse macrophages by neurotensin and neuromedin N, J. Neuroimmunol, vol.42, pp.97-104, 1993.

S. Deaglio, T. Vaisitti, L. Bergui, L. Bonello, A. L. Horenstein et al., CD38 and CD100 lead a network of surface receptors relaying positive signals for B-CLL growth and survival, Blood, vol.105, pp.3042-3050, 2005.

S. Deaglio, T. Vaisitti, S. Aydin, L. Bergui, G. D'arena et al., CD38 and ZAP-70 are functionally linked and mark CLL cells with high migratory potential, Blood, vol.110, pp.4012-4021, 2007.

G. Dechant, Molecular interactions between neurotrophin receptors, Cell Tissue Res, vol.305, pp.229-238, 2001.

J. Defoiche, C. Debacq, B. Asquith, Y. Zhang, A. Burny et al., Reduction of B cell turnover in chronic lymphocytic leukaemia, Br. J. Haematol, vol.143, pp.240-247, 2008.

G. Del-poeta, M. Bo, M. I. Del-principe, F. Pozzo, F. M. Rossi et al., Clinical significance of c.7544-7545 delCT NOTCH1 mutation in chronic lymphocytic leukaemia, Br. J. Haematol, vol.160, pp.415-418, 2013.

J. Delgado, G. Pratt, N. Phillips, J. Briones, C. Fegan et al., Beta2-microglobulin is a better predictor of treatmentfree survival in patients with chronic lymphocytic leukaemia if adjusted according to glomerular filtration rate, Br. J. Haematol, vol.145, pp.801-805, 2009.

I. E. Demir, E. Tieftrunk, S. Schorn, H. Friess, and G. O. Ceyhan, Nerve growth factor & TrkA as novel therapeutic targets in cancer, Biochim. Biophys. Acta, vol.1866, pp.37-50, 2016.

Y. Demont, C. Corbet, A. Page, Y. Ataman-Önal, G. Choquet-kastylevsky et al., Pro-nerve growth factor induces autocrine stimulation of breast cancer cell invasion through tropomyosinrelated kinase A (TrkA) and sortilin protein, J. Biol. Chem, vol.287, pp.1923-1931, 2012.

X. Deng, F. Gao, T. Flagg, J. Anderson, and W. S. May, Bcl2's flexible loop domain regulates p53 binding and survival, Mol. Cell. Biol, vol.26, pp.4421-4434, 2006.

S. Derenne, B. Monia, N. M. Dean, J. K. Taylor, M. Rapp et al., Antisense strategy shows that Mcl-1 rather than Bcl-2 or Bcl-x(L) is an essential survival protein of human myeloma cells, Blood, vol.100, pp.194-199, 2002.

F. Di-raimondo, R. Giustolisi, S. Lerner, E. Cacciola, S. O'brien et al., Retrospective study of the prognostic role of serum thymidine kinase level in CLL patients with active disease treated with fludarabine, Ann. Oncol, vol.12, pp.621-625, 2001.

E. Dicou, J. Vincent, and J. Mazella, Neurotensin receptor-3/sortilin mediates neurotensin-induced cytokine/chemokine expression in a murine microglial cell line, J. Neurosci. Res, vol.78, pp.92-99, 2004.

G. Dighiero and T. J. Hamblin, Chronic lymphocytic leukaemia, Lancet, vol.371, 2008.
URL : https://hal.archives-ouvertes.fr/pasteur-00838819

W. Ding, T. R. Knox, R. C. Tschumper, W. Wu, S. M. Schwager et al., Platelet-derived growth factor (PDGF)-PDGF receptor interaction activates bone marrow-derived mesenchymal stromal cells derived from chronic lymphocytic leukemia: implications for an angiogenic switch, Blood, vol.116, pp.2984-2993, 2010.

W. Ding, B. R. Laplant, T. G. Call, S. A. Parikh, J. F. Leis et al., Pembrolizumab in patients with CLL and Richter transformation or with relapsed CLL, Blood, vol.129, pp.3419-3427, 2017.

P. R. Dobner, Multitasking with neurotensin in the central nervous system, Cell. Mol. Life Sci, vol.62, pp.1946-1963, 2005.

P. R. Dobner, Neurotensin and pain modulation, Peptides, vol.27, pp.2405-2414, 2006.

R. C. Doebele, L. E. Davis, A. Vaishnavi, A. T. Le, A. Estrada-bernal et al., An Oncogenic NTRK Fusion in a Patient with Soft-Tissue Sarcoma with Response to the Tropomyosin-Related Kinase Inhibitor LOXO-101, Cancer Discov, vol.5, pp.1049-1057, 2015.

H. Döhner, S. Stilgenbauer, M. R. James, A. Benner, T. Weilguni et al., 11q deletions identify a new subset of B-cell chronic lymphocytic leukemia characterized by extensive nodal involvement and inferior prognosis, Blood, vol.89, pp.2516-2522, 1997.

H. Döhner, S. Stilgenbauer, A. Benner, E. Leupolt, A. Kröber et al., Genomic aberrations and survival in chronic lymphocytic leukemia, N. Engl. J. Med, vol.343, pp.1910-1916, 2000.

R. E. Dolmetsch, R. S. Lewis, C. C. Goodnow, and J. I. Healy, Differential activation of transcription factors induced by Ca2+ response amplitude and duration, Nature, vol.386, pp.855-858, 1997.

Z. Dong, Q. Lei, R. Yang, S. Zhu, X. Ke et al., Inhibition of neurotensin receptor 1 induces intrinsic apoptosis via let-7a-3p/Bcl-w axis in glioblastoma, Br. J. Cancer, vol.116, pp.1572-1584, 2017.

L. Dubanet, H. Bentayeb, B. Petit, A. Olivrie, S. Saada et al., Anti-apoptotic role and clinical relevance of neurotrophins in diffuse large B-cell lymphomas, Br. J. Cancer, vol.113, pp.934-944, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01500853

J. A. Dubovsky, D. L. Chappell, B. K. Harrington, K. Agrawal, L. A. Andritsos et al., Lymphocyte cytosolic protein 1 is a chronic lymphocytic leukemia membrane-associated antigen critical to niche homing, Blood, vol.122, pp.3308-3316, 2013.

I. Dubuc, P. Sarret, C. Labbé-jullié, J. M. Botto, E. Honoré et al., Identification of the receptor subtype involved in the analgesic effect of neurotensin, J. Neurosci, vol.19, pp.503-510, 1999.

D. Duluc, Y. Delneste, F. Tan, M. Moles, L. Grimaud et al., Tumor-associated leukemia inhibitory factor and IL-6, 2007.

, Licence CC BY

M. Erreni, A. Mantovani, A. , and P. , Tumor-associated Macrophages (TAM) and Inflammation in Colorectal Cancer, Cancer Microenviron, vol.4, pp.141-154, 2010.

J. Escamilla, S. Schokrpur, C. Liu, S. J. Priceman, D. Moughon et al., CSF1 receptor targeting in prostate cancer reverses macrophage-mediated resistance to androgen blockade therapy, Cancer Res, vol.75, pp.950-962, 2015.

G. Esposito, C. Perrino, A. Cannavo, G. G. Schiattarella, F. Borgia et al., EGFR trans-activation by urotensin II receptor is mediated by ?-arrestin recruitment and confers cardioprotection in pressure overload-induced cardiac hypertrophy, Basic Res. Cardiol, vol.106, pp.577-589, 2011.

B. M. Evers, R. J. Bold, J. A. Ehrenfried, J. Li, C. M. Townsend et al., Characterization of functional neurotensin receptors on human lymphocytes, Surgery, vol.116, pp.139-140, 1994.

S. Evrard, P. Gaussem, D. Helley, and L. Darnige, Facteurs pronostiques de la leucémie lymphoïde chronique : apport des marqueurs biologiques récents, Ann Biol Clin, vol.63, 2005.

G. Fabbri, S. Rasi, D. Rossi, V. Trifonov, H. Khiabanian et al., Analysis of the chronic lymphocytic leukemia coding genome: role of NOTCH1 mutational activation, Journal of Experimental Medicine, vol.208, pp.1389-1401, 2011.

S. Fabris, L. Mosca, G. Cutrona, M. Lionetti, L. Agnelli et al., Chromosome 2p gain in monoclonal B-cell lymphocytosis and in early stage chronic lymphocytic leukemia, Am. J. Hematol, vol.88, pp.24-31, 2013.

S. Faderl, M. J. Keating, K. Do, S. Liang, H. M. Kantarjian et al., Expression profile of 11 proteins and their prognostic significance in patients with chronic lymphocytic leukemia (CLL), Leukemia, vol.16, pp.1045-1052, 2002.

F. Fais, F. Ghiotto, S. Hashimoto, B. Sellars, A. Valetto et al., Chronic lymphocytic leukemia B cells express restricted sets of mutated and unmutated antigen receptors, J Clin Invest, vol.102, pp.1515-1525, 1998.

C. Falciani, J. Brunetti, C. Pagliuca, S. Menichetti, L. Vitellozzi et al., Design and in vitro evaluation of branched peptide conjugates: turning nonspecific cytotoxic drugs into tumor-selective agents, ChemMedChem, vol.5, pp.567-574, 2010.

C. Falciani, B. Lelli, J. Brunetti, S. Pileri, A. Cappelli et al., Modular branched neurotensin peptides for tumor target tracing and receptor-mediated therapy: a proof-of-concept, Curr Cancer Drug Targets, vol.10, pp.695-704, 2010.

E. Falisi, E. Novella, C. Visco, N. Guercini, F. Maura et al., B-cell receptor configuration and mutational analysis of patients with chronic lymphocytic leukaemia and trisomy 12 reveal recurrent molecular abnormalities, Hematological Oncology, vol.32, pp.22-30, 2014.

, Licence CC BY

G. Gaidano, R. Foà, and R. Dalla-favera, Molecular pathogenesis of chronic lymphocytic leukemia, J. Clin. Invest, vol.122, pp.3432-3438, 2012.

T. F. Gajewski, H. Schreiber, and Y. Fu, Innate and adaptive immune cells in the tumor microenvironment, Nat. Immunol, vol.14, pp.1014-1022, 2013.

C. Galustian, B. Meyer, M. Labarthe, K. Dredge, D. Klaschka et al., The anti-cancer agents lenalidomide and pomalidomide inhibit the proliferation and function of T regulatory cells, Cancer Immunol. Immunother, vol.58, pp.1033-1045, 2009.

E. García-garayoa, P. Bläuenstein, A. Blanc, V. Maes, D. Tourwé et al., A stable neurotensin-based radiopharmaceutical for targeted imaging and therapy of neurotensin receptor-positive tumours, Eur. J. Nucl. Med. Mol. Imaging, vol.36, pp.37-47, 2009.

R. García-muñoz, V. R. Galiacho, and L. Llorente, Immunological aspects in chronic lymphocytic leukemia (CLL) development, Ann. Hematol, vol.91, pp.981-996, 2012.

R. Garg, W. Wierda, A. Ferrajoli, L. Abruzzo, S. Pierce et al., The prognostic difference of monoallelic versus biallelic deletion of 13q in chronic lymphocytic leukemia, Cancer, vol.118, pp.3531-3537, 2012.

L. Gaviglio, A. Gross, N. Metzler-nolte, and M. Ravera, Synthesis and in vitro cytotoxicity of cis,cis,trans-diamminedichloridodisuccinatoplatinum(IV)-peptide bioconjugates, Metallomics, vol.4, pp.260-266, 2012.

L. Gendron, A. Perron, M. D. Payet, N. Gallo-payet, P. Sarret et al., , 2004.

, Low-affinity neurotensin receptor (NTS2) signaling: internalization-dependent activation of extracellular signal-regulated kinases 1/2, Mol. Pharmacol, vol.66, pp.1421-1430

R. Van-gent, A. P. Kater, S. A. Otto, A. Jaspers, J. A. Borghans et al., In vivo dynamics of stable chronic lymphocytic leukemia inversely correlate with somatic hypermutation levels and suggest no major leukemic turnover in bone marrow, Cancer Res, vol.68, pp.10137-10144, 2008.

M. Gentile, G. Cutrona, A. Neri, S. Molica, M. Ferrarini et al., , 2009.

, Predictive value of beta2-microglobulin (beta2-m) levels in chronic lymphocytic leukemia since Binet A stages, Haematologica, vol.94, pp.887-888

D. Gesty-palmer, L. Yuan, B. Martin, W. H. Wood, M. Lee et al., ?-arrestin-selective G protein-coupled receptor agonists engender unique biological efficacy in vivo, Mol. Endocrinol, vol.27, pp.296-314, 2013.

H. Ghamlouch, F. Nguyen-khac, and O. A. Bernard, Chronic lymphocytic leukaemia genomics and the precision medicine era, Br. J. Haematol, vol.178, pp.852-870, 2017.

P. Ghia, L. Granziero, M. Chilosi, and F. Caligaris-cappio, Chronic B cell malignancies and bone marrow microenvironment, Semin. Cancer Biol, vol.12, pp.149-155, 2002.

P. Ghia, G. Strola, L. Granziero, M. Geuna, G. Guida et al., Chronic lymphocytic leukemia B cells are endowed with the capacity to attract CD4+, CD40L+ T cells by producing CCL22, European Journal of Immunology, vol.32, pp.1403-1413, 2002.

P. Ghia, G. Guida, S. Stella, D. Gottardi, M. Geuna et al.,

F. Cappio, The pattern of CD38 expression defines a distinct subset of chronic lymphocytic leukemia (CLL) patients at risk of disease progression, Blood, vol.101, pp.1262-1269, 2003.

P. Ghia, P. Circosta, C. Scielzo, A. Vallario, A. Camporeale et al., Differential effects on CLL cell survival exerted by different microenvironmental elements, Curr. Top. Microbiol. Immunol, vol.294, pp.135-145, 2005.

P. Ghia, A. M. Ferreri, and F. Caligaris-cappio, Chronic lymphocytic leukemia, Crit. Rev. Oncol. Hematol, vol.64, pp.234-246, 2007.
URL : https://hal.archives-ouvertes.fr/hal-02318055

A. K. Ghosh and N. E. Kay, Critical Signal Transduction Pathways in CLL, Adv Exp Med Biol, vol.792, pp.215-239, 2013.

S. Ghosh, M. J. May, and E. B. Kopp, NF-kappa B and Rel proteins: evolutionarily conserved mediators of immune responses, Annu. Rev. Immunol, vol.16, pp.225-260, 1998.

P. Giannoni, S. Scaglione, R. Quarto, R. Narcisi, M. Parodi et al., An interaction between hepatocyte growth factor and its receptor (c-MET) prolongs the survival of chronic lymphocytic leukemic cells through STAT3 phosphorylation: a potential role of mesenchymal cells in the disease, Haematologica, vol.96, pp.1015-1023, 2011.

P. Giannoni, G. Pietra, G. Travaini, R. Quarto, G. Shyti et al., Chronic lymphocytic leukemia nurse-like cells express hepatocyte growth factor receptor (c-MET) and indoleamine 2,3-dioxygenase and display features of immunosuppressive type 2 skewed macrophages, Haematologica, vol.99, pp.1078-1087, 2014.

E. Giraudo, M. Inoue, and D. Hanahan, An amino-bisphosphonate targets MMP-9-expressing macrophages and angiogenesis to impair cervical carcinogenesis, J Clin Invest, vol.114, pp.623-633, 2004.

S. Gobessi, L. Laurenti, P. G. Longo, L. Carsetti, V. Berno et al., Inhibition of constitutive and BCR-induced Syk activation downregulates Mcl-1 and induces apoptosis in chronic lymphocytic leukemia B cells, Leukemia, vol.23, pp.686-697, 2009.

V. Goede, K. Fischer, R. Busch, A. Engelke, B. Eichhorst et al., Obinutuzumab plus chlorambucil in patients with CLL and coexisting conditions, N. Engl. J. Med, vol.370, pp.1101-1110, 2014.

M. R. Gold, M. P. Scheid, L. Santos, M. Dang-lawson, R. A. Roth et al., The B Cell Antigen Receptor Activates the Akt (Protein Kinase B)/Glycogen Synthase Kinase-3 Signaling Pathway Via Phosphatidylinositol 3-Kinase, The Journal of Immunology, vol.163, pp.1894-1905, 1999.

L. R. Goldin, R. M. Pfeiffer, X. Li, and K. Hemminki, Familial risk of lymphoproliferative tumors in families of patients with chronic lymphocytic leukemia: results from the Swedish Family-Cancer Database, Blood, vol.104, pp.1850-1854, 2004.

D. Gonzalez, P. Martinez, R. Wade, S. Hockley, D. Oscier et al., Mutational Status of the TP53 Gene As a Predictor of Response and Survival in Patients With Chronic Lymphocytic Leukemia: Results From the LRF CLL4, Trial. JCO, vol.29, pp.2223-2229, 2011.

, Licence CC BY

C. C. Goodnow, J. Sprent, B. F. Groth, S. De, and C. G. Vinuesa, Cellular and genetic mechanisms of self tolerance and autoimmunity, 2005.

S. Gordon and A. Mantovani, Diversity and plasticity of mononuclear phagocytes, Eur. J. Immunol, vol.41, pp.2470-2472, 2011.

D. J. De-gorter, E. A. Beuling, R. Kersseboom, S. Middendorp, J. M. Van-gils et al., Bruton's tyrosine kinase and phospholipase Cgamma2 mediate chemokine-controlled B cell migration and homing, Immunity, vol.26, pp.93-104, 2007.

D. Gottardi, A. Alfarano, A. M. De-leo, A. Stacchini, M. Aragno et al., In leukaemic CD5+ B cells the expression of BCL-2 gene family is shifted toward protection from apoptosis, Br. J. Haematol, vol.94, pp.612-618, 1996.

L. Granziero, P. Circosta, C. Scielzo, E. Frisaldi, S. Stella et al., CD100/Plexin-B1 interactions sustain proliferation and survival of normal and leukemic CD5+ B lymphocytes, Blood, vol.101, pp.1962-1969, 2003.

M. Greaves, Clonal expansion in B-CLL: Fungal drivers or self-service?, Journal of Experimental Medicine, vol.210, pp.1-3, 2013.

S. I. Grivennikov, F. R. Greten, K. , and M. , Immunity, inflammation, and cancer. Cell, vol.140, pp.883-899, 2010.

L. Gu, S. Tseng, R. M. Horner, C. Tam, M. Loda et al., Control of TH2 polarization by the chemokine monocyte chemoattractant protein-1, Nature, vol.404, pp.407-411, 2000.

A. Guarini, S. Chiaretti, S. Tavolaro, R. Maggio, N. Peragine et al., BCR ligation induced by IgM stimulation results in gene expression and functional changes only in IgVH unmutated chronic lymphocytic leukemia (CLL) cells, Blood, vol.112, pp.782-792, 2008.

A. Guarini, M. Marinelli, S. Tavolaro, E. Bellacchio, M. Magliozzi et al., ATM gene alterations in chronic lymphocytic leukemia patients induce a distinct gene expression profile and predict disease progression, Haematologica, vol.97, pp.47-55, 2012.

S. Guha, O. Rey, and E. Rozengurt, Neurotensin induces protein kinase C-dependent protein kinase D activation and DNA synthesis in human pancreatic carcinoma cell line PANC-1, Cancer Res, vol.62, pp.1632-1640, 2002.

S. Guha, J. A. Lunn, C. Santiskulvong, and E. Rozengurt, Neurotensin stimulates protein kinase C-dependent mitogenic signaling in human pancreatic carcinoma cell line PANC-1, Cancer Res, vol.63, pp.2379-2387, 2003.

X. Gui, G. Guzman, P. R. Dobner, and S. S. Kadkol, Increased neurotensin receptor-1 expression during progression of colonic adenocarcinoma, Peptides, vol.29, pp.1609-1615, 2008.

D. Gully, M. Canton, R. Boigegrain, F. Jeanjean, J. C. Molimard et al., Biochemical and pharmacological profile of a potent and selective nonpeptide antagonist of the neurotensin receptor, Proc. Natl. Acad. Sci. U.S.A, vol.90, pp.65-69, 1993.

D. Gully, B. Labeeuw, R. Boigegrain, F. Oury-donat, A. Bachy et al.,

R. Suaud-chagny, M. F. Santucci, V. Vita, and N. , Biochemical and pharmacological activities of SR 142948A, a new potent neurotensin receptor antagonist, J. Pharmacol. Exp. Ther, vol.280, pp.802-812, 1997.

M. D. Gunn, V. N. Ngo, K. M. Ansel, E. H. Ekland, J. G. Cyster et al., , 1998.

, A B-cell-homing chemokine made in lymphoid follicles activates Burkitt's lymphoma receptor-1, Nature, vol.391, pp.799-803

C. Guruvayoorappan, Tumor Versus Tumor-Associated Macrophages: How Hot is the Link?, Integr Cancer Ther, vol.7, pp.90-95, 2008.

C. Haase, R. Bergmann, J. Oswald, D. Zips, and J. Pietzsch, Neurotensin Receptors in Adeno-and Squamous Cell Carcinoma, Anticancer Res, vol.26, pp.3527-3533, 2006.

C. Haferlach, F. Dicker, S. Schnittger, W. Kern, and T. Haferlach, Comprehensive genetic characterization of CLL: a study on 506 cases analysed with chromosome banding analysis, interphase FISH, IgVH status and immunophenotyping, Leukemia, vol.21, pp.2442-2451, 2007.

T. Hagemann, J. Wilson, F. Burke, H. Kulbe, N. F. Li et al., Ovarian cancer cells polarize macrophages toward a tumor-associated phenotype, J. Immunol, vol.176, pp.5023-5032, 2006.

F. Hallböök, C. F. Ibáñez, and H. Persson, Evolutionary studies of the nerve growth factor family reveal a novel member abundantly expressed in Xenopus ovary, Neuron, vol.6, pp.845-858, 1991.

M. Hallek, Update on diagnosis, risk stratification, and treatment, Am. J. Hematol, vol.90, pp.446-460, 2015.

M. Hallek, Chronic lymphocytic leukemia: 2017 update on diagnosis, risk stratification, and treatment: HALLEK, American Journal of Hematology, vol.92, pp.946-965, 2017.

M. Hallek, I. Langenmayer, C. Nerl, W. Knauf, H. Dietzfelbinger et al., Elevated serum thymidine kinase levels identify a subgroup at high risk of disease progression in early, nonsmoldering chronic lymphocytic leukemia, Blood, vol.93, pp.1732-1737, 1999.

M. Hallek, B. D. Cheson, D. Catovsky, F. Caligaris-cappio, G. Dighiero et al., Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the International Workshop on Chronic Lymphocytic Leukemia updating the National Cancer Institute-Working Group 1996 guidelines, Blood, vol.111, pp.5446-5456, 2008.

M. Hallek, K. Fischer, G. Fingerle-rowson, A. M. Fink, R. Busch et al., Addition of rituximab to fludarabine and cyclophosphamide in patients with chronic lymphocytic leukaemia: a randomised, open-label, Lancet, vol.376, pp.1164-1174, 2010.

M. Hallek, B. D. Cheson, D. Catovsky, F. Caligaris-cappio, G. Dighiero et al., iwCLL guidelines for diagnosis, indications for treatment, response assessment, and supportive management of CLL, Blood, vol.131, pp.2745-2760, 2018.

T. J. Hamblin, Z. Davis, A. Gardiner, D. G. Oscier, and F. K. Stevenson, Unmutated Licence CC, 1999.

, APRIL. J. Immunol, vol.172, pp.3268-3279

T. E. Heinen, R. P. Santos, A. Da-rocha, M. P. Santos, P. L. Lopez et al., Trk inhibition reduces cell proliferation and potentiates the effects of chemotherapeutic agents in Ewing sarcoma, Oncotarget, vol.7, pp.34860-34880, 2016.

K. Heinig, M. Gätjen, M. Grau, V. Stache, I. Anagnostopoulos et al., Access to follicular dendritic cells is a pivotal step in murine chronic lymphocytic leukemia B-cell activation and proliferation, Cancer Discov, vol.4, pp.1448-1465, 2014.

B. L. Hempstead, D. Martin-zanca, D. R. Kaplan, L. F. Parada, and M. V. Chao, , 1991.

, High-affinity NGF binding requires coexpression of the trk proto-oncogene and the lowaffinity NGF receptor, Nature, vol.350, pp.678-683

Y. Herishanu, P. Pérez-galán, D. Liu, A. Biancotto, S. Pittaluga et al., The lymph node microenvironment promotes B-cell receptor signaling, NF-kappaB activation, and tumor proliferation in chronic lymphocytic leukemia, Blood, vol.117, pp.563-574, 2011.

T. M. Herndon, S. Chen, N. S. Saba, J. Valdez, C. Emson et al., Direct in vivo Evidence for Increased Proliferation of CLL Cells in Lymph Nodes Compared to Bone Marrow and Peripheral Blood, Leukemia, vol.31, pp.1340-1347, 2017.

K. Herrup and E. M. Shooter, Properties of the beta nerve growth factor receptor of avian dorsal root ganglia, Proc. Natl. Acad. Sci. U.S.A, vol.70, pp.3884-3888, 1973.

S. Hewamana, S. Alghazal, T. T. Lin, M. Clement, C. Jenkins et al., The NF-?B subunit Rel A is associated with in vitro survival and clinical disease progression in chronic lymphocytic leukemia and represents a promising therapeutic target, Blood, vol.111, pp.4681-4689, 2008.

J. Hillis, M. O'dwyer, and A. M. Gorman, Neurotrophins and B-cell malignancies, Cell. Mol. Life Sci, vol.73, pp.41-56, 2016.

P. Hillmen, T. Robak, A. Janssens, K. G. Babu, J. Kloczko et al., Chlorambucil plus ofatumumab versus chlorambucil alone in previously untreated patients with chronic lymphocytic leukaemia (COMPLEMENT 1): a randomised, multicentre, open-label phase 3 trial, Lancet, vol.385, pp.1873-1883, 2015.

J. Hoellenriegel, S. A. Meadows, M. Sivina, W. G. Wierda, H. Kantarjian et al., The phosphoinositide 3'-kinase delta inhibitor, CAL-101, inhibits B-cell receptor signaling and chemokine networks in chronic lymphocytic leukemia, Blood, vol.118, pp.3603-3612, 2011.

J. Hoellenriegel, G. P. Coffey, U. Sinha, A. Pandey, M. Sivina et al., Selective, novel spleen tyrosine kinase (Syk) inhibitors suppress chronic lymphocytic leukemia B-cell activation and migration, Leukemia, vol.26, pp.1576-1583, 2012.

J. Hoellenriegel, D. Zboralski, C. Maasch, N. Y. Rosin, W. G. Wierda et al.,

A. Kruschinski and J. A. Burger, The Spiegelmer NOX-A12, a novel CXCL12 inhibitor, interferes with chronic lymphocytic leukemia cell motility and causes chemosensitization, Blood, vol.123, pp.1032-1039, 2014.

M. Holgado-madruga, D. K. Moscatello, D. R. Emlet, R. Dieterich, and A. J. Wong, , 1997.

, Grb2-associated binder-1 mediates phosphatidylinositol 3-kinase activation and the promotion of cell survival by nerve growth factor, Proc. Natl. Acad. Sci. U.S.A, vol.94, pp.12419-12424

B. Holst, N. D. Holliday, A. Bach, C. E. Elling, H. M. Cox et al., Common structural basis for constitutive activity of the ghrelin receptor family, J. Biol. Chem, vol.279, pp.53806-53817, 2004.

R. Hoogeboom, T. A. Wormhoudt, M. R. Schipperus, A. W. Langerak, D. K. Dunn-walters et al., A novel chronic lymphocytic leukemia subset expressing mutated IGHV3-7-encoded rheumatoid factor B-cell receptors that are functionally proficient, Leukemia, vol.27, pp.738-740, 2013.

E. J. Huang and L. F. Reichardt, Trk receptors: roles in neuronal signal transduction, Annu. Rev. Biochem, vol.72, pp.609-642, 2003.

S. Huang, M. Van-arsdall, S. Tedjarati, M. Mccarty, W. Wu et al., Contributions of Stromal Metalloproteinase-9 to Angiogenesis and Growth of Human Ovarian Carcinoma in Mice, J Natl Cancer Inst, vol.94, pp.1134-1142, 2002.

R. Hughes, B. Qian, C. Rowan, M. Muthana, I. Keklikoglou et al., Perivascular M2 Macrophages Stimulate Tumor Relapse after Chemotherapy, Cancer Res, vol.75, pp.3479-3491, 2015.

L. Hui, C. , and Y. , Tumor microenvironment: Sanctuary of the devil, Cancer Letters, vol.368, pp.7-13, 2015.

S. A. Hussain, C. M. Cheney, A. J. Johnson, T. S. Lin, M. R. Grever et al., Mcl-1 is a relevant therapeutic target in acute and chronic lymphoid malignancies: down-regulation enhances rituximab-mediated apoptosis and complement-dependent cytotoxicity, Clin. Cancer Res, vol.13, pp.2144-2150, 2007.

S. Ibrahim, M. Keating, K. A. Do, S. O'brien, Y. O. Huh et al., CD38 expression as an important prognostic factor in B-cell chronic lymphocytic leukemia, Blood, vol.98, pp.181-186, 2001.

A. Iguchi, H. Matsunaga, T. Nomura, M. Gotoh, and N. Sakamoto, Glucoregulatory effects of intrahypothalamic injections of bombesin and other peptides, Endocrinology, vol.114, pp.2242-2246, 1984.

M. Ito, Y. Minamiya, H. Kawai, S. Saito, H. Saito et al., Tumor-Derived TGF?-1 Induces Dendritic Cell Apoptosis in the Sentinel Lymph Node, The Journal of Immunology, vol.176, pp.5637-5643, 2006.

R. Iyer, A. E. Evans, X. Qi, R. Ho, J. E. Minturn et al., Lestaurtinib enhances the antitumor efficacy of chemotherapy in murine xenograft models of neuroblastoma, Clin. Cancer Res, vol.16, pp.1478-1485, 2010.

R. Iyer, C. R. Varela, J. E. Minturn, R. Ho, A. M. Simpson et al., AZ64 inhibits TrkB and enhances the efficacy of Licence CC, 2012.

, chemotherapy and local radiation in neuroblastoma xenografts, Cancer Chemother. Pharmacol, vol.70, pp.477-486

S. M. Jaglowski, A. S. Ruppert, N. A. Heerema, A. Bingman, J. M. Flynn et al., Complex karyotype predicts for inferior outcomes following reduced-intensity conditioning allogeneic transplant for chronic lymphocytic leukaemia, British Journal of Haematology, vol.159, pp.82-87, 2012.

N. Jain, P. A. Thompson, J. A. Burger, G. Borthakur, P. Bose et al., Ibrutinib, fludarabine, cyclophosphamide, and obinutuzumab (GA101) (iFCG) for previously untreated patients with chronic lymphocytic leukemia (CLL) with mutated IGHV and non-del (17p), JCO, vol.35, pp.7522-7522, 2017.

N. Jain, P. A. Thompson, A. Ferrajoli, J. A. Burger, G. Borthakur et al., Combined Venetoclax and Ibrutinib for Patients with Previously Untreated High-Risk CLL, and Relapsed/Refractory CLL: A Phase II Trial, Blood, vol.130, pp.429-429, 2017.

P. Jain, G. M. Nogueras-gonzález, R. Kanagal-shamanna, U. Rozovski, N. Sarwari et al., The absolute percent deviation of IGHV mutation rather than a 98% cut-off predicts survival of chronic lymphocytic leukaemia patients treated with fludarabine, cyclophosphamide and rituximab, Br. J. Haematol, vol.180, pp.33-40, 2018.

D. F. James, L. Werner, J. R. Brown, W. G. Wierda, J. C. Barrientos et al., Lenalidomide and rituximab for the initial treatment of patients with chronic lymphocytic leukemia: a multicenter clinicaltranslational study from the chronic lymphocytic leukemia research consortium, J. Clin. Oncol, vol.32, pp.2067-2073, 2014.

P. Jansen, K. Giehl, J. R. Nyengaard, K. Teng, O. Lioubinski et al., Roles for the pro-neurotrophin receptor sortilin in neuronal development, aging and brain injury, Nat. Neurosci, vol.10, pp.1449-1457, 2007.

F. Jeanneteau, M. J. Garabedian, and M. V. Chao, Activation of Trk neurotrophin receptors by glucocorticoids provides a neuroprotective effect, Proc. Natl. Acad. Sci. U.S.A, vol.105, pp.4862-4867, 2008.

R. Ji, Lymphatic endothelial cells, tumor lymphangiogenesis and metastasis: New insights into intratumoral and peritumoral lymphatics, Cancer Metastasis Rev, vol.25, pp.677-694, 2006.

L. Jia, A. Clear, F. Liu, J. Matthews, N. Uddin et al., Extracellular HMGB1 promotes differentiation of nurselike cells in chronic lymphocytic leukemia, Blood, vol.123, pp.1709-1719, 2014.

R. Jitschin, M. Braun, M. Büttner, K. Dettmer-wilde, J. Bricks et al., CLL-cells induce IDOhi CD14+HLA-DRlo myeloid-derived suppressor cells that inhibit T-cell responses and promote TRegs, Blood, vol.124, pp.750-760, 2014.

R. Jitschin, M. Braun, M. Qorraj, D. Saul, K. Le-blanc et al.,

, Stromal cell-mediated glycolytic switch in CLL cells involves Notch-c-Myc signaling, Blood, vol.125, pp.3432-3436

, Licence CC BY

G. L. Johnson and R. Lapadat, Mitogen-activated protein kinase pathways mediated by ERK, JNK, and p38 protein kinases, Science, vol.298, pp.1911-1912, 2002.

J. A. Jones, T. Robak, J. R. Brown, F. T. Awan, X. Badoux et al., Efficacy and safety of idelalisib in combination with ofatumumab for previously treated chronic lymphocytic leukaemia: an open-label, randomised phase 3 trial, Lancet Haematol, vol.4, pp.114-126, 2017.

J. A. Jones, A. R. Mato, W. G. Wierda, M. S. Davids, M. Choi et al., Venetoclax for chronic lymphocytic leukaemia progressing after ibrutinib: an interim analysis of a multicentre, open-label, phase 2 trial, Lancet Oncol, vol.19, pp.65-75, 2018.

E. Joo, H. E. Broxmeyer, H. Kwon, H. B. Kang, S. Kim et al., Enhancement of Cell Survival by Stromal Cell-Derived Factor-1/CXCL12 Involves Activation of CREB and Induction of Mcl-1 and c-Fos in Factor-Dependent Human Cell Line MO7e, Stem Cells and Development, vol.13, pp.563-570, 2004.

D. H. Josephs, H. J. Bax, and S. N. Karagiannis, Tumour-associated macrophage polarisation and re-education with immunotherapy, Front Biosci (Elite Ed), vol.7, pp.293-308, 2015.

M. R. Junttila and F. J. De-sauvage, Influence of tumour micro-environment heterogeneity on therapeutic response, Nature, vol.501, pp.346-354, 2013.

R. Kaaks, D. Sookthai, A. ?uczy?ska, C. C. Oakes, S. Becker et al., Lag times between lymphoproliferative disorder and clinical diagnosis of chronic lymphocytic leukemia: a prospective analysis using plasma soluble CD23, Cancer Epidemiol. Biomarkers Prev, vol.24, pp.538-545, 2015.

S. Kao, R. K. Jaiswal, W. Kolch, and G. E. Landreth, Identification of the mechanisms regulating the differential activation of the mapk cascade by epidermal growth factor and nerve growth factor in PC12 cells, J. Biol. Chem, vol.276, pp.18169-18177, 2001.

M. Karp, G. , and K. , Antigen stimulation in the development of chronic lymphocytic leukemia, Post?py Higieny i Medycyny Do?wiadczalnej, vol.67, pp.1204-1213, 2013.

M. K. Kashyap, D. Kumar, H. Jones, C. I. Amaya-chanaga, M. Y. Choi et al., Ulocuplumab (BMS-936564 / MDX1338): a fully human anti-CXCR4 antibody induces cell death in chronic lymphocytic leukemia mediated through a reactive oxygen species-dependent pathway, Oncotarget, vol.7, pp.2809-2822, 2016.

G. Katrinakis, D. Kyriakou, H. Papadaki, I. Kalokyri, F. Markidou et al., Defective Natural Killer Cell Activity in B-Cell Chronic Lymphocytic Leukaemia Is Associated with Impaired Release of Natural Killer Cytotoxic Factor(s) but Not of Tumour Necrosis Factor-?, AHA, vol.96, pp.16-23, 1996.

N. E. Kay and J. M. Zarling, Impaired natural killer activity in patients with chronic lymphocytic leukemia is associated with a deficiency of azurophilic cytoplasmic granules in putative NK cells, Blood, vol.63, pp.305-309, 1984.

N. E. Kay, S. M. O'brien, A. R. Pettitt, and S. Stilgenbauer, The role of prognostic factors in assessing "high-risk" subgroups of patients with chronic lymphocytic leukemia, 2007.

, apoptosis induced by oxidative stress, The Journal of Biological Chemistry, vol.289, pp.21205-21216

M. Krajewska, S. Krajewski, J. M. Zapata, T. Van-arsdale, R. D. Gascoyne et al., TRAF-4 expression in epithelial progenitor cells. Analysis in normal adult, fetal, and tumor tissues, Am. J. Pathol, vol.152, pp.1549-1561, 1998.

M. Kraus, M. B. Alimzhanov, N. Rajewsky, and K. Rajewsky, Survival of resting mature B lymphocytes depends on BCR signaling via the Igalpha/beta heterodimer, Cell, vol.117, pp.787-800, 2004.

A. Kröber, T. Seiler, A. Benner, L. Bullinger, E. Brückle et al., V(H) mutation status, CD38 expression level, genomic aberrations, and survival in chronic lymphocytic leukemia, Blood, vol.100, pp.1410-1416, 2002.

A. Kröber, J. Bloehdorn, S. Hafner, A. Bühler, T. Seiler et al., Additional genetic high-risk features such as 11q deletion, 17p deletion, and V3-21 usage characterize discordance of ZAP-70 and VH mutation status in chronic lymphocytic leukemia, J. Clin. Oncol, vol.24, pp.969-975, 2006.

J. S. Kruk, M. S. Vasefi, J. J. Heikkila, and M. A. Beazely, Reactive oxygen species are required for 5-HT-induced transactivation of neuronal platelet-derived growth factor and TrkB receptors, but not for ERK1/2 activation, PLoS ONE, vol.8, p.77027, 2013.

J. S. Kruk, M. S. Vasefi, H. Liu, J. J. Heikkila, and M. A. Beazely, 5-HT(1A) receptors transactivate the platelet-derived growth factor receptor type beta in neuronal cells, Cell Signal, vol.25, pp.133-143, 2013.

R. Krzysiek, E. A. Lefèvre, W. Zou, A. Foussat, J. Bernard et al., Antigen receptor engagement selectively induces macrophage inflammatory protein-1 alpha (MIP-1 alpha) and MIP-1 beta chemokine production in human B cells, J. Immunol, vol.162, pp.4455-4463, 1999.

K. Kubota, M. Moriyama, S. Furukawa, H. A. Rafiul, Y. Maruse et al., CD163 + CD204 + tumor-associated macrophages contribute to T cell regulation via interleukin-10 and PD-L1 production in oral squamous cell carcinoma, Scientific Reports, vol.7, p.1755, 2017.

T. Kurosaki, Regulation of B-cell signal transduction by adaptor proteins, Nat. Rev. Immunol, vol.2, pp.354-363, 2002.

E. Labouyrie, M. Parrens, A. De-mascarel, B. Bloch, and J. P. Merlio, Distribution of NGF receptors in normal and pathologic human lymphoid tissues, J. Neuroimmunol, vol.77, pp.161-173, 1997.

E. Labouyrie, P. Dubus, A. Groppi, F. X. Mahon, J. Ferrer et al., Expression of neurotrophins and their receptors in human bone marrow, Am. J. Pathol, vol.154, pp.405-415, 1999.

D. P. Lad, S. Varma, N. Varma, M. U. Sachdeva, P. Bose et al., , 2015.

, Regulatory T-cell and T-helper 17 balance in chronic lymphocytic leukemia progression and autoimmune cytopenias, Leuk. Lymphoma, vol.56, pp.2424-2428

, Licence CC BY

L. Lagneaux, A. Delforge, D. Bron, C. De-bruyn, and P. Stryckmans, Chronic lymphocytic leukemic B cells but not normal B cells are rescued from apoptosis by contact with normal bone marrow stromal cells, Blood, vol.91, pp.2387-2396, 1998.

K. P. Lam, R. Kühn, and K. Rajewsky, In vivo ablation of surface immunoglobulin on mature B cells by inducible gene targeting results in rapid cell death, Cell, vol.90, pp.1073-1083, 1997.

F. Lamballe, R. Klein, and M. Barbacid, trkC, a new member of the trk family of tyrosine protein kinases, is a receptor for neurotrophin-3, Cell, vol.66, pp.967-979, 1991.

A. Lambiase, L. Bracci-laudiero, S. Bonini, S. Bonini, G. Starace et al., Human CD4+ T cell clones produce and release nerve growth factor and express high-affinity nerve growth factor receptors, J. Allergy Clin. Immunol, vol.100, pp.408-414, 1997.

M. C. Lanasa, Novel insights into the biology of CLL, Hematology Am Soc Hematol Educ Program, pp.70-76, 2010.

D. A. Landau, S. L. Carter, P. Stojanov, A. Mckenna, K. Stevenson et al., Evolution and Impact of Subclonal Mutations in Chronic Lymphocytic Leukemia, Cell, vol.152, pp.714-726, 2013.

D. A. Landau, C. Sun, D. Rosebrock, S. E. Herman, J. Fein et al., The evolutionary landscape of chronic lymphocytic leukemia treated with ibrutinib targeted therapy, Nature Communications, vol.8, p.2185, 2017.

A. M. Lange and H. Lo, Inhibiting TRK Proteins in Clinical Cancer Therapy, Cancers, p.10, 2018.

B. J. Lannutti, S. A. Meadows, S. E. Herman, A. Kashishian, B. Steiner et al., CAL-101, a p110delta selective phosphatidylinositol-3-kinase inhibitor for the treatment of B-cell malignancies, inhibits PI3K signaling and cellular viability, Blood, vol.117, pp.591-594, 2011.

P. Laurent, P. Clerc, M. G. Mattei, P. Forgez, X. Dumont et al., Chromosomal localization of mouse and human neurotensin receptor genes, Mamm. Genome, vol.5, pp.303-306, 1994.

T. W. Lebien and T. F. Tedder, B lymphocytes: how they develop and function, Blood, vol.112, pp.1570-1580, 2008.

F. S. Lee and M. V. Chao, Activation of Trk neurotrophin receptors in the absence of neurotrophins, Proceedings of the National Academy of Sciences, vol.98, pp.3555-3560, 2001.

B. Lee, H. Gao, E. N. Cohen, X. Badoux, W. G. Wierda et al., Treatment with lenalidomide modulates T-cell immunophenotype and cytokine production in patients with chronic lymphocytic leukemia, Cancer, vol.117, pp.3999-4008, 2011.

F. S. Lee, R. Rajagopal, A. H. Kim, P. C. Chang, and M. V. Chao, Activation of Trk neurotrophin receptor signaling by pituitary adenylate cyclase-activating polypeptides, J. Biol. Chem, vol.277, pp.9096-9102, 2002.

J. S. Lee, D. O. Dixon, H. M. Kantarjian, M. J. Keating, and M. Talpaz, Prognosis of Licence CC BY-NC-ND 3.0 chronic lymphocytic leukemia: a multivariate regression analysis of 325 untreated patients, Blood, vol.69, pp.929-936, 1987.

R. Lee, P. Kermani, K. K. Teng, and B. L. Hempstead, Regulation of cell survival by secreted proneurotrophins, Science, vol.294, pp.1945-1948, 2001.

M. Leserer, A. Gschwind, and A. Ullrich, Epidermal growth factor receptor signal transactivation, IUBMB Life, vol.49, pp.405-409, 2000.

V. Lessmann and T. Brigadski, Mechanisms, locations, and kinetics of synaptic BDNF secretion: an update, Neurosci. Res, vol.65, pp.11-22, 2009.

V. Lessmann, K. Gottmann, and M. Malcangio, Neurotrophin secretion: current facts and future prospects, Prog. Neurobiol, vol.69, pp.341-374, 2003.

C. E. Lewis and J. W. Pollard, Distinct Role of Macrophages in Different Tumor Microenvironments, Cancer Res, vol.66, pp.605-612, 2006.

J. Li, J. Song, Y. Y. Zaytseva, Y. Liu, P. Rychahou et al., An obligatory role for neurotensin in high-fatdiet-induced obesity, Nature, vol.533, pp.411-415, 2016.

Z. Li, G. Beutel, M. Rhein, J. Meyer, C. Koenecke et al., High-affinity neurotrophin receptors and ligands promote leukemogenesis, Blood, vol.113, pp.2028-2037, 2009.

Z. Li, Y. Zhang, Y. Tong, J. Tong, and C. J. Thiele, Trk inhibitor attenuates the BDNF/TrkB-induced protection of neuroblastoma cells from etoposide in vitro and in vivo, Cancer Biol. Ther, vol.16, pp.477-483, 2015.

R. H. Lipsky, K. Xu, D. Zhu, C. Kelly, A. Terhakopian et al., Nuclear factor kappaB is a critical determinant in N-methyl-D-aspartate receptormediated neuroprotection, J. Neurochem, vol.78, pp.254-264, 2001.

Q. Liu and H. Wang, Anti-cancer drug discovery and development: Bcl-2 family small molecule inhibitors, Commun Integr Biol, vol.5, pp.557-565, 2012.

J. Liu, M. Agopiantz, J. Poupon, Z. Wu, P. Just et al., Neurotensin receptor 1 antagonist SR48692 improves response to carboplatin by enhancing apoptosis and inhibiting drug efflux in ovarian cancer, Clin Cancer Res clincanres, p.861, 2017.

P. G. Longo, L. Laurenti, S. Gobessi, A. Petlickovski, M. Pelosi et al., The Akt signaling pathway determines the different proliferative capacity of chronic lymphocytic leukemia B-cells from patients with progressive and stable disease, Leukemia, vol.21, pp.110-120, 2007.

P. G. Longo, L. Laurenti, S. Gobessi, S. Sica, G. Leone et al., The Akt/Mcl-1 pathway plays a prominent role in mediating antiapoptotic signals downstream of the B-cell receptor in chronic lymphocytic leukemia B cells, Blood, vol.111, pp.846-855, 2008.

M. López-fraga, R. Fernández, J. P. Albar, and M. Hahne, Biologically active APRIL is secreted following intracellular processing in the Golgi apparatus by furin convertase, EMBO Rep, vol.2, pp.945-951, 2001.

, Licence CC BY

P. Lu, V. M. Weaver, and Z. Werb, The extracellular matrix: A dynamic niche in cancer progression, J Cell Biol, vol.196, pp.395-406, 2012.

G. Lutzny, T. Kocher, M. Schmidt-supprian, M. Rudelius, L. Klein-hitpass et al., Protein Kinase C-?-Dependent Activation of NF-?B in Stromal Cells Is Indispensable for the Survival of Chronic Lymphocytic Leukemia B Cells In Vivo, Cancer Cell, vol.23, pp.77-92, 2013.

D. Ma, Z. Chen, K. P. Patel, B. M. Mishra, H. Yao et al., Array comparative genomic hybridization analysis identifies recurrent gain of chromosome 2p25.3 involving the ACP1 and MYCN genes in chronic lymphocytic leukemia, Clin Lymphoma Myeloma Leuk, vol.11, issue.1, pp.17-24, 2011.

S. Ma, D. M. Brander, J. F. Seymour, T. J. Kipps, J. C. Barrientos et al., Deep and Durable Responses Following Venetoclax (ABT-199 / GDC-0199) Combined with Rituximab in Patients with Relapsed/Refractory Chronic Lymphocytic Leukemia: Results from a Phase 1b Study, Blood, vol.126, pp.830-830, 2015.

A. W. Macfarlane, M. Jillab, M. R. Smith, R. K. Alpaugh, M. E. Cole et al., , 2017.

F. Mackay and P. Schneider, Cracking the BAFF code, Nat. Rev. Immunol, vol.9, pp.491-502, 2009.

K. J. Maddocks, A. S. Ruppert, G. Lozanski, N. A. Heerema, W. Zhao et al., Etiology of Ibrutinib Therapy Discontinuation and Outcomes in Patients With Chronic Lymphocytic Leukemia, JAMA Oncol, vol.1, pp.80-87, 2015.

H. Maeno, K. Yamada, Y. Santo-yamada, K. Aoki, Y. Sun et al., Comparison of mice deficient in the high-or low-affinity neurotensin receptors, Ntsr1 or Ntsr2, reveals a novel function for Ntsr2 in thermal nociception, Brain Res, vol.998, pp.122-129, 2004.

R. Maffei, S. Fiorcari, J. Bulgarelli, S. Martinelli, I. Castelli et al., Physical contact with endothelial cells through ?1-and ?2-integrins rescues chronic lymphocytic leukemia cells from spontaneous and drug-induced apoptosis and induces a peculiar gene expression profile in leukemic cells, Haematologica, vol.97, pp.952-960, 2012.

R. Maffei, J. Bulgarelli, S. Fiorcari, S. Martinelli, I. Castelli et al., Endothelin-1 promotes survival and chemoresistance in chronic lymphocytic leukemia B cells through ETA receptor, PLoS ONE, vol.9, p.98818, 2014.

P. C. Maisonpierre, L. Belluscio, B. Friedman, R. F. Alderson, S. J. Wiegand et al., NT-3, BDNF, and NGF in the developing rat nervous system: parallel as well as reciprocal patterns of expression, Neuron, vol.5, pp.501-509, 1990.

G. Maki, G. M. Hayes, A. Naji, T. Tyler, E. D. Carosella et al., NK resistance of tumor cells from multiple myeloma and chronic lymphocytic Licence CC BY-NC-ND 3.0 leukemia patients: implication of HLA-G, Leukemia, vol.22, pp.998-1006, 2008.

F. Malavasi, S. Deaglio, R. Damle, G. Cutrona, M. Ferrarini et al., CD38 and chronic lymphocytic leukemia: a decade later, Blood, vol.118, pp.3470-3478, 2011.

J. Malcikova, J. Smardova, L. Rocnova, B. Tichy, P. Kuglik et al., Monoallelic and biallelic inactivation of TP53 gene in chronic lymphocytic leukemia: selection, impact on survival, and response to DNA damage, Blood, vol.114, pp.5307-5314, 2009.

A. Mantovani and M. Locati, Tumor-associated macrophages as a paradigm of macrophage plasticity, diversity, and polarization: lessons and open questions, Arterioscler. Thromb. Vasc. Biol, vol.33, pp.1478-1483, 2013.

A. Mantovani and A. Sica, Macrophages, innate immunity and cancer: balance, tolerance, and diversity, Current Opinion in Immunology, vol.22, pp.231-237, 2010.

A. Mantovani, S. Sozzani, M. Locati, P. Allavena, and A. Sica, Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes, Trends Immunol, vol.23, pp.549-555, 2002.

A. Mantovani, F. Marchesi, A. Malesci, L. Laghi, A. et al., Tumourassociated macrophages as treatment targets in oncology, Nature Reviews Clinical Oncology, vol.14, pp.399-416, 2017.

J. J. Maoret, Y. Anini, C. Rouyer-fessard, D. Gully, and M. Laburthe, Neurotensin and a non-peptide neurotensin receptor antagonist control human colon cancer cell growth in cell culture and in cells xenografted into nude mice, Int. J. Cancer, vol.80, pp.448-454, 1999.

R. Marasca, R. Maffei, S. Martinelli, S. Fiorcari, J. Bulgarelli et al., Clinical heterogeneity of de novo 11q deletion chronic lymphocytic leukaemia: prognostic relevance of extent of 11q deleted nuclei inside leukemic clone, Hematological Oncology, vol.31, pp.88-95, 2013.

A. M. Marini, X. Jiang, X. Wu, F. Tian, D. Zhu et al., Role of brain-derived neurotrophic factor and NF-kappaB in neuronal plasticity and survival: From genes to phenotype, Restor. Neurol. Neurosci, vol.22, pp.121-130, 2004.

I. Marondel, B. Renault, J. Lieman, D. Ward, and R. Kucherlapati, Physical mapping of the human neurotensin gene (NTS) between markers D12S1444 and D12S81 on chromosome 12q21, Genomics, vol.38, pp.243-245, 1996.

M. Marquez, O. Hernández-uzcátegui, A. Cornejo, P. Vargas, D. Costa et al., , 2015.

, Bone marrow stromal mesenchymal cells induce down regulation of CD20 expression on B-CLL: implications for rituximab resistance in CLL, Br. J. Haematol, vol.169, pp.211-218

S. Martin, V. Navarro, J. P. Vincent, and J. Mazella, Neurotensin receptor-1 and -3 complex modulates the cellular signaling of neurotensin in the HT29 cell line, Gastroenterology, vol.123, pp.1135-1143, 2002.

S. Martin, J. Vincent, and J. Mazella, Involvement of the neurotensin receptor-3 in the neurotensin-induced migration of human microglia, J. Neurosci, vol.23, pp.1198-1205, 2003.
URL : https://hal.archives-ouvertes.fr/hal-00091093

F. O. Martinez, G. , and S. , The M1 and M2 paradigm of macrophage activation: time for reassessment, 1000.

, Licence CC BY

A. Martínez-trillos, M. Pinyol, A. Navarro, M. Aymerich, P. Jares et al., Mutations in TLR/MYD88 pathway identify a subset of young chronic lymphocytic leukemia patients with favorable outcome, Blood, vol.123, pp.3790-3796, 2014.

I. Maruyama and I. N. Maruyama, Mechanisms of Activation of Receptor Tyrosine Kinases: Monomers or Dimers, Cells, vol.3, pp.304-330, 2014.

F. Massa, C. Devader, S. Lacas-gervais, S. Béraud-dufour, T. Coppola et al., Impairement of HT29 Cancer Cells Cohesion by the Soluble Form of Neurotensin Receptor-3, Genes Cancer, vol.5, pp.240-249, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02108990

A. R. Mato, B. T. Hill, N. Lamanna, P. M. Barr, C. S. Ujjani et al., Optimal sequencing of ibrutinib, idelalisib, and venetoclax in chronic lymphocytic leukemia: results from a multicenter study of 683 patients, Ann. Oncol, vol.28, pp.1050-1056, 2017.

Y. Matsuda, M. Haneda, K. Kadomatsu, and T. Kobayashi, A proliferation-inducing ligand sustains the proliferation of human naïve (CD27?) B cells and mediates their differentiation into long-lived plasma cells in vitro via transmembrane activator and calcium modulator and cyclophilin ligand interactor and B-cell mature antigen, Cellular Immunology, vol.295, pp.127-136, 2015.

C. Matthews, M. A. Catherwood, T. C. Morris, P. J. Kettle, M. B. Drake et al., Serum TK levels in CLL identify Binet stage A patients within biologically defined prognostic subgroups most likely to undergo disease progression, Eur. J. Haematol, vol.77, pp.309-317, 2006.

E. Matutes, K. Owusu-ankomah, R. Morilla, J. Garcia-marco, A. Houlihan et al., The immunological profile of B-cell disorders and proposal of a scoring system for the diagnosis of CLL, Leukemia, vol.8, pp.1640-1645, 1994.

F. R. Mauro, E. Giammartini, M. Gentile, I. Sperduti, V. Valle et al., Clinical features and outcome of familial chronic lymphocytic leukemia, Haematologica, vol.91, pp.1117-1120, 2006.

J. Mazella and J. Vincent, Internalization and recycling properties of neurotensin receptors, Peptides, vol.27, pp.2488-2492, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00171827

J. Mazella, J. M. Botto, E. Guillemare, T. Coppola, P. Sarret et al., , 1996.

, Structure, functional expression, and cerebral localization of the levocabastine-sensitive neurotensin/neuromedin N receptor from mouse brain, J. Neurosci, vol.16, pp.5613-5620

F. Mcclanahan, B. Hanna, S. Miller, A. J. Clear, P. Lichter et al., PD-L1 checkpoint blockade prevents immune dysfunction and leukemia development in a mouse model of chronic lymphocytic leukemia, Blood, vol.126, pp.203-211, 2015.

J. Meldolesi, Neurotrophin Trk Receptors: New Targets for Cancer Therapy, Rev. Physiol. Biochem. Pharmacol, vol.174, pp.67-79, 2018.

S. Menguy, M. Prochazkova-carlotti, M. Beylot-barry, F. Saltel, B. Vergier et al., PD-L1 and PD-L2 Are Differentially Expressed by Macrophages or Tumor Cells in Primary Cutaneous Diffuse Large B-Cell Lymphoma, Leg Type, Am. J. Surg. Pathol, vol.42, pp.326-334, 2018.

, Licence CC BY

B. T. Messmer, D. Messmer, S. L. Allen, J. E. Kolitz, P. Kudalkar et al., In vivo measurements document the dynamic cellular kinetics of chronic lymphocytic leukemia B cells, J. Clin. Invest, vol.115, pp.755-764, 2005.

J. Michels, J. W. O'neill, C. L. Dallman, A. Mouzakiti, F. Habens et al., Mcl-1 is required for Akata6 B-lymphoma cell survival and is converted to a cell death molecule by efficient caspase-mediated cleavage, Oncogene, vol.23, pp.4818-4827, 2004.

F. Mignini, M. Sabbatini, V. D'andrea, and C. Cavallotti, Neuropeptides of human thymus in normal and pathological conditions, Peptides, vol.32, pp.920-928, 2011.

T. Mijatovic, P. Gailly, V. Mathieu, N. De-nève, P. Yeaton et al., Neurotensin is a versatile modulator of in vitro human pancreatic ductal adenocarcinoma cell (PDAC) migration, Cell. Oncol, vol.29, pp.315-326, 2007.

C. D. Mills and K. Ley, M1 and M2 Macrophages: The Chicken and the Egg of Immunity, J Innate Immun, vol.6, pp.716-726, 2014.

E. Mira, L. Carmona-rodríguez, M. Tardáguila, I. Azcoitia, A. González-martín et al., A lovastatin-elicited genetic program inhibits M2 macrophage polarization and enhances T cell infiltration into spontaneous mouse mammary tumors, Oncotarget, vol.4, pp.2288-2301, 2013.

J. B. Mitchem, D. J. Brennan, B. L. Knolhoff, B. A. Belt, Y. Zhu et al., Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses, Cancer Res, vol.73, pp.1128-1141, 2013.

S. P. Mitra, Neurotensin and Neurotensin Receptors in health and diseases: A brief review, INDIAN J. BIOCHEM. BIOPHYS, vol.54, p.17, 2017.

M. Mitre, A. Mariga, and M. V. Chao, Neurotrophin signalling: novel insights into mechanisms and pathophysiology, Clin. Sci, vol.131, pp.13-23, 2017.

T. Miyashita and J. C. Reed, Tumor suppressor p53 is a direct transcriptional activator of the human bax gene, Cell, vol.80, pp.293-299, 1995.

T. Miyashita, S. Krajewski, M. Krajewska, H. G. Wang, H. K. Lin et al., Tumor suppressor p53 is a regulator of bcl-2 and bax gene expression in vitro and in vivo, Oncogene, vol.9, pp.1799-1805, 1994.

Y. Miyata, K. Ohba, S. Kanda, K. Nomata, J. Eguchi et al., Pathological function of prostaglandin E2 receptors in transitional cell carcinoma of the upper urinary tract, Virchows Arch, vol.448, pp.822-829, 2006.

I. S. Mohammad, J. Zhuang, A. Pettitt, M. Oates, L. et al., Use of differentiated THP. 1 cells to mimic nurse-like cells for its pro-survival effect on chronic lymphocytic leukaemia cells, LEUKEMIA & LYMPHOMA, pp.102-104, 2017.

A. Mohr, Y. Renaudineau, C. Bagacean, J. Pers, C. Jamin et al., , 2016.

, Regulatory B lymphocyte functions should be considered in chronic lymphocytic leukemia

, Licence CC BY

S. Molica, Infections in chronic lymphocytic leukemia: risk factors, and impact on survival, and treatment, Leuk. Lymphoma, vol.13, pp.203-214, 1994.

S. Molica, D. Levato, M. Dell'olio, R. Matera, M. Minervini et al., Cellular expression and serum circulating levels of CD23 in B-cell chronic lymphocytic leukemia. Implications for prognosis, Haematologica, vol.81, pp.428-433, 1996.

M. Montillo, T. Hamblin, M. Hallek, E. Montserrat, and E. Morra, Chronic lymphocytic leukemia: novel prognostic factors and their relevance for risk-adapted therapeutic strategies, Haematologica, vol.90, pp.391-399, 2005.

E. Montserrat, New prognostic markers in CLL, Hematology Am Soc Hematol Educ Program, pp.279-284, 2006.

S. A. Moodie and A. Wolfman, The 3Rs of life: Ras, Raf and growth regulation, Trends in Genetics, vol.10, pp.44-48, 1994.

T. W. Moody, J. Chiles, M. Casibang, E. Moody, D. Chan et al., , 2001.

, SR48692 is a neurotensin receptor antagonist which inhibits the growth of small cell lung cancer cells, Peptides, vol.22, pp.109-115

T. W. Moody, D. C. Chan, S. A. Mantey, P. Moreno, and R. T. Jensen, SR48692 inhibits non-small cell lung cancer proliferation in an EGF receptor-dependent manner, Life Sci, vol.100, pp.25-34, 2014.

T. W. Moody, B. Nuche-berenguer, T. Nakamura, and R. T. Jensen, EGFR Transactivation by Peptide G Protein-Coupled Receptors in Cancer, Curr Drug Targets, vol.17, pp.520-528, 2016.

R. N. Moore, A. P. Osmand, J. A. Dunn, J. G. Joshi, J. W. Koontz et al., Neurotensin regulation of macrophage colony-stimulating factor-stimulated in vitro myelopoiesis, J. Immunol, vol.142, pp.2689-2694, 1989.

F. Morabito, L. Mosca, G. Cutrona, L. Agnelli, G. Tuana et al., Clinical monoclonal B lymphocytosis versus Rai 0 chronic lymphocytic leukemia: A comparison of cellular, cytogenetic, molecular, and clinical features, Clin. Cancer Res, vol.19, pp.5890-5900, 2013.

E. J. Moreau, E. Matutes, R. P. A'hern, A. M. Morilla, R. M. Morilla et al., Improvement of the chronic lymphocytic leukemia scoring system with the monoclonal antibody SN8 (CD79b), Am. J. Clin. Pathol, vol.108, pp.378-382, 1997.

A. Morilla, D. Gonzalez-de-castro, I. Del-giudice, N. Osuji, M. Else et al., Combinations of ZAP-70, CD38 and IGHV mutational status as predictors of time to first treatment in CLL, Leukemia & Lymphoma, vol.49, pp.2108-2115, 2008.

A. Morotti, S. Mila, P. Accornero, E. Tagliabue, and C. Ponzetto, K252a inhibits the oncogenic properties of Met, the HGF receptor, Oncogene, vol.21, pp.4885-4893, 2002.

V. A. Morrison, Infectious complications of chronic lymphocytic leukaemia: pathogenesis, spectrum of infection, preventive approaches, Best Pract Res Clin Haematol, vol.23, pp.145-153, 2010.

, Licence CC BY

M. Motta, L. Rassenti, B. J. Shelvin, S. Lerner, T. J. Kipps et al., Increased expression of CD152 (CTLA-4) by normal T lymphocytes in untreated patients with B-cell chronic lymphocytic leukemia, Leukemia, vol.19, pp.1788-1793, 2005.

L. I. Moura, L. Silva, E. C. Leal, A. Tellechea, M. T. Cruz et al., , 2013.

, Neurotensin Modulates the Migratory and Inflammatory Response of Macrophages under Hyperglycemic Conditions, Biomed Res Int, 2013.

I. Munk-pedersen and J. Reed, Microenvironmental interactions and survival of CLL B-cells, Leuk. Lymphoma, vol.45, pp.2365-2372, 2004.

C. Murdoch, A. Giannoudis, and C. E. Lewis, Mechanisms regulating the recruitment of macrophages into hypoxic areas of tumors and other ischemic tissues, Blood, vol.104, pp.2224-2234, 2004.

W. C. Mustain, P. G. Rychahou, and B. M. Evers, The role of neurotensin in physiologic and pathologic processes, Curr Opin Endocrinol Diabetes Obes, vol.18, pp.75-82, 2011.

M. Muzio, B. Apollonio, C. Scielzo, M. Frenquelli, I. Vandoni et al., Constitutive activation of distinct BCR-signaling pathways in a subset of CLL patients: a molecular signature of anergy, Blood, vol.112, pp.188-195, 2008.

R. M. Myers, J. W. Shearman, M. O. Kitching, A. Ramos-montoya, D. E. Neal et al., Cancer, chemistry, and the cell: molecules that interact with the neurotensin receptors, ACS Chem. Biol, vol.4, pp.503-525, 2009.

C. Nabhan, G. Raca, and Y. L. Wang, Predicting Prognosis in Chronic Lymphocytic Leukemia in the Contemporary Era, JAMA Oncol, vol.1, pp.965-974, 2015.

K. D. Nadezhdin, I. García-carpio, S. A. Goncharuk, K. S. Mineev, A. S. Arseniev et al., Structural Basis of p75 Transmembrane Domain Dimerization, J. Biol. Chem. jbc, 2016.

B. P. Nelson, R. Gupta, G. W. Dewald, S. F. Paternoster, S. T. Rosen et al.,

, Chronic Lymphocytic Leukemia FISH PanelImpact on Diagnosis, Am J Clin Pathol, vol.128, pp.323-332

E. V. Neste, V. Robin, J. Francart, A. Hagemeijer, M. Stul et al., Chromosomal translocations independently predict treatment failure, treatment-free survival and overall survival in B-cell chronic lymphocytic leukemia patients treated with cladribine, Leukemia, vol.21, pp.1715-1722, 2007.

A. P. Ng, A. , and W. S. , Haematopoietic stem cells: past, present and future, Cell Death Discovery, vol.3, p.17002, 2017.

F. Nguyen-khac, S. Chevret, C. Roux, C. Touzeau, A. Cosson et al., Mutational and Cytogenetic Analyses Of 177 CLL Patients With Trisomy 12: A Retrospective Study Of The CLL French Intergroup, Blood, vol.122, pp.4144-4144, 2013.

M. Nishio, T. Endo, N. Tsukada, J. Ohata, S. Kitada et al., Nurselike cells express BAFF and APRIL, which can promote survival of chronic lymphocytic leukemia cells via a paracrine pathway distinct from that of SDF-1?, Blood, vol.106, pp.1012-1020, 2005.

, Licence CC BY

R. L. Nosheny, F. Ahmed, A. Yakovlev, E. M. Meyer, K. Ren et al., Brain-derived neurotrophic factor prevents the nigrostriatal degeneration induced by human immunodeficiency virus-1 glycoprotein 120 in vivo, Eur. J. Neurosci, vol.25, pp.2275-2284, 2007.

D. Nouel, M. P. Faure, J. A. St-pierre, R. Alonso, R. Quirion et al., Differential binding profile and internalization process of neurotensin via neuronal and glial receptors, J. Neurosci, vol.17, pp.1795-1803, 1997.

A. J. Novak, R. J. Bram, N. E. Kay, and D. F. Jelinek, Aberrant expression of Blymphocyte stimulator by B chronic lymphocytic leukemia cells: a mechanism for survival, Blood, vol.100, pp.2973-2979, 2002.

C. Nunes, R. Wong, M. Mason, C. Fegan, S. Man et al., Expansion of a CD8(+)PD-1(+) replicative senescence phenotype in early stage CLL patients is associated with inverted CD4:CD8 ratios and disease progression, Clin. Cancer Res, vol.18, pp.678-687, 2012.

A. Nykjaer and T. E. Willnow, Sortilin: a receptor to regulate neuronal viability and function, Trends Neurosci, vol.35, pp.261-270, 2012.

A. Nykjaer and T. E. Willnow, Sortilin: a receptor to regulate neuronal viability and function, Trends in Neurosciences, vol.35, pp.261-270, 2012.

A. Nykjaer, R. Lee, K. K. Teng, P. Jansen, P. Madsen et al., Sortilin is essential for proNGFinduced neuronal cell death, Nature, vol.427, pp.843-848, 2004.

A. Nykjaer, T. E. Willnow, and C. M. Petersen, p75NTR--live or let die, Curr. Opin. Neurobiol, vol.15, pp.49-57, 2005.

R. H. Oakley, S. A. Laporte, J. A. Holt, L. S. Barak, C. et al., Molecular determinants underlying the formation of stable intracellular G protein-coupled receptor-betaarrestin complexes after receptor endocytosis*, J. Biol. Chem, vol.276, pp.19452-19460, 2001.

S. O'brien, R. R. Furman, S. Coutre, I. W. Flinn, J. A. Burger et al., Single-agent ibrutinib in treatment-naïve and relapsed/refractory chronic lymphocytic leukemia: a 5-year experience, Blood, vol.131, pp.1910-1919, 2018.

E. Oda, R. Ohki, H. Murasawa, J. Nemoto, T. Shibue et al., Noxa, a BH3-only member of the Bcl-2 family and candidate mediator of p53-induced apoptosis, Science, vol.288, pp.1053-1058, 2000.

L. Ohman, L. Franzén, U. Rudolph, L. Birnbaumer, and E. H. Hörnquist, Regression of Peyer's patches in G alpha i2 deficient mice prior to colitis is associated with reduced expression of Bcl-2 and increased apoptosis, Gut, vol.51, pp.392-397, 2002.

Y. Ohtaki, G. Ishii, K. Nagai, S. Ashimine, T. Kuwata et al., Stromal Macrophage Expressing CD204 is Associated with Tumor Aggressiveness in Lung Adenocarcinoma, Journal of Thoracic Oncology, vol.5, pp.1507-1515, 2010.

H. Okuda, A. Kobayashi, B. Xia, M. Watabe, S. K. Pai et al., , 2012.

U. Olszewski, H. , and G. , Neurotensin signaling induces intracellular alkalinization and interleukin-8 expression in human pancreatic cancer cells, Mol Oncol, vol.3, pp.204-213, 2009.

A. Os, S. Bürgler, A. P. Ribes, A. Funderud, D. Wang et al., Chronic Lymphocytic Leukemia Cells Are Activated and Proliferate in Response to Specific T Helper Cells, Cell Reports, vol.4, pp.566-577, 2013.

D. G. Oscier, M. J. Rose-zerilli, N. Winkelmann, D. Gonzalez-de-castro, B. Gomez et al., The clinical significance of NOTCH1 and SF3B1 mutations in the UK LRF CLL4 trial, Blood, vol.121, pp.468-475, 2013.

U. Otten, P. Ehrhard, and R. Peck, Nerve growth factor induces growth and differentiation of human B lymphocytes, Proc. Natl. Acad. Sci. U.S.A, vol.86, pp.10059-10063, 1989.

Q. Ouyang, X. Gong, H. Xiao, J. Zhou, M. Xu et al., Neurotensin promotes the progression of malignant glioma through NTSR1 and impacts the prognosis of glioma patients, Mol. Cancer, vol.14, p.21, 2015.

Q. Ouyang, J. Zhou, W. Yang, H. Cui, M. Xu et al., Oncogenic role of neurotensin and neurotensin receptors in various cancers, Clin. Exp. Pharmacol. Physiol, vol.44, pp.841-846, 2017.

J. Paggetti, F. Haderk, M. Seiffert, B. Janji, U. Distler et al., Exosomes released by chronic lymphocytic leukemia cells induce the transition of stromal cells into cancer-associated fibroblasts, Blood, vol.126, pp.1106-1117, 2015.

C. P. Pallasch, S. Ulbrich, R. Brinker, M. Hallek, R. A. Uger et al., Disruption of T cell suppression in chronic lymphocytic leukemia by CD200 blockade, Leukemia Research, vol.33, pp.460-464, 2009.

S. Palmer, C. A. Hanson, C. S. Zent, L. F. Porrata, B. Laplant et al., Prognostic importance of T and NK-cells in a consecutive series of newly diagnosed patients with chronic lymphocytic leukaemia, British Journal of Haematology, vol.141, pp.607-614, 2008.

P. T. Pang, H. K. Teng, E. Zaitsev, N. T. Woo, K. Sakata et al., Cleavage of proBDNF by tPA/plasmin is essential for long-term hippocampal plasticity, Science, vol.306, pp.487-491, 2004.

H. M. Parry, T. Stevens, C. Oldreive, B. Zadran, T. Mcskeane et al., NK cell function is markedly impaired in patients with chronic lymphocytic leukaemia but is preserved in patients with small lymphocytic lymphoma, Oncotarget, vol.7, pp.68513-68526, 2016.

A. Patapoutian and L. F. Reichardt, Trk receptors: mediators of neurotrophin action, Current Opinion in Neurobiology, vol.11, pp.272-280, 2001.

A. B. Patel, I. Tsilioni, S. E. Leeman, and T. C. Theoharides, Neurotensin stimulates Licence CC, 2016.

, sortilin and mTOR in human microglia inhibitable by methoxyluteolin, a potential therapeutic target for autism, Proc. Natl. Acad. Sci. U.S.A

R. N. Pearse, S. L. Swendeman, Y. Li, D. Rafii, and B. L. Hempstead, A neurotrophin axis in myeloma: TrkB and BDNF promote tumor-cell survival, Blood, vol.105, pp.4429-4436, 2005.

I. M. Pedersen, S. Kitada, L. M. Leoni, J. M. Zapata, J. G. Karras et al., Protection of CLL B cells by a follicular dendritic cell line is dependent on induction of Mcl-1, Blood, vol.100, pp.1795-1801, 2002.

C. Pepper, T. Hoy, and D. P. Bentley, Bcl-2/Bax ratios in chronic lymphocytic leukaemia and their correlation with in vitro apoptosis and clinical resistance, Br. J. Cancer, vol.76, pp.935-938, 1997.

C. Pepper, T. Hoy, and P. Bentley, Elevated Bcl-2/Bax Are a Consistent Feature of Apoptosis Resistance in B-cell Chronic Lymphocytic Leukaemia and Are Correlated with In Vivo Chemoresistance, Leukemia & Lymphoma, vol.28, pp.355-361, 1998.

C. Pepper, T. T. Lin, G. Pratt, S. Hewamana, P. Brennan et al., Mcl-1 expression has in vitro and in vivo significance in chronic lymphocytic leukemia and is associated with other poor prognostic markers, Blood, vol.112, pp.3807-3817, 2008.

E. G. Perdiguero and F. Geissmann, Myb-Independent Macrophages: A Family of Cells That Develops with Their Tissue of Residence and Is Involved in Its Homeostasis, Cold Spring Harb Symp Quant Biol, vol.78, pp.91-100, 2013.

L. Pereira-da-silva, B. Neves, L. Moura, M. T. Cruz, and E. Carvalho, , 2014.

, Neurotensin Decreases the Proinflammatory Status of Human Skin Fibroblasts and Increases Epidermal Growth Factor Expression

A. Philippen, S. Diener, T. Zenz, H. Döhner, S. Stilgenbauer et al., , 2010.

, SYK carries no activating point mutations in patients with chronic lymphocytic leukaemia (CLL), British Journal of Haematology, vol.150, pp.633-636

S. Pinet, B. Bessette, N. Vedrenne, A. Lacroix, L. Richard et al., TrkB-containing exosomes promote the transfer of glioblastoma aggressiveness to YKL-40-inactivated glioblastoma cells, Oncotarget, vol.7, pp.50349-50364, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01819184

A. Poggi, C. Prevosto, S. Catellani, I. Rocco, A. Garuti et al., Engagement of CD31 delivers an activating signal that contributes to the survival of chronic lymphocytic leukemia cells, British Journal of Haematology, vol.151, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00573089

A. Polk, Y. Lu, T. Wang, E. Seymour, N. G. Bailey et al., Colony-Stimulating Factor-1 Receptor Is Required for Nurse-like, Cell Survival in Chronic Lymphocytic Leukemia. Clin. Cancer Res, vol.22, pp.6118-6128, 2016.

D. A. Pollyea, S. Coutre, L. Gore, N. Adler, P. Harris et al., A Dose Escalation Study of Ibrutinib with Lenalidomide for Relapsed and Refractory Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma, Blood, vol.124, pp.1987-1987, 2014.

, Licence CC BY

S. Ponader, S. Chen, J. J. Buggy, K. Balakrishnan, V. Gandhi et al., The Bruton tyrosine kinase inhibitor PCI-32765 thwarts chronic lymphocytic leukemia cell survival and tissue homing in vitro and in vivo, Blood, vol.119, pp.1182-1189, 2012.

M. Ponzoni, C. Doglioni, and F. Caligaris-cappio, Chronic lymphocytic leukemia: the pathologist's view of lymph node microenvironment, Seminars in Diagnostic Pathology, vol.28, pp.161-166, 2011.

N. Prenzel, E. Zwick, H. Daub, M. Leserer, R. Abraham et al., EGF receptor transactivation by G-protein-coupled receptors requires metalloproteinase cleavage of proHB-EGF, Nature, vol.402, pp.884-888, 1999.

D. Puehringer, N. Orel, P. Lüningschrör, N. Subramanian, T. Herrmann et al., EGF transactivation of Trk receptors regulates the migration of newborn cortical neurons, Nat. Neurosci, vol.16, pp.407-415, 2013.

A. Puiggros, J. Delgado, A. Rodriguez-vicente, R. Collado, A. Aventín et al., Biallelic losses of 13q do not confer a poorer outcome in chronic lymphocytic leukaemia: analysis of 627 patients with isolated 13q deletion, British Journal of Haematology, vol.163, pp.47-54, 2013.

N. Purroy, P. Abrisqueta, J. Carabia, C. Carpio, C. Palacio et al., Co-culture of primary CLL cells with bone marrow mesenchymal cells, CD40 ligand and CpG ODN promotes proliferation of chemoresistant CLL cells phenotypically comparable to those proliferating in vivo, Oncotarget, vol.6, pp.7632-7643, 2015.

B. Qian, H. Zhang, J. Li, T. He, E. Yeo et al., FLT1 signaling in metastasis-associated macrophages activates an inflammatory signature that promotes breast cancer metastasis, J. Exp. Med, vol.212, pp.1433-1448, 2015.

Z. Qin, G. Noffz, M. Mohaupt, and T. Blankenstein, Interleukin-10 prevents dendritic cell accumulation and vaccination with granulocyte-macrophage colony-stimulating factor gene-modified tumor cells, The Journal of Immunology, vol.159, pp.770-776, 1997.

D. F. Quail, J. , and J. A. , Microenvironmental regulation of tumor progression and metastasis, Nat. Med, vol.19, pp.1423-1437, 2013.

S. Quijano, A. López, A. Rasillo, J. M. Sayagués, S. Barrena et al., Impact of trisomy 12, del(13q), del(17p), and del(11q) on the immunophenotype, DNA ploidy status, and proliferative rate of leukemic B-cells in chronic lymphocytic leukemia, Cytometry B Clin Cytom, vol.74, pp.139-149, 2008.

K. R. Rai, A. Sawitsky, E. P. Cronkite, A. D. Chanana, R. N. Levy et al., Clinical staging of chronic lymphocytic leukemia, Blood, vol.46, pp.219-234, 1975.

R. Rajagopal and M. V. Chao, A role for Fyn in Trk receptor transactivation by Gprotein-coupled receptor signaling, Mol. Cell. Neurosci, vol.33, pp.36-46, 2006.

M. Ramez, M. Bagot, M. Nikolova, L. Boumsell, N. Vita et al., Functional characterization of neurotensin receptors in human cutaneous T cell lymphoma malignant lymphocytes, J. Invest. Dermatol, vol.117, pp.687-693, 2001.

, Licence CC BY

E. Rampazzo, L. Bonaldi, L. Trentin, C. Visco, S. Keppel et al., Telomere length and telomerase levels delineate subgroups of B-cell chronic lymphocytic leukemia with different biological characteristics and clinical outcomes, Haematologica, vol.97, pp.56-63, 2012.

A. G. Ramsay, A. J. Johnson, A. M. Lee, G. Gorgün, R. Le-dieu et al., Chronic lymphocytic leukemia T cells show impaired immunological synapse formation that can be reversed with an immunomodulating drug, J. Clin. Invest, vol.118, pp.2427-2437, 2008.

A. G. Ramsay, R. Evans, S. Kiaii, L. Svensson, N. Hogg et al., , 2013.

, Chronic lymphocytic leukemia cells induce defective LFA-1-directed T-cell motility by altering Rho GTPase signaling that is reversible with lenalidomide, Blood, vol.121, pp.2704-2714

A. Rao, C. Luo, and P. G. Hogan, Transcription factors of the NFAT family: regulation and function, Annu. Rev. Immunol, vol.15, pp.707-747, 1997.

L. Z. Rassenti, L. Huynh, T. L. Toy, L. Chen, M. J. Keating et al., ZAP-70 compared with immunoglobulin heavy-chain gene mutation status as a predictor of disease progression in chronic lymphocytic leukemia, N. Engl. J. Med, vol.351, pp.893-901, 2004.

L. Z. Rassenti, S. Jain, M. J. Keating, W. G. Wierda, M. R. Grever et al., Relative value of ZAP-70, CD38, and immunoglobulin mutation status in predicting aggressive disease in chronic lymphocytic leukemia, Blood, vol.112, pp.1923-1930, 2008.

J. C. Reed, Mechanisms of Bcl-2 family protein function and dysfunction in health and disease, Behring Inst. Mitt, pp.72-100, 1996.

L. F. Reichardt, Neurotrophin-regulated signalling pathways, Philos. Trans. R. Soc. Lond., B, Biol. Sci, vol.361, pp.1545-1564, 2006.

K. S. Reiners, D. Topolar, A. Henke, V. R. Simhadri, J. Kessler et al., Soluble ligands for NK cell receptors promote evasion of chronic lymphocytic leukemia cells from NK cell anti-tumor activity, Blood, vol.121, pp.3658-3665, 2013.

A. Remaury, N. Vita, S. Gendreau, M. Jung, M. Arnone et al., Targeted inactivation of the neurotensin type 1 receptor reveals its role in body temperature control and feeding behavior but not in analgesia, Brain Res, vol.953, pp.63-72, 2002.

C. René, N. Prat, A. Thuizat, M. Broctawik, O. Avinens et al., , 2014.

M. Reth, Antigen receptor tail clue, Nature, vol.338, pp.383-384, 1989.

D. Ribatti, G. Mangialardi, and A. Vacca, Stephen Paget and the "seed and soil" theory of metastatic dissemination, Clin. Exp. Med, vol.6, pp.145-149, 2006.

F. Richard, S. Barroso, J. Martinez, C. Labbé-jullié, and P. Kitabgi, Agonism, inverse agonism, and neutral antagonism at the constitutively active human neurotensin Licence CC BY-NC-ND 3.0 receptor 2, Mol. Pharmacol, vol.60, pp.1392-1398, 2001.

S. J. Richardson, C. Matthews, M. A. Catherwood, H. D. Alexander, B. S. Carey et al., ZAP-70 expression is associated with enhanced ability to respond to migratory and survival signals in B-cell chronic lymphocytic leukemia (B-CLL), Blood, vol.107, pp.3584-3592, 2006.

J. C. Riches, J. K. Davies, F. Mcclanahan, R. Fatah, S. Iqbal et al., T cells from CLL patients exhibit features of T-cell exhaustion but retain capacity for cytokine production, Blood, vol.121, pp.1612-1621, 2013.

F. Rioux, R. Quirion, R. Kérouac, and S. St-pierre, The pressor effects of neurotensin and related peptides in rats, Neuropeptides, vol.2, pp.295-299, 1982.

A. W. Roberts, M. S. Davids, J. M. Pagel, B. S. Kahl, S. D. Puvvada et al., Targeting BCL2 with Venetoclax in Relapsed Chronic Lymphocytic Leukemia, New England Journal of Medicine, vol.374, pp.311-322, 2016.

L. E. Robertson, W. Plunkett, K. Mcconnell, M. J. Keating, and T. J. Mcdonnell, , 1996.

, Bcl-2 expression in chronic lymphocytic leukemia and its correlation with the induction of apoptosis and clinical outcome, Leukemia, vol.10, pp.456-459

A. Rodríguez-tébar, G. Dechant, R. Götz, and Y. A. Barde, Binding of neurotrophin-3 to its neuronal receptors and interactions with nerve growth factor and brainderived neurotrophic factor, EMBO J, vol.11, pp.917-922, 1992.

V. Rolli, M. Gallwitz, T. Wossning, A. Flemming, W. W. Schamel et al., Amplification of B cell antigen receptor signaling by a Syk/ITAM positive feedback loop, Mol. Cell, vol.10, pp.1057-1069, 2002.

R. W. Rooswinkel, B. Van-de-kooij, E. De-vries, M. Paauwe, R. Braster et al., Antiapoptotic potency of Bcl-2 proteins primarily relies on their stability, not binding selectivity, Blood, vol.123, pp.2806-2815, 2014.

E. Rosati, R. Sabatini, G. Rampino, A. Tabilio, M. Di-ianni et al., Constitutively activated Notch signaling is involved in survival and apoptosis resistance of B-CLL cells, Blood, vol.113, pp.856-865, 2009.

S. Roselli, J. Pundavela, Y. Demont, S. Faulkner, S. Keene et al., Sortilin is associated with breast cancer aggressiveness and contributes to tumor cell adhesion and invasion, Oncotarget, vol.6, pp.10473-10486, 2015.

A. Rosén, F. Murray, C. Evaldsson, and R. Rosenquist, Antigens in chronic lymphocytic leukemia-Implications for cell origin and leukemogenesis, Seminars in Cancer Biology, vol.20, pp.400-409, 2010.

S. Rosenblum, T. N. Smith, N. Wang, J. Y. Chua, E. Westbroek et al.,

, BDNF Pretreatment of Human Embryonic-Derived Neural Stem Cells Improves Cell Survival and Functional Recovery after Transplantation in Hypoxic-Ischemic Stroke, Cell Transplant, vol.24, pp.2449-2461

M. J. Rose-zerilli, J. Forster, H. Parker, A. Parker, A. E. Rodríguez et al.,

A. Steele, A. J. Collins, A. Young, and B. D. , ATM mutation rather than BIRC3 deletion and/or mutation predicts reduced survival in 11q-deleted chronic lymphocytic leukemia: data from the UK LRF CLL4 trial, Haematologica, vol.99, pp.736-742, 2014.

P. Rosini, G. De-chiara, M. Lucibello, E. Garaci, F. Cozzolino et al., NGF withdrawal induces apoptosis in CESS B cell line through p38 MAPK activation and Bcl-2 phosphorylation, Biochem. Biophys. Res. Commun, vol.278, pp.753-759, 2000.

D. Rossi and G. Gaidano, ATM and chronic lymphocytic leukemia: mutations, and not only deletions, matter, Haematologica, vol.97, pp.5-8, 2012.

D. Rossi, E. Sozzi, A. Puma, L. De-paoli, S. Rasi et al., The prognosis of clinical monoclonal B cell lymphocytosis differs from prognosis of Rai 0 chronic lymphocytic leukaemia and is recapitulated by biological risk factors, Br. J. Haematol, vol.146, pp.64-75, 2009.

D. Rossi, M. Cerri, C. Deambrogi, E. Sozzi, S. Cresta et al., The Prognostic Value of TP53 Mutations in Chronic Lymphocytic Leukemia Is Independent of Del17p13: Implications for Overall Survival and Chemorefractoriness, Clin Cancer Res, vol.15, pp.995-1004, 2009.

D. Rossi, A. Bruscaggin, V. Spina, S. Rasi, H. Khiabanian et al., Mutations of the SF3B1 splicing factor in chronic lymphocytic leukemia: association with progression and fludarabine-refractoriness, Blood, vol.118, pp.6904-6908, 2011.

D. Rossi, M. Fangazio, S. Rasi, T. Vaisitti, S. Monti et al., Disruption of BIRC3 associates with fludarabine chemorefractoriness in TP53 wild-type chronic lymphocytic leukemia, Blood, vol.119, pp.2854-2862, 2012.

D. Rossi, S. Rasi, G. Fabbri, V. Spina, M. Fangazio et al., Mutations of NOTCH1 are an independent predictor of survival in chronic lymphocytic leukemia, Blood, vol.119, pp.521-529, 2012.

D. Rossi, V. Spina, F. Forconi, D. Capello, M. Fangazio et al., Molecular history of Richter syndrome: origin from a cell already present at the time of chronic lymphocytic leukemia diagnosis, Int. J. Cancer, vol.130, pp.3006-3010, 2012.

D. Rossi, S. Rasi, V. Spina, A. Bruscaggin, S. Monti et al., Integrated mutational and cytogenetic analysis identifies new prognostic subgroups in chronic lymphocytic leukemia, Blood, vol.121, pp.1403-1412, 2013.

D. Rossi, L. Terzi-di-bergamo, L. D. Paoli, M. Cerri, G. Ghilardi et al., Molecular prediction of durable remission after first-line fludarabine-cyclophosphamide-rituximab in chronic lymphocytic leukemia, Blood, vol.126, pp.1921-1924, 2015.

D. J. Rossi, C. H. Jamieson, and I. L. Weissman, Stems cells and the pathways to aging and cancer, Cell, vol.132, pp.681-696, 2008.

W. H. Rostène, A. , and M. J. , Neurotensin and neuroendocrine regulation, 1997.

, Licence CC BY

, Front Neuroendocrinol, vol.18, pp.115-173

G. Roussy, M. Dansereau, L. Doré-savard, K. Belleville, N. Beaudet et al., Spinal NTS1 receptors regulate nociceptive signaling in a rat formalin tonic pain model, Journal of Neurochemistry, vol.105, pp.1100-1114, 2008.

P. P. Roux and P. A. Barker, Neurotrophin signaling through the p75 neurotrophin receptor, Prog. Neurobiol, vol.67, pp.203-233, 2002.

P. P. Roux, A. L. Bhakar, T. E. Kennedy, and P. A. Barker, The p75 neurotrophin receptor activates Akt (protein kinase B) through a phosphatidylinositol 3-kinase-dependent pathway, J. Biol. Chem, vol.276, pp.23097-23104, 2001.

C. Rozman and E. Montserrat, Chronic lymphocytic leukemia, N. Engl. J. Med, vol.333, pp.1052-1057, 1995.

B. Ruffell and L. M. Coussens, Macrophages and therapeutic resistance in cancer, Cancer Cell, vol.27, pp.462-472, 2015.

J. Ruland and T. W. Mak, Transducing signals from antigen receptors to nuclear factor kappaB, Immunol. Rev, vol.193, pp.93-100, 2003.

L. Rushton, A. R. Schnatter, G. Tang, and D. C. Glass, Acute myeloid and chronic lymphoid leukaemias and exposure to low-level benzene among petroleum workers, Br J Cancer, vol.110, pp.783-787, 2014.

S. Saada, P. Marget, A. Fauchais, M. Lise, G. Chemin et al., Differential Expression of Neurotensin and Specific Receptors, NTSR1 and NTSR2, in Normal and Malignant Human B Lymphocytes, The Journal of Immunology, vol.189, pp.5293-5303, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01819265

B. Samah, F. Porcheray, and G. Gras, Neurotrophins modulate monocyte chemotaxis without affecting macrophage function, Clin. Exp. Immunol, vol.151, pp.476-486, 2008.

J. Sánchez-sánchez, J. Arévalo, J. Sánchez-sánchez, and J. C. Arévalo, A Review on Ubiquitination of Neurotrophin Receptors: Facts and Perspectives, International Journal of Molecular Sciences, vol.18, p.630, 2017.

M. Sarfati, S. Chevret, C. Chastang, G. Biron, P. Stryckmans et al., Prognostic importance of serum soluble CD23 level in chronic lymphocytic leukemia, Blood, vol.88, pp.4259-4264, 1996.

P. Sarret, A. Beaudet, J. P. Vincent, and J. Mazella, Regional and cellular distribution of low affinity neurotensin receptor mRNA in adult and developing mouse brain, 1998.

, Comp. Neurol, vol.394, pp.344-356

P. Sarret, L. Gendron, P. Kilian, H. M. Nguyen, N. Gallo-payet et al., Pharmacology and functional properties of NTS2 neurotensin receptors in cerebellar granule cells, J. Biol. Chem, vol.277, pp.36233-36243, 2002.

P. Sarret, A. Perron, T. Stroh, and A. Beaudet, Immunohistochemical distribution of NTS2 neurotensin receptors in the rat central nervous system, J. Comp. Neurol, vol.461, pp.520-538, 2003.

E. Satterwhite, T. Sonoki, T. G. Willis, L. Harder, R. Nowak et al., The BCL11 gene family: involvement of BCL11A in lymphoid malignancies, Blood, vol.98, pp.3413-3420, 2001.

D. Saulep-easton, F. B. Vincent, P. S. Quah, A. Wei, S. B. Ting et al., The BAFF receptor TACI controls IL-10 production by regulatory B cells and CLL B cells, Leukemia, vol.30, pp.163-172, 2016.

J. K. Sax, P. Fei, M. E. Murphy, E. Bernhard, S. J. Korsmeyer et al., BID regulation by p53 contributes to chemosensitivity, Nat. Cell Biol, vol.4, pp.842-849, 2002.

L. Scarfò, A. J. Ferreri, and P. Ghia, Chronic lymphocytic leukaemia, Critical Reviews in Oncology/Hematology, vol.104, pp.169-182, 2016.

A. M. Scharenberg, L. A. Humphries, and D. J. Rawlings, Calcium signalling and cellfate choice in B cells, Nat. Rev. Immunol, vol.7, pp.778-789, 2007.

M. Schena, D. Gottardi, P. Ghia, L. Larsson, M. Carlsson et al., The Role of Bcl-2 in the Pathogenesis of B Chronic Lymphocytic Leukemia, Leukemia & Lymphoma, vol.11, pp.173-179, 1993.

T. Schioppa, B. Uranchimeg, A. Saccani, S. K. Biswas, A. Doni et al., Regulation of the Chemokine Receptor CXCR4 by Hypoxia, Journal of Experimental Medicine, vol.198, pp.1391-1402, 2003.

A. Schober, K. Huber, J. Fey, and K. Unsicker, Distinct populations of macrophages in the adult rat adrenal gland: a subpopulation with neurotrophin-4-like immunoreactivity, Cell Tissue Res, vol.291, pp.365-373, 1998.

S. F. Schoppmann, P. Birner, J. Stöckl, R. Kalt, R. Ullrich et al., Tumor-Associated Macrophages Express Lymphatic Endothelial Growth Factors and Are Related to Peritumoral Lymphangiogenesis, Am J Pathol, vol.161, pp.947-956, 2002.

A. Schotte, W. Rostène, and P. M. Laduron, Different subcellular localization of neurotensin-receptor and neurotensin-acceptor sites in the rat brain dopaminergic system, J. Neurochem, vol.50, pp.1026-1031, 1988.

F. Schriever and D. Huhn, New directions in the diagnosis and treatment of chronic lymphocytic leukaemia, Drugs, vol.63, pp.953-969, 2003.

R. Schroers, F. Griesinger, L. Trümper, D. Haase, B. Kulle et al., Combined analysis of ZAP-70 and CD38 expression as a predictor of disease progression in B-cell chronic lymphocytic leukemia, Leukemia, vol.19, pp.750-758, 2005.

B. Schuhmann, A. Dietrich, S. Sel, C. Hahn, M. Klingenspor et al., A role for brain-derived neurotrophic factor in B cell development, J. Neuroimmunol, vol.163, pp.15-23, 2005.

A. Schulz, C. Dürr, T. Zenz, H. Döhner, S. Stilgenbauer et al., Lenalidomide reduces survival of chronic lymphocytic leukemia cells in primary cocultures by altering the myeloid microenvironment, Blood, vol.121, pp.2503-2511, 2013.

C. Schulz, E. G. Perdiguero, L. Chorro, H. Szabo-rogers, N. Cagnard et al., A Lineage of Myeloid Cells Licence CC, 2012.

, Independent of Myb and Hematopoietic Stem Cells, Science, vol.336, pp.86-90

S. Schulz, C. Röcken, M. P. Ebert, and S. Schulz, Immunocytochemical identification of low-affinity NTS2 neurotensin receptors in parietal cells of human gastric mucosa, J. Endocrinol, vol.191, pp.121-128, 2006.

C. Scielzo, B. Apollonio, L. Scarfò, A. Janus, M. Muzio et al., The functional in vitro response to CD40 ligation reflects a different clinical outcome in patients with chronic lymphocytic leukemia, Leukemia, vol.25, pp.1760-1767, 2011.

F. Secreto, M. Manske, T. Price-troska, S. Ziesmer, L. S. Hodge et al., B-cell activating factor-receptor specific activation of tumor necrosis factor receptor associated factor 6 and the phosphatidyl inositol 3-kinase pathway in lymphoma B cells, Leukemia & Lymphoma, vol.55, pp.1884-1892, 2014.

I. Sehgal, S. Powers, B. Huntley, G. Powis, M. Pittelkow et al., , 1994.

, Neurotensin is an autocrine trophic factor stimulated by androgen withdrawal in human prostate cancer, Proc. Natl. Acad. Sci. U.S.A, vol.91, pp.4673-4677

L. Sellmann, S. Gesk, C. Walter, M. Ritgen, L. Harder et al., Trisomy 19 is associated with trisomy 12 and mutated IGHV genes in B-chronic lymphocytic leukaemia, British Journal of Haematology, vol.138, pp.217-220, 2007.

L. Sellmann, D. De-beer, M. Bartels, B. Opalka, H. Nückel et al., Telomeres and prognosis in patients with chronic lymphocytic leukaemia, Int. J. Hematol, vol.93, pp.74-82, 2011.

J. F. Seymour, T. J. Kipps, B. Eichhorst, P. Hillmen, J. D'rozario et al., Venetoclax-Rituximab in Relapsed or Refractory Chronic Lymphocytic Leukemia, N. Engl. J. Med, vol.378, pp.1107-1120, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02348612

T. D. Shanafelt, P. Ghia, M. C. Lanasa, O. Landgren, and A. C. Rawstron, , 2010.

, Monoclonal B-cell lymphocytosis (MBL): biology, natural history and clinical management, Leukemia, vol.24, pp.512-520

M. Sheng and C. Sala, PDZ domains and the organization of supramolecular complexes, Annu. Rev. Neurosci, vol.24, pp.1-29, 2001.

S. Shimizu, J. Tsukada, T. Sugimoto, N. Kikkawa, K. Sasaki et al., Identification of a novel therapeutic target for head and neck squamous cell carcinomas: a role for the neurotensin-neurotensin receptor 1 oncogenic signaling pathway, Int. J. Cancer, vol.123, pp.1816-1823, 2008.

A. Sica and A. Mantovani, Macrophage plasticity and polarization: in vivo veritas, J Clin Invest, vol.122, pp.787-795, 2012.

A. Sica, A. Saccani, B. Bottazzi, N. Polentarutti, A. Vecchi et al., Autocrine Production of IL-10 Mediates Defective IL-12 Production and NF-?B Activation in Tumor-Associated Macrophages, The Journal of Immunology, vol.164, pp.762-767, 2000.

E. Sierra-filardi, C. Nieto, A. Domínguez-soto, R. Barroso, P. Sánchez-mateos et al., , 2014.

, Licence CC BY

, -CSF: identification of CCL2/CCR2-dependent gene expression profile, CCL2 shapes macrophage polarization by GM-CSF and M, vol.192, pp.3858-3867

L. Da-silva, B. M. Neves, L. Moura, M. T. Cruz, and E. Carvalho, Neurotensin downregulates the pro-inflammatory properties of skin dendritic cells and increases epidermal growth factor expression, Biochim. Biophys. Acta, vol.1813, pp.1863-1871, 2011.

K. S. Siveen and G. Kuttan, Role of macrophages in tumour progression, Immunol. Lett, vol.123, pp.97-102, 2009.

M. Sivina, E. Hartmann, T. J. Kipps, L. Rassenti, D. Krupnik et al., CCL3 (MIP-1?) plasma levels and the risk for disease progression in chronic lymphocytic leukemia, Blood, vol.117, pp.1662-1669, 2011.

S. M. Smith, B. N. Pitcher, S. Jung, N. L. Bartlett, N. Wagner-johnston et al., Safety and tolerability of idelalisib, lenalidomide, and rituximab in relapsed and refractory lymphoma: the Alliance for Clinical Trials in Oncology A051201 and A051202 phase 1 trials, Lancet Haematol, vol.4, pp.176-182, 2017.

L. F. Sniderhan, T. M. Garcia-bates, M. Burgart, S. H. Bernstein, R. P. Phipps et al., Neurotrophin signaling through tropomyosin receptor kinases contributes to survival and proliferation of non-Hodgkin lymphoma, Exp. Hematol, vol.37, pp.1295-1309, 2009.

S. Somaï, A. Gompel, W. Rostène, and P. Forgez, Neurotensin counteracts apoptosis in breast cancer cells, Biochem. Biophys. Res. Commun, vol.295, pp.482-488, 2002.

F. Souazé, V. Viardot-foucault, N. Roullet, M. Toy-miou-leong, A. Gompel et al., Neurotensin receptor 1 gene activation by the Tcf/beta-catenin pathway is an early event in human colonic adenomas, Carcinogenesis, vol.27, pp.708-716, 2006.

F. Souazé, S. Dupouy, V. Viardot-foucault, E. Bruyneel, S. Attoub et al., Expression of neurotensin and NT1 receptor in human breast cancer: a potential role in tumor progression, Cancer Res, vol.66, pp.6243-6249, 2006.

M. Spaargaren, E. A. Beuling, M. L. Rurup, H. P. Meijer, M. D. Klok et al., The B cell antigen receptor controls integrin activity through Btk and PLCgamma2, J. Exp. Med, vol.198, pp.1539-1550, 2003.

D. E. Spaner, Y. Shi, D. White, J. Mena, C. Hammond et al., Immunomodulatory effects of Toll-like receptor-7 activation on chronic lymphocytic leukemia cells, Leukemia, vol.20, pp.286-295, 2006.

H. E. Speedy, D. Bernardo, M. C. Sava, G. P. Dyer, M. J. Holroyd et al., A genome-wide association study identifies multiple susceptibility loci for chronic lymphocytic leukemia, Nat. Genet, vol.46, pp.56-60, 2014.

J. E. Stadanlick, M. Kaileh, F. G. Karnell, J. L. Scholz, J. P. Miller et al., Tonic B cell antigen receptor signals supply an NF-kappaB substrate for prosurvival BLyS signaling, Nat. Immunol, vol.9, pp.1379-1387, 2008.

, Licence CC BY

B. Stamatopoulos, N. Meuleman, C. De-bruyn, K. Pieters, P. Mineur et al., AMD3100 disrupts the cross-talk between chronic lymphocytic leukemia cells and a mesenchymal stromal or nurselike cell-based microenvironment: pre-clinical evidence for its association with chronic lymphocytic leukemia treatments, Haematologica, vol.97, pp.608-615, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02061460

J. Starczynski, C. Pepper, G. Pratt, L. Hooper, A. Thomas et al., Common Polymorphism G(-248)A in the Promoter Region of the bax Gene Results in Significantly Shorter Survival in Patients With Chronic Lymphocytic Leukemia Once Treatment Is Initiated, JCO, vol.23, pp.1514-1521, 2005.

C. Sternini, D. Su, J. Arakawa, R. De-giorgio, D. W. Rickman et al., Cellular localization of Pan-trk immunoreactivity and trkC mRNA in the enteric nervous system, J. Comp. Neurol, vol.368, pp.597-607, 1996.

M. Steurer, M. Montillo, L. Scarfò, F. R. Mauro, J. Andel et al., Results from a Phase IIa Study of the Anti-CXCL12 Spiegelmer Olaptesed Pegol (NOX-A12) in Combination with Bendamustine/Rituximab in Patients with Chronic Lymphocytic Leukemia, Blood, vol.124, pp.1996-1996, 2014.

F. K. Stevenson and F. Caligaris-cappio, Chronic lymphocytic leukemia: revelations from the B-cell receptor, Blood, vol.103, pp.4389-4395, 2004.

F. K. Stevenson, S. Krysov, A. J. Davies, A. J. Steele, and G. Packham, B-cell receptor signaling in chronic lymphocytic leukemia, Blood, vol.118, pp.4313-4320, 2011.

Z. M. Sthoeger, M. Wakai, D. B. Tse, V. P. Vinciguerra, S. L. Allen et al., Production of autoantibodies by CD5-expressing B lymphocytes from patients with chronic lymphocytic leukemia, Journal of Experimental Medicine, vol.169, pp.255-268, 1989.

S. Stilgenbauer, S. Sander, L. Bullinger, A. Benner, E. Leupolt et al., Clonal evolution in chronic lymphocytic leukemia: acquisition of high-risk genomic aberrations associated with unmutated VH, resistance to therapy, and short survival, Haematologica, vol.92, pp.1242-1245, 2007.

S. Stilgenbauer, T. Zenz, D. Winkler, A. Bühler, R. F. Schlenk et al., Subcutaneous Alemtuzumab in Fludarabine-Refractory Chronic Lymphocytic Leukemia: Clinical Results and Prognostic Marker Analyses From the CLL2H Study of the German Chronic Lymphocytic Leukemia Study Group, JCO, vol.27, pp.3994-4001, 2009.

S. Stilgenbauer, A. Schnaiter, P. Paschka, T. Zenz, M. Rossi et al., Gene mutations and treatment outcome in chronic lymphocytic leukemia: results from the CLL8 trial, Blood, vol.123, pp.3247-3254, 2014.

P. Strati and F. Caligaris-cappio, A matter of debate in chronic lymphocytic leukemia: is the occurrence of autoimmune disorders an indicator of chronic lymphocytic leukemia therapy?, Curr Opin Oncol, vol.23, pp.455-460, 2011.

P. Strati, M. J. Keating, S. M. O'brien, J. Burger, A. Ferrajoli et al., Eradication of bone marrow minimal residual disease may prompt early treatment discontinuation in CLL, Blood, vol.123, pp.3727-3732, 2014.

, Licence CC BY

K. Suda, Tumor-associated macrophages-additional effectors at anti-PD-1/PD-L1 therapy?, J Thorac Dis, vol.9, pp.4197-4200, 2017.

Y. Sun, H. Maeno, S. Aoki, and K. Wada, Mouse neurotensin receptor 2 gene (Ntsr2): genomic organization, transcriptional regulation and genetic mapping on chromosome 12, Molecular Brain Research, vol.95, pp.167-171, 2001.

S. Suresh and A. E. Irvine, The NOTCH signaling pathway in normal and malignant blood cell production, J. Cell Commun. Signal, vol.9, pp.5-13, 2015.

M. A. Swartz, N. Iida, E. W. Roberts, S. Sangaletti, M. H. Wong et al., Tumor microenvironment complexity: emerging roles in cancer therapy, Cancer Res, vol.72, pp.2473-2480, 2012.

T. Sweeney, S. E. Spurgeon, J. W. Tyner, A. Agarwal, H. Ho et al., Colony Stimulating Factor 1 Receptor (CSF1R) As a Potential Novel Therapeutic Target in CLL, Blood, vol.126, pp.4439-4439, 2015.

S. H. Swerdlow, E. Campo, S. A. Pileri, N. L. Harris, H. Stein et al., The 2016 revision of the World Health Organization classification of lymphoid neoplasms, Blood, vol.127, pp.2375-2390, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01800015

S. L. Swift, J. E. Burns, and N. J. Maitland, Altered expression of neurotensin receptors is associated with the differentiation state of prostate cancer, Cancer Res, vol.70, pp.347-356, 2010.

T. Tachibana, H. Onodera, T. Tsuruyama, A. Mori, S. Nagayama et al., Increased intratumor Valpha24-positive natural killer T cells: a prognostic factor for primary colorectal carcinomas, Clin. Cancer Res, vol.11, pp.7322-7327, 2005.

A. Tajbakhsh, A. Mokhtari-zaer, M. Rezaee, F. Afzaljavan, M. Rivandi et al., Therapeutic Potentials of BDNF/TrkB in Breast Cancer; Current Status and Perspectives, Journal of Cellular Biochemistry, vol.118, pp.2502-2515, 2017.

, Licence CC BY-NC-ND 3.0 therapeutic strategies, Cancer, vol.103, pp.216-228

A. M. Tsimberidou, S. M. O'brien, J. E. Cortes, S. Faderl, M. Andreeff et al., Phase II study of fludarabine, cytarabine (Ara-C), cyclophosphamide, cisplatin and GM-CSF (FACPGM) in patients with Richter's syndrome or refractory lymphoproliferative disorders, Leuk. Lymphoma, vol.43, pp.767-772, 2002.

N. Tsukada, J. A. Burger, N. J. Zvaifler, and T. J. Kipps, Distinctive features of "nurselike" cells that differentiate in the context of chronic lymphocytic leukemia, Blood, vol.99, pp.1030-1037, 2002.

S. Tsutsui, K. Yasuda, K. Suzuki, K. Tahara, H. Higashi et al., Macrophage infiltration and its prognostic implications in breast cancer: The relationship with VEGF expression and microvessel density, Oncology Reports, vol.14, pp.425-431, 2005.

C. Ujjani, H. Wang, A. Skarbnik, N. Trivedi, P. Ramzi et al., A phase 1 study of lenalidomide and ibrutinib in combination with rituximab in relapsed and refractory CLL, Blood Adv, vol.2, pp.762-768, 2018.

R. T. Uren and A. M. Turnley, Regulation of neurotrophin receptor (Trk) signaling: suppressor of cytokine signaling 2 (SOCS2) is a new player, Front Mol Neurosci, vol.7, 2014.

C. B. Vaegter, P. Jansen, A. W. Fjorback, S. Glerup, S. Skeldal et al., Sortilin associates with Trk receptors to enhance anterograde transport and neurotrophin signaling, Nat. Neurosci, vol.14, pp.54-61, 2011.

A. R. Vaillant, I. Mazzoni, C. Tudan, M. Boudreau, D. R. Kaplan et al., , 1999.

, Depolarization and neurotrophins converge on the phosphatidylinositol 3-kinase-Akt pathway to synergistically regulate neuronal survival, J. Cell Biol, vol.146, pp.955-966

T. Vaisitti, S. Aydin, D. Rossi, F. Cottino, L. Bergui et al., CD38 increases CXCL12-mediated signals and homing of chronic lymphocytic leukemia cells, Leukemia, vol.24, pp.958-969, 2010.

N. C. Valerie, E. V. Casarez, J. O. Dasilva, M. E. Dunlap-brown, S. J. Parsons et al., Inhibition of neurotensin receptor 1 selectively sensitizes prostate cancer to ionizing radiation, Cancer Res, vol.71, pp.6817-6826, 2011.

I. Van-der-auwera, W. Yu, L. Suo, L. Van-neste, P. Van-dam et al., Array-based DNA methylation profiling for breast cancer subtype discrimination, PLoS ONE, vol.5, 2010.

F. Vari, D. Arpon, C. Keane, M. S. Hertzberg, D. Talaulikar et al., Immune evasion via PD-1/PD-L1 on NK-cells and monocyte/macrophages is more prominent in Hodgkin lymphoma than DLBCL, 2018.

M. Vasiadi, A. P. Mondolfi, K. Alysandratos, A. Therianou, A. Katsarou-katsari et al., Neurotensin serum levels and skin gene expression are increased in atopic dermatitis, Br. J. Dermatol, vol.169, pp.695-699, 2013.

J. A. Vega, O. García-suárez, J. Hannestad, M. Pérez-pérez, and A. Germanà, , 2003.

, Licence CC BY

, Neurotrophins and the immune system, J. Anat, vol.203, pp.1-19

C. Vérollet, G. M. Charrière, A. Labrousse, C. Cougoule, V. Le-cabec et al., Extracellular proteolysis in macrophage migration: losing grip for a breakthrough, Eur. J. Immunol, vol.41, pp.2805-2813, 2011.

L. Véronèse, O. Tournilhac, M. Callanan, N. Prie, F. Kwiatkowski et al., Telomeres and chromosomal instability in chronic lymphocytic leukemia, Leukemia, vol.27, pp.490-493, 2013.

M. Vias, G. Burtt, Z. Culig, A. Veerakumarasivam, D. E. Neal et al., A role for neurotensin in bicalutamide resistant prostate cancer cells, Prostate, vol.67, pp.190-202, 2007.

J. P. Vincent, J. Mazella, and P. Kitabgi, Neurotensin and neurotensin receptors, Trends Pharmacol. Sci, vol.20, pp.302-309, 1999.
URL : https://hal.archives-ouvertes.fr/hal-00171827

N. Vita, P. Laurent, S. Lefort, P. Chalon, X. Dumont et al., Cloning and expression of a complementary DNA encoding a high affinity human neurotensin receptor, FEBS Lett, vol.317, pp.139-142, 1993.

N. Vita, F. Oury-donat, P. Chalon, M. Guillemot, M. Kaghad et al., Neurotensin is an antagonist of the human neurotensin NT2 receptor expressed in Chinese hamster ovary cells, Eur. J. Pharmacol, vol.360, pp.265-272, 1998.

K. H. Vousden and X. Lu, Live or let die: the cell's response to p53, Nat. Rev. Cancer, vol.2, pp.594-604, 2002.

R. Vrhovac, A. Delmer, R. Tang, J. P. Marie, R. Zittoun et al.,

, Prognostic significance of the cell cycle inhibitor p27Kip1 in chronic B-cell lymphocytic leukemia, Blood, vol.91, pp.4694-4700

N. Walker, I. Lepee-lorgeoux, J. Fournier, C. Betancur, W. Rostene et al., Tissue distribution and cellular localization of the levocabastine-sensitive neurotensin receptor mRNA in adult rat brain, Brain Res. Mol. Brain Res, vol.57, pp.193-200, 1998.

E. Walsby, A. Buggins, S. Devereux, C. Jones, G. Pratt et al., Development and characterization of a physiologically relevant model of lymphocyte migration in chronic lymphocytic leukemia, Blood, vol.123, pp.3607-3617, 2014.

G. Wang, Y. Liu, X. Wang, Y. Zhang, Y. Zhang et al., The SDF-1/CXCR4 axis promotes recovery after spinal cord injury by mediating bone marrowderived from mesenchymal stem cells, Oncotarget, vol.8, pp.11629-11640, 2017.

J. Wang, N. Li, H. Li, L. Cui, W. et al., Pancreatic cancer bears overexpression of neurotensin and neurotensin receptor subtype-1 and SR 48692 counteracts neurotensin induced cell proliferation in human pancreatic ductal carcinoma cell line PANC, 2011.

, Neuropeptides, vol.45, pp.151-156

J. Wang, J. Chao, W. Chen, M. Kuo, J. J. Yen et al., , 1999.

, The Antiapoptotic Gene mcl-1 Is Up-Regulated by the Phosphatidylinositol 3-Kinase/Akt Signaling Pathway through a Transcription Factor Complex Containing CREB, Molecular and Cellular Biology, vol.19, pp.6195-6206

L. Wang, H. Friess, Z. Zhu, H. Graber, A. Zimmermann et al.,

M. W. Büchler, Neurotensin receptor-1 mRNA analysis in normal pancreas and pancreatic disease, Clin. Cancer Res, vol.6, pp.566-571, 2000.

L. Wang, M. S. Lawrence, Y. Wan, P. Stojanov, C. Sougnez et al., SF3B1 and Other Novel Cancer Genes in Chronic Lymphocytic Leukemia, New England Journal of Medicine, vol.365, pp.2497-2506, 2011.

T. Wang, S. Luo, C. Lin, P. Chang, C. et al., Modulation of p75 neurotrophin receptor under hypoxic conditions induces migration and invasion of C6 glioma cells, Clin. Exp. Metastasis, vol.32, pp.73-81, 2015.

W. Wang, H. Zhu, Y. Wu, Y. Xia, J. Wu et al., Elevated absolute NK cell counts in peripheral blood predict good prognosis in chronic lymphocytic leukemia, J. Cancer Res. Clin. Oncol, vol.144, pp.449-457, 2018.

F. L. Watson, M. A. Porcionatto, A. Bhattacharyya, C. D. Stiles, and R. A. Segal, , 1999.

, TrkA glycosylation regulates receptor localization and activity, J. Neurobiol, vol.39, pp.323-336

E. Wawrzyniak, A. Kotkowska, J. Z. Blonski, M. Siemieniuk-rys, E. Ziolkowska et al., Clonal evolution in CLL patients as detected by FISH versus chromosome banding analysis, and its clinical significance, European Journal of Haematology, vol.92, pp.91-101, 2014.

E. Weagel, C. Smith, P. G. Liu, R. Robison, and K. Neill, Macrophage Polarization and Its Role in Cancer, Journal of Clinical & Cellular Immunology, vol.6, pp.1-8, 2015.

F. Weber, S. N. Byrne, S. Le, D. A. Brown, S. N. Breit et al., Transforming growth factor-?<Subscript>1</Subscript> immobilises dendritic cells within skin tumours and facilitates tumour escape from the immune system, Cancer Immunol Immunother, vol.54, pp.898-906, 2005.

A. T. Weeraratna, S. L. Dalrymple, J. C. Lamb, S. R. Denmeade, S. Miknyoczki et al., Pan-trk inhibition decreases metastasis and enhances host survival in experimental models as a result of its selective induction of apoptosis of prostate cancer cells, Clin. Cancer Res, vol.7, pp.2237-2245, 2001.

I. Weerdt, . De, S. Terpstra, T. Hofland, R. Lameris et al., Chronic Lymphocytic Leukemia (CLL) Cells Are Susceptible to ??-T Cell Mediated Killing, Provided CLL-Derived ??-T Cell Dysfunction Can be Reversed, Blood, vol.126, pp.2914-2914, 2015.

N. S. Weiss, Geographical variation in the incidence of the leukemias and lymphomas, Natl Cancer Inst Monogr, pp.139-142, 1979.

E. J. Wherry, T cell exhaustion, Nat. Immunol, vol.12, pp.492-499, 2011.

W. Wierda, S. O&apos;brien, S. Wen, S. Faderl, G. Garcia-manero et al., Chemoimmunotherapy with fludarabine, cyclophosphamide, and rituximab for relapsed and refractory chronic lymphocytic leukemia, J. Clin. Oncol, vol.23, pp.4070-4078, 2005.

W. G. Wierda, S. O&apos;brien, X. Wang, S. Faderl, A. Ferrajoli et al., Multivariable Model for Time to First Licence CC, 2011.

, Treatment in Patients With Chronic Lymphocytic Leukemia. JCO, vol.29, pp.4088-4095

A. Wiestner, A. Rosenwald, T. S. Barry, G. Wright, R. E. Davis et al., ZAP-70 expression identifies a chronic lymphocytic leukemia subtype with unmutated immunoglobulin genes, inferior clinical outcome, and distinct gene expression profile, Blood, vol.101, pp.4944-4951, 2003.

S. Willimott and S. D. Wagner, Post-transcriptional and post-translational regulation of Bcl2, Biochem. Soc. Trans, vol.38, pp.1571-1575, 2010.

C. M. Wilson, T. Naves, F. Vincent, B. Melloni, F. Bonnaud et al., Sortilin mediates the release and transfer of exosomes in concert with two tyrosine kinase receptors, Journal of Cell Science, vol.127, pp.3983-3997, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01059696

I. P. Witz and O. Levy-nissenbaum, The tumor microenvironment in the post-PAGET era, Cancer Lett, vol.242, pp.1-10, 2006.

M. W. Wooten, M. L. Seibenhener, V. Mamidipudi, M. T. Diaz-meco, P. A. Barker et al., The atypical protein kinase C-interacting protein p62 is a scaffold for NF-kappaB activation by nerve growth factor, J. Biol. Chem, vol.276, pp.7709-7712, 2001.

J. A. Woyach, A. J. Johnson, and J. C. Byrd, The B-cell receptor signaling pathway as a therapeutic target in CLL, Blood, vol.120, pp.1175-1184, 2012.

J. A. Woyach, G. Lozanski, A. S. Ruppert, A. Lozanski, K. A. Blum et al., Outcome of patients with relapsed or refractory chronic lymphocytic leukemia treated with flavopiridol: impact of genetic features, Leukemia, vol.26, pp.1442-1444, 2012.

J. A. Woyach, K. Smucker, L. L. Smith, A. Lozanski, Y. Zhong et al., Prolonged lymphocytosis during ibrutinib therapy is associated with distinct molecular characteristics and does not indicate a suboptimal response to therapy, Blood, vol.123, pp.1810-1817, 2014.

J. A. Woyach, R. R. Furman, T. Liu, H. G. Ozer, M. Zapatka et al., Resistance Mechanisms for the Bruton's Tyrosine Kinase Inhibitor Ibrutinib, New England Journal of Medicine, vol.370, pp.2286-2294, 2014.

J. A. Woyach, A. S. Ruppert, D. Guinn, A. Lehman, J. S. Blachly et al., BTKC481S-Mediated Resistance to Ibrutinib in Chronic Lymphocytic Leukemia, J. Clin. Oncol, vol.35, pp.1437-1443, 2017.

C. J. Wu, CLL clonal heterogeneity: an ecology of competing subpopulations, Blood, vol.120, pp.4117-4118, 2012.

C. Wu, C. F. Lai, and W. C. Mobley, Nerve growth factor activates persistent Rap1 signaling in endosomes, J. Neurosci, vol.21, pp.5406-5416, 2001.

L. Wu, M. Adams, T. Carter, R. Chen, G. Muller et al., lenalidomide enhances natural killer cell and monocyte-mediated antibodydependent cellular cytotoxicity of rituximab-treated CD20+ tumor cells, Clin. Cancer Res, vol.14, pp.4650-4657, 2008.

Z. Wu, D. Martinez-fong, J. Trédaniel, and P. Forgez, Neurotensin and its high Licence CC, 2013.

D. Xia, W. Li, L. Zhang, H. Qian, S. Yao et al., RNA interference-mediated knockdown of brain-derived neurotrophic factor (BDNF) promotes cell cycle arrest and apoptosis in B-cell lymphoma cells, Neoplasma, vol.61, pp.523-532, 2014.

P. Xiao, X. Long, L. Zhang, Y. Ye, J. Guo et al., Neurotensin/IL-8 pathway orchestrates local inflammatory response and tumor invasion by inducing M2 polarization of Tumor-Associated macrophages and epithelialmesenchymal transition of hepatocellular carcinoma cells, OncoImmunology, vol.7, p.1440166, 2018.

N. Yaktapour, R. Übelhart, J. Schüler, K. Aumann, C. Dierks et al., Insulin-like growth factor-1 receptor (IGF1R) as a novel target in chronic lymphocytic leukemia, Blood, vol.122, pp.1621-1633, 2013.

M. Yamada, M. Yamada, A. Lombet, P. Forgez, R. et al., Distinct functional characteristics of levocabastine sensitive rat neurotensin NT2 receptor expressed in Chinese hamster ovary cells, Life Sci, vol.62, pp.375-380, 1998.

T. Yamamoto, Y. Yamanashi, and K. Toyoshima, Association of Src-Family Kinase Lyn with B-Cell Antigen Receptor, Immunological Reviews, vol.132, pp.187-206, 1993.

H. Yan and M. V. Chao, Disruption of cysteine-rich repeats of the p75 nerve growth factor receptor leads to loss of ligand binding, J. Biol. Chem, vol.266, pp.12099-12104, 1991.

H. Yano, R. Torkin, L. A. Martin, M. V. Chao, and K. K. Teng, Proneurotrophin-3 is a neuronal apoptotic ligand: evidence for retrograde-directed cell killing, J. Neurosci, vol.29, pp.14790-14802, 2009.

E. C. Yeiser, N. J. Rutkoski, A. Naito, J. Inoue, and B. D. Carter, Neurotrophin signaling through the p75 receptor is deficient in traf6-/-mice, J. Neurosci, vol.24, pp.10521-10529, 2004.

L. Yi, H. Xiao, M. Xu, X. Ye, J. Hu et al., , 2011.

, Glioma-initiating cells: A predominant role in microglia/macrophages tropism to glioma, Journal of Neuroimmunology, vol.232, pp.75-82

R. D. York, H. Yao, T. Dillon, C. L. Ellig, S. P. Eckert et al.,

M. Younes, Z. Wu, S. Dupouy, A. M. Lupo, N. Mourra et al., Neurotensin (NTS) and its receptor (NTSR1) causes EGFR, HER2 and HER3 over-expression and their autocrine/paracrine activation in lung tumors, confirming responsiveness to erlotinib, Oncotarget, vol.5, pp.8252-8269, 2014.

J. H. Young, Epidemiology and management of infectious complications of contemporary management of chronic leukemias, Infect Disord Drug Targets, vol.11, pp.3-10, 2011.

L. Ysebaert and J. Fournié, Genomic and phenotypic characterization of nurse-like cells that promote drug resistance in chronic lymphocytic leukemia, Leuk. Lymphoma, vol.52, pp.1404-1406, 2011.

J. Yu, Z. Wang, K. W. Kinzler, B. Vogelstein, and L. Zhang, PUMA mediates the apoptotic response to p53 in colorectal cancer cells, Proc. Natl. Acad. Sci. U.S.A, vol.100, 2003.

M. R. Yuille, R. S. Houlston, and D. Catovsky, Anticipation in familial chronic lymphocytic leukaemia, Leukemia, vol.12, pp.1696-1698, 1998.

P. E. Zage, T. C. Graham, L. Z. Zeng, W. Fang, C. Pien et al., The selective Trk inhibitor AZ623 inhibits brain-derived neurotrophic factor-mediated neuroblastoma cell proliferation and signaling and is synergistic with topotecan, Cancer, vol.117, pp.1321-1391, 2011.

B. F. Zamarron, C. , and W. , Dual roles of immune cells and their factors in cancer development and progression, Int. J. Biol. Sci, vol.7, pp.651-658, 2011.

G. P. Zapata and J. M. , Mouse Models of Chronic Lymphocytic Leukemia, 2012.

R. Zaynagetdinov, T. P. Sherrill, V. V. Polosukhin, W. Han, J. A. Ausborn et al., A critical role for macrophages in promotion of urethane-induced lung carcinogenesis, J. Immunol, vol.187, pp.5703-5711, 2011.

A. D. Zelenetz, J. C. Barrientos, J. R. Brown, B. Coiffier, J. Delgado et al., Idelalisib or placebo in combination with bendamustine and rituximab in patients with relapsed or refractory chronic lymphocytic leukaemia: interim results from a phase 3, randomised, double-blind, placebo-controlled trial, Lancet Oncol, vol.18, pp.297-311, 2017.

C. S. Zent and N. E. Kay, Autoimmune complications in chronic lymphocytic leukaemia (CLL), Best Pract Res Clin Haematol, vol.23, pp.47-59, 2010.

T. Zenz, D. Vollmer, M. Trbusek, J. Smardova, A. Benner et al., TP53 mutation profile in chronic lymphocytic leukemia: evidence for a disease specific profile from a comprehensive analysis of 268 mutations, Leukemia, vol.24, pp.2072-2079, 2010.

T. Zenz, B. Eichhorst, R. Busch, T. Denzel, S. Häbe et al., TP53 Mutation and Survival in Chronic Lymphocytic Leukemia, vol.28, pp.4473-4479, 2010.

F. Zeppernick, R. Ahmadi, B. Campos, C. Dictus, B. M. Helmke et al., Stem Cell Marker CD133 Affects Clinical Outcome in Glioma Patients, Clin Cancer Res, vol.14, pp.123-129, 2008.

X. Q. Zhang, J. W. Mu, H. B. Wang, J. Jolkkonen, T. T. Liu et al., Increased protein expression levels of pCREB, 2016.

, /CXCR4 in the hippocampus may be associated with enhanced neurogenesis induced by environmental enrichment, Mol Med Rep, vol.14, pp.2231-2237

Y. Zhang, P. Matthiesen, S. Harder, R. Siebert, G. Castoldi et al., A 3-cM commonly deleted region in 6q21 in leukemias and lymphomas delineated by fluorescence in situ hybridization, Genes Chromosomes Cancer, vol.27, pp.52-58, 2000.

D. Zhao, S. Kuhnt-moore, H. Zeng, J. S. Wu, M. P. Moyer et al., , 2003.

, Licence CC BY

, Neurotensin stimulates IL-8 expression in human colonic epithelial cells through Rho GTPase-mediated NF-kappa B pathways, Am. J. Physiol, vol.284, pp.1397-1404

D. Zhao, Y. Zhan, H. Zeng, H. W. Koon, M. P. Moyer et al., , 2007.

, Neurotensin stimulates expression of early growth response gene-1 and EGF receptor through MAP kinase activation in human colonic epithelial cells, Int. J. Cancer, vol.120, pp.1652-1656

D. Zhao, K. Bakirtzi, Y. Zhan, H. Zeng, H. W. Koon et al., Insulinlike growth factor-1 receptor transactivation modulates the inflammatory and proliferative responses of neurotensin in human colonic epithelial cells, J. Biol. Chem, vol.286, pp.6092-6099, 2011.

H. Zhao, X. Zhang, X. Chen, Y. Li, Z. Ke et al., Isoliquiritigenin, a flavonoid from licorice, blocks M2 macrophage polarization in colitis-associated tumorigenesis through downregulating PGE2 and IL-6, Toxicology and Applied Pharmacology, vol.279, pp.311-321, 2014.

J. Zhou, L. Yi, Q. Ouyang, L. Xu, H. Cui et al., Neurotensin signaling regulates stem-like traits of glioblastoma stem cells through activation of IL, 2014.

, CXCR1/STAT3 pathway, vol.26, pp.2896-2902

, Licence CC BY

. Wilson, Finally, sortilin has been shown to be implicated in the development 64 of different cancers, Regulatory roles of Sortilin and SorLA in immune-related processes 3, 2016.

. Fauchais, Few studies showed the expression of sortilin and SorLA and their potential functions in lymphoid 66 tissues and bone marrow (BM), 2003.

. Yabe-wada, SorLA expression was detected in monocytes, T and B cells and 69 hematopoietic precursors, p.70, 2000.

, Licence CC BY

, Regulatory roles of Sortilin and SorLA in immune-related processes

C. , 191 a transcriptional repressor and regulator of inflammation pathway. ATF3 is a member of the cyclic 192 adenosine monophosphate (cAMP) response element binding protein (CREB), containing a DNA 193 binding region and a bZIP domain, Another transcriptional regulation of SORT1 implicates the activating transcription factor 3 (ATF3), 1994.

. Liang, ATF3 promoter possesses many binding sites of 196 transcription factors such as ATF/CRE (C-rich element), activator protein 1 (AP1) and NF-?B, 197 suggesting that its expression is regulated by stress conditions like pro-inflammatory environment 198, ATF3 homodimerization represses transcription, but conversely, its heterodimerization with Jun 195 proteins activates transcription, vol.202, 1996.

. Rosenberger, These results highlight ATF3 role in the modulation of 206 host immune response under stress or physiopathological conditions, 1999.

A. , ATF3 binding to the proximal promoter region of SORT1 represses SORT1 transcription, p.209, 2012.

D. Ai, J. M. Baez, H. Jiang, D. M. Conlon, A. Hernandez-ono et al., Activation of ER stress and mTORC1 463 suppresses hepatic sortilin-1 levels in obese mice, J. Clin. Invest, vol.122, pp.1677-1687, 2012.

O. M. Andersen, J. Reiche, V. Schmidt, M. Gotthardt, R. Spoelgen et al., , p.465

T. Breiderhoff, P. Jansen, and X. Wu, Neuronal sorting protein-related receptor 466 sorLA/LR11 regulates processing of the amyloid precursor protein, Proc. Natl. Acad. Sci. U. S. A, vol.467, issue.102, pp.13461-13466, 2005.

R. N. Aravalli, P. K. Peterson, and J. R. Lokensgard, Toll-like receptors in defense and 469 damage of the central nervous system, J. Neuroimmune Pharmacol. Off. J. Soc. NeuroImmune 470 Pharmacol, vol.2, pp.297-312, 2007.

A. E. Arrant, V. C. Onyilo, D. E. Unger, and E. D. Roberson, Progranulin Gene Therapy 472 Improves Lysosomal Dysfunction and Microglial Pathology Associated with Frontotemporal 473 Dementia and Neuronal Ceroid Lipofuscinosis, J. Neurosci, vol.38, pp.2341-2358, 2018.

F. Blasi and P. Carmeliet, uPAR: a versatile signalling orchestrator, Nat. Rev. Mol. Cell 475 Biol, vol.3, pp.932-943, 2002.

N. D. Boespflug, S. Kumar, J. W. Mcalees, J. D. Phelan, H. L. Grimes et al., , p.477

M. Karp and C. L. , ATF3 is a novel regulator of mouse neutrophil migration, Blood, vol.123, pp.478-2084, 2014.

M. Canuel, A. Korkidakis, K. Konnyu, and C. R. Morales, Sortilin mediates the lysosomal Licence CC, 2008.

, Regulatory roles of Sortilin and SorLA in immune-related processes 16 targeting of cathepsins D and H, Biochem. Biophys. Res. Commun, vol.373, pp.292-297

A. Carlo, C. Gustafsen, G. Mastrobuoni, M. S. Nielsen, T. Burgert et al., , p.482

A. Nykjaer, J. Herz, and J. Heeren, The pro-neurotrophin receptor sortilin is a major 483 neuronal apolipoprotein E receptor for catabolism of amyloid-? peptide in the brain, J. Neurosci. Off. 484 J. Soc. Neurosci, vol.33, pp.358-370, 2013.

L. Changhua, Y. Jindong, L. Defa, Z. Lidan, Q. Shiyan et al., Conjugated 486 linoleic acid attenuates the production and gene expression of proinflammatory cytokines in weaned 487 pigs challenged with lipopolysaccharide, J. Nutr, vol.135, pp.239-244, 2005.

I. F. Charo and M. B. Taubman, Chemokines in the pathogenesis of vascular disease, 2004.

. Res, , vol.95, pp.858-866

B. P. Chen, G. Liang, J. Whelan, H. , and T. , ATF3 and ATF3 delta Zip. Transcriptional 491 repression versus activation by alternatively spliced isoforms, J. Biol. Chem, vol.269, pp.15819-15826, 1994.

X. Chen, J. Chang, Q. Deng, J. Xu, T. A. Nguyen et al., , p.493

K. F. Chung and J. , Progranulin Does Not Bind Tumor Necrosis Factor (TNF) Receptors 494 and Is Not a Direct Regulator of TNF-Dependent Signaling or Bioactivity in Immune or Neuronal 495 Cells, J. Neurosci, vol.33, pp.9202-9213, 2013.

D. C. Chow, L. A. Wenning, W. M. Miller, and E. T. Papoutsakis, Modeling pO2 497 Distributions in the Bone Marrow Hematopoietic Compartment, 2001.

, Biophys. J, vol.81, pp.685-696

S. Crabé, A. Guay-giroux, A. J. Tormo, D. Duluc, R. Lissilaa et al.,

U. Bigouagou, F. Lefouili, I. Cognet, and W. Ferlin, The IL-27 p28 Subunit Binds, p.501, 2009.

. Cytokine-like, Factor 1 to Form a Cytokine Regulating NK and T Cell Activities Requiring IL-6R 502 for Signaling, J. Immunol, vol.183, pp.7692-7702

E. Dicou, J. Vincent, and J. Mazella, Neurotensin receptor-3/sortilin mediates 504 neurotensin-induced cytokine/chemokine expression in a murine microglial cell line, J. Neurosci, 2004.

. Res, , vol.78, pp.92-99

S. B. Dumanis, T. Burgert, S. Caglayan, A. Füchtbauer, E. Füchtbauer et al., Distinct Functions for Anterograde and Retrograde Sorting of SORLA, p.508, 2015.

, Amyloidogenic Processes in the Brain, J. Neurosci, vol.35, pp.12703-12713

S. F. Evans, K. Irmady, K. Ostrow, T. Kim, A. Nykjaer et al., Neuronal Brain-derived Neurotrophic Factor Is Synthesized in Excess, with Levels 511 Regulated by Sortilin-mediated Trafficking and Lysosomal Degradation, J. Biol. Chem, vol.286, pp.29556-512, 2011.

A. Fauchais, F. Lalloue, M. Lise, A. Boumediene, J. Preud&apos;homme et al., Role of Endogenous Brain-Derived Neurotrophic Factor and Sortilin in B 515 Cell Survival, J. Immunol, vol.181, pp.3027-3038, 2008.

J. Gehrmann, Y. Matsumoto, and G. W. Kreutzberg, Microglia: intrinsic immuneffector cell 517 of the brain, Brain Res. Brain Res. Rev, vol.20, pp.269-287, 1995.

M. Gilchrist, V. Thorsson, B. Li, A. G. Rust, M. Korb et al., Systems biology approaches identify ATF3 as a negative regulator of Toll-like 520 receptor 4, Nature, vol.441, pp.173-178, 2006.

C. Goettsch, J. D. Hutcheson, M. Aikawa, H. Iwata, T. Pham et al., Sortilin mediates vascular calcification via its Licence CC, 2016.

, Regulatory roles of Sortilin and SorLA in immune-related processes 17 recruitment into extracellular vesicles, J. Clin. Invest, vol.126, pp.1323-1336

C. Goettsch, M. Kjolby, A. , and E. , Sortilin and Its Multiple Roles in Cardiovascular 525 and Metabolic Diseases, Arterioscler. Thromb. Vasc. Biol, vol.38, pp.19-25, 2018.

S. Gordon, Alternative activation of macrophages, Nat. Rev. Immunol, vol.3, pp.23-35, 2003.
URL : https://hal.archives-ouvertes.fr/hal-00474829

J. Gu, A. Johns, J. Morser, W. P. Dole, D. R. Greaves et al., Urokinase 528 plasminogen activator receptor promotes macrophage infiltration into the vascular wall of ApoE 529 deficient mice, J. Cell. Physiol, vol.204, pp.73-82, 2005.

M. G. Gutierrez, B. B. Mishra, L. Jordao, E. Elliott, E. Anes et al., NF-B 531 Activation Controls Phagolysosome Fusion-Mediated Killing of Mycobacteria by, 2008.

. Immunol, , vol.181, pp.2651-2663

T. Hai and M. G. Hartman, The molecular biology and nomenclature of the activating 534 transcription factor/cAMP responsive element binding family of transcription factors: activating 535 transcription factor proteins and homeostasis, Gene, vol.273, pp.1-11, 2001.

T. Hai, C. D. Wolfgang, D. K. Marsee, A. E. Allen, and U. Sivaprasad, ATF3 and stress 537 responses, Gene Expr, vol.7, pp.321-335, 1999.

Y. Hashimoto, C. Zhang, J. Kawauchi, I. Imoto, M. T. Adachi et al., An alternatively spliced isoform of transcriptional repressor ATF3 and its 540 induction by stress stimuli, Nucleic Acids Res, vol.30, pp.2398-2406, 2002.

S. Herda, F. Raczkowski, H. Mittrücker, G. Willimsky, K. Gerlach et al., The Sorting Receptor Sortilin Exhibits a 543 Dual Function in Exocytic Trafficking of Interferon-? and Granzyme A in T Cells, Immunity, vol.37, pp.854-866, 2012.

I. Hermans-borgmeyer, G. Hermey, A. Nykjaer, and C. Schaller, Expression of the 100-546 kDa neurotensin receptor sortilin during mouse embryonal development, Brain Res. Mol. Brain Res, vol.547, pp.216-219, 1999.

G. Hermey, The Vps10p-domain receptor family, Cell. Mol. Life Sci, vol.66, pp.2677-2689, 2009.

G. Hermey, S. S. Sjøgaard, C. M. Petersen, A. Nykjaer, and J. Gliemann, Tumour necrosis 550 factor ?-converting enzyme mediates ectodomain shedding of Vps10p-domain receptor family 551 members, Biochem. J, vol.395, pp.285-293, 2006.

C. Hivelin, J. Mazella, and T. Coppola, Sortilin derived propeptide regulation during 553 adipocyte differentiation and inflammation, Biochem. Biophys. Res. Commun, vol.482, pp.87-92, 2017.

H. H. Ho, T. T. Antoniv, J. Ji, and L. B. Ivashkiv, Lipopolysaccharide-Induced 555 Expression of Matrix Metalloproteinases in Human Monocytes Is Suppressed by IFN-? via 556 Superinduction of ATF-3 and Suppression of AP-1, J. Immunol, vol.181, pp.5089-5097, 2008.

W. Hoetzenecker, B. Echtenacher, E. Guenova, K. Hoetzenecker, F. Woelbing et al., ROS-induced ATF3 causes susceptibility to 559 secondary infections during sepsis-associated immunosuppression, Nat. Med, vol.18, pp.128-134, 2012.

L. Jacobsen, P. Madsen, S. K. Moestrup, A. H. Lund, N. Tommerup et al., Molecular characterization of a novel human hybrid-562 type receptor that binds the alpha2-macroglobulin receptor-associated protein, J. Biol. Chem, vol.271, pp.31379-31383, 1996.

L. Jacobsen, P. Madsen, C. Jacobsen, M. S. Nielsen, J. Gliemann et al., , 2001.

, Licence CC BY

, Regulatory roles of Sortilin and SorLA in immune-related processes 18

, Activation and Functional Characterization of the Mosaic Receptor SorLA/LR11, J. Biol. Chem, vol.276, pp.22788-22796

K. Jadhav and Y. Zhang, Activating transcription factor 3 in immune response and 568 metabolic regulation, Liver Res, vol.1, pp.96-102, 2017.

J. Jian, S. Zhao, Q. Tian, E. Gonzalez-gugel, J. J. Mundra et al., , p.570

R. Brunetti and C. Liu, Progranulin directly binds to the CRD2 and CRD3 of TNFR 571 extracellular domains, FEBS Lett, vol.587, pp.3428-3436, 2013.

V. Kaddai, J. Jager, T. Gonzalez, R. Najem-lendom, S. Bonnafous et al., , p.573

Y. Brustel, P. Gual, J. Tanti, and M. Cormont, Involvement of TNF-? in abnormal 574 adipocyte and muscle sortilin expression in obese mice and humans, Diabetologia, vol.52, pp.932-940, 2009.

C. H. Khuu, R. M. Barrozo, T. Hai, and S. L. Weinstein, Activating transcription factor 3 576 (ATF3) represses the expression of CCL4 in murine macrophages, Mol. Immunol, vol.44, pp.1598-1605, 2007.

Y. Kitago, M. Nagae, Z. Nakata, M. Yagi-utsumi, S. Takagi-niidome et al., Structural basis for amyloidogenic peptide recognition by sorLA, 2015.

, Struct. Mol. Biol, vol.22, pp.199-206

M. Kjolby, O. M. Andersen, T. Breiderhoff, A. W. Fjorback, K. M. Pedersen et al., Sort1, encoded by the cardiovascular risk 582 locus 1p13.3, is a regulator of hepatic lipoprotein export, Cell Metab, vol.12, pp.213-223, 2010.

R. Klingenberg, N. Gerdes, R. M. Badeau, A. Gisterå, D. Strodthoff et al., Depletion of FOXP3+ regulatory T 585 cells promotes hypercholesterolemia and atherosclerosis, J. Clin. Invest, vol.123, pp.1323-1334, 2013.

S. C. Klinger, S. Glerup, M. K. Raarup, M. C. Mari, M. Nyegaard et al., , p.587

S. K. Nilsson, M. M. Kjaergaard, and O. Bakke, SorLA regulates the activity of lipoprotein 588 lipase by intracellular trafficking, J Cell Sci, vol.124, pp.1095-1105, 2011.

J. Kofler and C. A. Wiley, Microglia: key innate immune cells of the brain, Toxicol. Pathol, vol.590, pp.103-114, 2011.

M. Koga, H. Kai, H. Yasukawa, T. Yamamoto, Y. Kawai et al., , p.592

K. Egashira and Y. Kataoka, Inhibition of Progression and Stabilization of Plaques by 593, 2007.

, Postnatal Interferon-? Function Blocking in ApoE-Knockout Mice, Circ. Res, vol.101, pp.348-356

S. Kumar-singh, Progranulin and TDP-43: Mechanistic Links and Future Directions, J. Mol, 2011.

. Neurosci, , vol.45, pp.561-573

L. I. Labzin, S. V. Schmidt, S. L. Masters, M. Beyer, W. Krebs et al., , p.597

J. L. Schultze and E. Latz, ATF3 Is a Key Regulator of Macrophage IFN Responses, 2015.

, Immunol, vol.195, pp.4446-4455

P. F. Lai, C. F. Cheng, H. Lin, T. L. Tseng, H. H. Chen et al., ATF3 protects 600 against LPS-induced inflammation in mice via inhibiting HMGB1 expression. Evid. Based 601 Complement, Alternat. Med, p.716481, 2013.

G. Landskron, M. De-la-fuente, P. Thuwajit, C. Thuwajit, and M. A. Hermoso, Chronic 603 Inflammation and Cytokines in the Tumor Microenvironment, 2014.

J. V. Larsen and C. M. Petersen, SorLA in interleukin-6 signaling and turnover, Mol. Cell, 2017.

, Biol, vol.37, pp.641-657

J. V. Larsen, M. Hansen, B. Moller, P. Madsen, J. Scheller et al.,

, Sortilin Facilitates Signaling of Ciliary Neurotrophic Factor and Related Helical Type 1, Regulatory roles of Sortilin and SorLA in immune-related processes 19, 2010.

, Cytokines Targeting the gp130/Leukemia Inhibitory Factor Receptor Heterodimer, Mol. Cell. Biol, vol.609, pp.4175-4187

J. V. Larsen, A. M. Kristensen, L. T. Pallesen, J. Bauer, C. B. Vaegter et al., Cytokine-Like Factor 1, an Essential Facilitator of Cardiotrophin-Like 612 Cytokine:Ciliary Neurotrophic Factor Receptor ? Signaling and sorLA-Mediated Turnover, 2016.

, Cell. Biol, vol.36, pp.1272-1286

Y. Lebovitz, V. M. Ringel-scaia, I. C. Allen, and M. H. Theus, Emerging Developments in 615 Microbiome and Microglia Research: Implications for Neurodevelopmental Disorders, 2018.

. Immunol, , vol.9, 1993.

S. Lehnardt, Innate immunity and neuroinflammation in the CNS: the role of microglia in 618 Toll-like receptor-mediated neuronal injury, Glia, vol.58, pp.253-263, 2010.

J. Lévesque, F. M. Helwani, and I. G. Winkler, The endosteal "osteoblastic" niche and its 620 role in hematopoietic stem cell homing and mobilization, Leukemia, vol.24, pp.1979-1992, 2010.

G. Liang, C. D. Wolfgang, B. P. Chen, T. Chen, H. et al., ATF3 Gene GENOMIC 622 ORGANIZATION, PROMOTER, AND REGULATION, J. Biol. Chem, vol.271, pp.1695-1701, 1996.

A. J. Lusis, Atherosclerosis. Nature, vol.407, pp.233-241, 2000.

A. V. Makeyev and S. A. Liebhaber, The poly(C)-binding proteins: a multiplicity of 625 functions and a search for mechanisms, vol.8, pp.265-278, 2002.

M. Muriel, B. Miriam, V. Zeuschner-dagmar, G. Willie, J. C. Griffith et al.,

M. Claus, J. Cullen-pete, J. Klumperman, G. Hans, and J. , SNX1 Defines an Early 628, 2007.

, Endosomal Recycling Exit for Sortilin and Mannose 6-Phosphate Receptors, Traffic, vol.9, pp.380-393

S. Martin, J. Vincent, and J. Mazella, Involvement of the neurotensin receptor-3 in the 630 neurotensin-induced migration of human microglia, J. Neurosci. Off. J. Soc. Neurosci, vol.23, p.1205, 2003.

S. Martin, E. Dicou, J. Vincent, and J. Mazella, Neurotensin and the neurotensin 633 receptor-3 in microglial cells, J. Neurosci. Res, vol.81, pp.322-326, 2005.

F. O. Martinez, A. Sica, A. Mantovani, and M. Locati, Macrophage activation and 635 polarization, Front. Biosci. J. Virtual Libr, vol.13, pp.453-461, 2008.

A. E. May, R. Schmidt, S. M. Kanse, T. Chavakis, R. W. Stephens et al., Urokinase receptor surface expression regulates monocyte adhesion in 638 acute myocardial infarction, Blood, vol.100, pp.3611-3617, 2002.

J. Mazella, Sortilin/neurotensin receptor-3: a new tool to investigate neurotensin signaling 640 and cellular trafficking?, Cell. Signal, vol.13, pp.1-6, 2001.

J. Mazella, N. Zsürger, V. Navarro, J. Chabry, M. Kaghad et al., The 100-kDa neurotensin receptor is gp95/sortilin, a non-G-protein-643 coupled receptor, J. Biol. Chem, vol.273, pp.26273-26276, 1998.

C. Mccarthy, P. O&apos;gaora, W. G. James, S. Mcclelland, D. J. Fitzgerald et al., , p.645

M. Gaetano, SorLA modulates atheroprotective properties of CLA by regulating monocyte 646 migration, Atherosclerosis, vol.213, pp.400-407, 2010.

S. Mcclelland, C. Cox, R. O&apos;connor, M. De-gaetano, C. Mccarthy et al., , p.648

O. Belton, Conjugated linoleic acid suppresses the migratory and inflammatory phenotype of Licence CC, 2010.

, Regulatory roles of Sortilin and SorLA in immune-related processes

E. Mendoza-barberá, J. Julve, S. K. Nilsson, A. Lookene, J. M. Martín-campos et al., , p.651

A. M. Lechuga-sancho, J. H. Sloan, P. Fuentes-prior, and F. Blanco-vaca, Structural and 652 functional analysis of APOA5 mutations identified in patients with severe hypertriglyceridemia, J. 653 Lipid Res, vol.54, pp.649-661, 2013.

M. Mojic, K. Takeda, and Y. Hayakawa, The Dark Side of IFN-?: Its Role in Promoting 655 Cancer Immunoevasion, Int. J. Mol. Sci, vol.19, 2017.

A. Morinville, S. Martin, M. Lavallée, J. Vincent, A. Beaudet et al., , 2004.

, Internalization and trafficking of neurotensin via NTS3 receptors in HT29 cells, Int. J. Biochem. Cell 658 Biol, vol.36, pp.2153-2168

M. B. Mortensen, M. Kjolby, S. Gunnersen, J. V. Larsen, J. Palmfeldt et al., Targeting sortilin in immune cells reduces proinflammatory cytokines and 661 atherosclerosis, J. Clin. Invest, vol.124, p.5317, 2014.

D. M. Mosser and J. P. Edwards, Exploring the full spectrum of macrophage activation, Nat. 663 Rev. Immunol, vol.8, pp.958-969, 2008.

M. S. Nielsen, P. Madsen, E. I. Christensen, A. Nykjaer, J. Gliemann et al., The sortilin cytoplasmic tail conveys Golgi-endosome transport and binds 666 the VHS domain of the GGA2 sorting protein, EMBO J, vol.20, pp.2180-2190, 2001.

S. K. Nilsson, A. Lookene, J. A. Beckstead, J. Gliemann, R. O. Ryan et al., , 2007.

, Apolipoprotein A-V interaction with members of the low density lipoprotein receptor gene family, 669 Biochemistry, vol.46, pp.3896-3904

S. K. Nilsson, S. Christensen, M. K. Raarup, R. O. Ryan, M. S. Nielsen et al., , 2008.

, Endocytosis of apolipoprotein A-V by members of the low density lipoprotein receptor and the 672 VPS10p domain receptor families, J. Biol. Chem, vol.283, pp.25920-25927

K. Nishii, C. Nakaseko, M. Jiang, N. Shimizu, M. Takeuchi et al., The Soluble Form of LR11 Protein Is a Regulator of Hypoxia-induced, p.675, 2013.

, Plasminogen Activator Receptor (uPAR)-mediated Adhesion of Immature Hematological Cells

, Biol. Chem, vol.288, pp.11877-11886

R. Nurieva, X. O. Yang, G. Martinez, Y. Zhang, A. D. Panopoulos et al., , p.678

S. S. Watowich and A. M. Jetten, Essential autocrine regulation by IL-21 in the generation 679 of inflammatory T cells, Nature, vol.448, pp.480-483, 2007.

A. C. Nyborg, T. B. Ladd, C. W. Zwizinski, J. J. Lah, and T. E. Golde, Sortilin, SorCS1b, and 681 SorLA Vps10p sorting receptors, are novel ?-secretase substrates, Mol. Neurodegener, vol.1, p.3, 2006.

A. Nykjaer, R. Lee, K. K. Teng, P. Jansen, P. Madsen et al., , p.683

M. Schwarz, E. Willnow, and T. E. , Sortilin is essential for proNGF-induced neuronal cell 684 death, Nature, vol.427, pp.843-848, 2004.

K. Offe, S. E. Dodson, J. T. Shoemaker, J. J. Fritz, M. Gearing et al., , 2006.

, The lipoprotein receptor LR11 regulates amyloid beta production and amyloid precursor protein 687 traffic in endosomal compartments, J. Neurosci. Off. J. Soc. Neurosci, vol.26, pp.1596-1603

K. Ohwaki, H. Bujo, M. Jiang, H. Yamazaki, W. J. Schneider et al., A Secreted 689 Soluble Form of LR11, Specifically Expressed in Intimal Smooth Muscle Cells, Accelerates 690 Formation of Lipid-Laden Macrophages, Arterioscler. Thromb. Vasc. Biol, vol.27, pp.1050-1056, 2007.

, Licence CC BY

, Regulatory roles of Sortilin and SorLA in immune-related processes 21

X. Pan, N. Zaarur, M. Singh, P. Morin, and K. V. Kandror, Sortilin and retromer mediate 692 retrograde transport of Glut4 in 3T3-L1 adipocytes, Mol. Biol. Cell, vol.28, pp.1667-1675, 2017.

A. B. Patel, I. Tsilioni, S. E. Leeman, and T. C. Theoharides, Neurotensin stimulates sortilin 694 and mTOR in human microglia inhibitable by methoxyluteolin, a potential therapeutic target for 695 autism, Proc. Natl. Acad. Sci. U. S. A, 2016.

K. M. Patel, A. Strong, J. Tohyama, X. Jin, C. R. Morales et al., , p.697

D. J. Rader, Macrophage Sortilin Promotes LDL Uptake, Foam Cell Formation, and 698 Atherosclerosis, Circ. Res, vol.116, pp.789-796, 2015.

D. H. Paushter, H. Du, T. Feng, and F. Hu, The lysosomal function of progranulin, a 700 guardian against neurodegeneration, Acta Neuropathol. (Berl.), vol.136, pp.1-17, 2018.

C. M. Petersen, M. S. Nielsen, A. Nykjaer, L. Jacobsen, N. Tommerup et al., , p.702

H. Roigaard, J. Gliemann, P. Madsen, and S. K. Moestrup, Molecular identification of a 703 novel candidate sorting receptor purified from human brain by receptor-associated protein affinity 704 chromatography, J. Biol. Chem, vol.272, pp.3599-3605, 1997.

J. Pirault, K. A. Polyzos, M. H. Petri, D. F. Ketelhuth, M. Bäck et al., The 706 inflammatory cytokine interferon-gamma inhibits sortilin-1 expression in hepatocytes via the 707 JAK/STAT pathway, Eur. J. Immunol, vol.47, pp.1918-1924, 2017.

E. M. Quistgaard, P. Madsen, M. K. Grøftehauge, P. Nissen, C. M. Petersen et al., Ligands bind to Sortilin in the tunnel of a ten-bladed ?-propeller domain, Nat. Struct. Mol, p.709, 2009.

, Biol, vol.16, pp.96-98

E. Reuter, J. Weber, M. Paterka, R. Ploen, T. Breiderhoff et al., Role of Sortilin in Models of Autoimmune Neuroinflammation, J. Immunol, vol.713, pp.5762-5769, 2015.

S. Rhost, É. Hughes, H. Harrison, S. Rafnsdottir, H. Jacobsson et al., , p.715

P. Fitzpatrick, D. Andersson, and K. Berger, Sortilin inhibition limits secretion-induced 716 progranulin-dependent breast cancer progression and cancer stem cell expansion, Breast Cancer Res, vol.717, issue.20, p.137, 2018.

M. Rohe, M. Synowitz, R. Glass, S. M. Paul, A. Nykjaer et al., , 2009.

, Derived Neurotrophic Factor Reduces Amyloidogenic Processing through Control of SORLA Gene 720 Expression, J. Neurosci, vol.29, pp.15472-15478

M. Rohe, D. Hartl, A. N. Fjorback, J. Klose, and T. E. Willnow, SORLA-Mediated 722 Trafficking of TrkB Enhances the Response of Neurons to BDNF, PLoS ONE, vol.8, 2013.

S. Rose-john, Interleukin-6 Family Cytokines, Cold Spring Harb. Perspect. Biol, vol.10, p.28415, 2018.

S. Roselli, J. Pundavela, Y. Demont, S. Faulkner, S. Keene et al., , p.726

M. M. Walker and H. Hondermarck, Sortilin is associated with breast cancer aggressiveness 727 and contributes to tumor cell adhesion and invasion, Oncotarget, vol.6, pp.10473-10486, 2015.

C. M. Rosenberger, A. E. Clark, P. M. Treuting, C. D. Johnson, A. et al., ATF3 729 regulates MCMV infection in mice by modulating IFN-? expression in natural killer cells, Proc. Natl, 2008.

. Acad and . Sci, , vol.105, pp.2544-2549

I. Ross, A bird's-eye view of macrophage biology, Lentiviruses and Macrophages: Licence CC, 2010.

, Regulatory roles of Sortilin and SorLA in immune-related processes 22

S. Sakai, C. Nakaseko, M. Takeuchi, C. Ohwada, N. Shimizu et al., , p.734

M. Jiang, Y. Sato, and H. Ebinuma, Circulating soluble LR11/SorLA levels are highly 735 increased and ameliorated by chemotherapy in acute leukemias, Clin. Chim. Acta, vol.413, pp.1542-1548, 2012.

J. Scheller, A. Chalaris, D. Schmidt-arras, R. , and S. , The pro-and anti-737 inflammatory properties of the cytokine interleukin-6, Biochim. Biophys. Acta, vol.1813, pp.878-888, 2011.

V. Schmidt and T. E. Willnow, Protein sorting gone wrong -VPS10P domain receptors in 739 cardiovascular and metabolic diseases, Atherosclerosis, vol.245, pp.194-199, 2016.

V. Schmidt, N. Schulz, X. Yan, A. Schürmann, S. Kempa et al., , p.741

G. Olivecrona and T. E. Willnow, SORLA facilitates insulin receptor signaling in adipocytes 742 and exacerbates obesity, J. Clin. Invest, vol.126, pp.2706-2720, 2016.

V. Schmidt, A. Subkhangulova, and T. E. Willnow, Sorting receptor SORLA: cellular 744 mechanisms and implications for disease, Cell. Mol. Life Sci, vol.74, pp.1475-1483, 2017.

H. Schuett, R. Oestreich, G. H. Waetzig, W. Annema, M. Luchtefeld et al., Transsignaling of interleukin-6 crucially 747 contributes to atherosclerosis in mice, Arterioscler. Thromb. Vasc. Biol, vol.32, pp.281-290, 2012.

M. N. Seaman, Identification of a novel conserved sorting motif required for retromer-749 mediated endosome-to-TGN retrieval, J Cell Sci, vol.120, pp.2378-2389, 2007.

S. Shalapour, K. , and M. , Immunity, inflammation, and cancer: an eternal fight between 751 good and evil, J. Clin. Invest, vol.125, pp.3347-3355, 2015.

J. Shi and K. V. Kandror, Sortilin is essential and sufficient for the formation of Glut4 753 storage vesicles in 3T3-L1 adipocytes, Dev. Cell, vol.9, pp.99-108, 2005.

A. Strong, Q. Ding, A. C. Edmondson, J. S. Millar, K. V. Sachs et al., , p.755

M. Y. Ai, D. Guo, and L. , Hepatic sortilin regulates both apolipoprotein B secretion and 756 LDL catabolism, J. Clin. Invest, vol.122, pp.2807-2816, 2012.

T. Suda, K. Takubo, and G. L. Semenza, Metabolic regulation of hematopoietic stem cells in 758 the hypoxic niche, Cell Stem Cell, vol.9, pp.298-310, 2011.

H. Y. Sung, J. Y. Lee, A. K. Park, Y. J. Moon, I. Jo et al., Aberrant Promoter Hypomethylation of Sortilin 1: A Moyamoya Disease, p.761, 2018.

. Biomarker, Aberrant Promoter Hypomethylation of Sortilin 1: A Moyamoya Disease Biomarker

, Stroke J. Stroke, vol.20, pp.350-361

W. Tang, Y. Lu, Q. Tian, Y. Zhang, F. Guo et al., , p.764

L. Kong, The growth factor progranulin binds to TNF receptors and is therapeutic 765 against inflammatory arthritis in mice, Science, vol.332, pp.478-484, 2011.

R. Tanimoto, C. Palladino, S. Xu, S. Buraschi, T. Neill et al., The perlecan-interacting growth factor progranulin 768 regulates ubiquitination, sorting, and lysosomal degradation of sortilin, Matrix Biol. J. Int. Soc. 769 Matrix Biol, vol.64, pp.27-39, 2017.

H. K. Teng, K. K. Teng, R. Lee, S. Wright, S. Tevar et al., , p.771

Z. Chen and F. S. Lee, ProBDNF induces neuronal apoptosis via activation of a 772 receptor complex of p75NTR and sortilin, J. Neurosci. Off. J. Soc. Neurosci, vol.25, pp.5455-5463, 2005.

M. Tjwa, N. Sidenius, R. Moura, S. Jansen, K. Theunissen et al., , p.774

M. Moons, L. Blasi, and F. , Membrane-anchored uPAR regulates the proliferation, marrow Licence CC, 2009.

, Regulatory roles of Sortilin and SorLA in immune-related processes 23 pool size, engraftment, and mobilization of mouse hematopoietic stem/progenitor cells, J. Clin. 776 Invest, vol.119, pp.1008-1018

A. Trumpp, M. Essers, W. , and A. , Awakening dormant haematopoietic stem cells, 2010.

, Rev. Immunol, vol.10, pp.201-209

M. D. Turner, B. Nedjai, T. Hurst, and D. J. Pennington, Cytokines and chemokines: At the 780 crossroads of cell signalling and inflammatory disease, Biochim. Biophys. Acta BBA -Mol. Cell 781 Res, vol.1843, pp.2563-2582, 2014.

D. Tzachanis, A. Berezovskaya, L. M. Nadler, and V. A. Boussiotis, Blockade of B7/CD28 783 in mixed lymphocyte reaction cultures results in the generation of alternatively activated 784 macrophages, which suppress T-cell responses, Blood, vol.99, pp.1465-1473, 2002.

C. L. Vázquez, A. Rodgers, S. Herbst, S. Coade, A. Gronow et al., , p.786

M. Kanzaki, A. Nykjaer, and M. G. Gutierrez, The proneurotrophin receptor sortilin is 787 required for Mycobacterium tuberculosis control by macrophages, Sci. Rep, vol.6, 2016.

A. Wahe, B. Kasmapour, C. Schmaderer, D. Liebl, K. Sandhoff et al., Golgi-to-phagosome transport of acid sphingomyelinase and prosaposin is 790 mediated by sortilin, J. Cell Sci, vol.123, pp.2502-2511, 2010.

A. J. Whittle, M. Jiang, V. Peirce, J. Relat, S. Virtue et al., , p.792

M. Takahashi and T. Murano, Soluble LR11/SorLA represses thermogenesis in adipose 793 tissue and correlates with BMI in humans, Nat. Commun, vol.6, p.8951, 2015.

C. M. Wilson, T. Naves, F. Vincent, B. Melloni, F. Bonnaud et al., Sortilin mediates the release and transfer of exosomes in concert with two tyrosine kinase 796 receptors, J. Cell Sci, vol.795, pp.3983-3997, 2014.

C. M. Wilson, T. Naves, S. Saada, S. Pinet, F. Vincent et al., The implications of sortilin/vps10p domain receptors in neurological and human diseases, 799 CNS Neurol. Disord. Drug Targets, vol.798, pp.1354-1365, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01827711

C. M. Wilson, T. Naves, H. Al-akhrass, F. Vincent, B. Melloni et al., A new role under sortilin's belt in cancer, Commun. Integr. Biol, vol.9, p.1130192, 2016.

T. Yabe-wada, S. Matsuba, K. Takeda, T. Sato, M. Suyama et al., , p.804

C. C. Philpott and A. Nakamura, TLR signals posttranscriptionally regulate the cytokine 805 trafficking mediator sortilin, Sci. Rep, vol.6, 2016.

T. Yabe-wada, S. Matsuba, M. Unno, and N. Onai, Crystal structure of the ligand-free 807 form of the Vps10 ectodomain of dimerized Sortilin at acidic pH, FEBS Lett, vol.592, pp.2647-2657, 2018.

M. Yang, Y. Lim, X. Li, J. Zhong, and X. Zhou, Precursor of Brain-derived 809 Neurotrophic Factor (proBDNF) Forms a Complex with Huntingtin-associated Protein-1 (HAP1), p.810, 2011.

, Sortilin That Modulates proBDNF Trafficking, Degradation, and Processing, J. Biol. Chem, vol.286, pp.16272-16284

X. Yu, Y. Fu, D. Zhang, K. Yin, and C. Tang, , 2013.

, Clin. Chim. Acta Int. J. Clin. Chem, vol.424, pp.245-252

Y. Yu, P. H. Correll, and J. P. Heuvel, Conjugated linoleic acid decreases production of pro-inf 815 lammatory products in macrophages: evidence for a PPARg-dependent mechanism

, Biophys. Acta, vol.11