R. M. Locksley, N. Killeen, M. J. Lenardo, . The, and . Superfamilies, Integrating Mammalian Biology. Cell, vol.104, pp.487-501, 2001.

G. A. Granger, S. J. Shacks, T. W. Williams, and W. P. Kolb, Lymphocyte in vitro Cytotoxicity: Specific Release of Lymphotoxin-like Materials from Tuberculin-sensitive Lymphoid Cells, Nature, vol.221, pp.1155-1157, 1969.

E. A. Carswell, L. J. Old, R. L. Kassel, S. Green, N. Fiore et al., An endotoxin-induced serum factor that causes necrosis of tumors, Proc Natl Acad Sci, vol.72, pp.3666-3670, 1975.

P. W. Gray, B. B. Aggarwal, C. V. Benton, T. S. Bringman, W. J. Henzel et al., Cloning and expression of cDNA for human lymphotoxin, a lymphokine with tumour necrosis activity, Nature, vol.312, pp.721-724, 1984.

D. Pennica, G. E. Nedwin, J. S. Hayflick, P. H. Seeburg, R. Derynck et al., Human tumour necrosis factor: precursor structure, expression and homology to lymphotoxin, Nature, vol.312, pp.724-729, 1984.

J. Bodmer, P. Schneider, and J. Tschopp, The molecular architecture of the TNF superfamily, Trends Biochem Sci, vol.27, pp.19-26, 2002.

G. Zhang, Tumor necrosis factor family ligand-receptor binding, Curr Opin Struct Biol, vol.14, pp.154-160, 2004.

B. B. Aggarwal, Signalling pathways of the TNF superfamily: a double-edged sword, Nat Rev Immunol, vol.3, pp.745-756, 2003.

M. Croft, C. A. Benedict, and C. F. Ware, Clinical targeting of the TNF and TNFR superfamilies, Nat Rev Drug Discov, vol.12, pp.147-168, 2013.

W. S. Simonet, D. L. Lacey, C. R. Dunstan, M. Kelley, M. Chang et al., Osteoprotegerin: A Novel Secreted Protein Involved in the Regulation of Bone Density, Cell, vol.89, pp.309-319, 1997.

E. Tsuda, M. Goto, S. Mochizuki, K. Yano, F. Kobayashi et al., Isolation of a Novel Cytokine from Human Fibroblasts That Specifically Inhibits Osteoclastogenesis, Biochem Biophys Res Commun, vol.234, pp.137-142, 1997.

H. Yasuda, N. Shima, N. Nakagawa, K. Yamaguchi, M. Kinosaki et al., Osteoclast differentiation factor is a ligand for osteoprotegerin/osteoclastogenesis-inhibitory factor and is identical to TRANCE/RANKL, Proc Natl Acad Sci U S A, vol.95, pp.3597-3602, 1998.

D. L. Lacey, E. Timms, H. Tan, M. J. Kelley, C. R. Dunstan et al., Osteoprotegerin Ligand Is a Cytokine that Regulates Osteoclast Differentiation and Activation, Cell, vol.93, pp.165-176, 1998.

D. M. Anderson, E. Maraskovsky, W. L. Billingsley, W. C. Dougall, M. E. Tometsko et al., A homologue of the TNF receptor and its ligand enhance T-cell growth and dendritic-cell function, Nature, vol.390, pp.175-179, 1997.

, The RANK/RANKL/OPG triad

B. R. Wong, R. Josien, S. Y. Lee, B. Sauter, H. Li et al., TRANCE (Tumor Necrosis Factor [TNF]-related Activation-induced Cytokine), a New TNF Family Member Predominantly Expressed in T cells, Is a Dendritic Cell-specific Survival Factor, J Exp Med, vol.186, pp.2075-2080, 1997.

H. Yasuda, N. Shima, N. Nakagawa, S. Mochizuki, K. Yano et al., Identity of Osteoclastogenesis Inhibitory Factor (OCIF) and Osteoprotegerin (OPG): A Mechanism by which OPG/OCIF Inhibits Osteoclastogenesis in Vitro, Endocrinology, vol.139, pp.1329-1337, 1998.

N. Nakagawa, M. Kinosaki, K. Yamaguchi, N. Shima, H. Yasuda et al., RANK Is the Essential Signaling Receptor for Osteoclast Differentiation Factor in Osteoclastogenesis, Biochem Biophys Res Commun, vol.253, pp.395-400, 1998.

H. Hsu, D. L. Lacey, C. R. Dunstan, I. Solovyev, A. Colombero et al., Tumor necrosis factor receptor family member RANK mediates osteoclast differentiation and activation induced by osteoprotegerin ligand, Proc Natl Acad Sci, vol.96, pp.3540-3545, 1999.

J. G. Emery, P. Mcdonnell, M. B. Burke, K. C. Deen, S. Lyn et al., Osteoprotegerin Is a Receptor for the Cytotoxic Ligand TRAIL, J Biol Chem, vol.273, pp.14363-14367, 1998.

L. Lum, B. R. Wong, R. Josien, J. D. Becherer, H. Erdjument-bromage et al., Evidence for a Role of a Tumor Necrosis Factor-? (TNF-?)-converting Enzyme-like Protease in Shedding of TRANCE, a TNF Family Member Involved in Osteoclastogenesis and Dendritic Cell Survival, J Biol Chem, vol.274, pp.13613-13618, 1999.

A. Hikita, I. Yana, H. Wakeyama, M. Nakamura, Y. Kadono et al., Negative Regulation of Osteoclastogenesis by Ectodomain Shedding of Receptor Activator of NF-?B Ligand, J Biol Chem, vol.281, pp.36846-36855, 2006.

C. C. Lynch, A. Hikosaka, H. B. Acuff, M. D. Martin, N. Kawai et al., MMP-7 promotes prostate cancer-induced osteolysis via the solubilization of RANKL, Cancer Cell, vol.7, pp.485-496, 2005.

J. Lam, C. A. Nelson, F. P. Ross, S. L. Teitelbaum, and D. H. Fremont, Crystal structure of the TRANCE/RANKL cytokine reveals determinants of receptor-ligand specificity, J Clin Invest, vol.108, pp.971-979, 2001.

C. A. Nelson, J. T. Warren, M. Wang, S. L. Teitelbaum, and D. H. Fremont, RANKL employs distinct binding modes to engage RANK and the osteoprotegerin decoy receptor. Struct Lond Engl, vol.20, pp.1971-1982, 1993.

T. Ikeda, M. Kasai, M. Utsuyama, and K. Hirokawa, Determination of Three Isoforms of the Receptor Activator of Nuclear Factor-?? Ligand and Their Differential Expression in Bone and Thymus, Endocrinology, vol.142, pp.1419-1426, 2001.

V. Kartsogiannis, H. Zhou, N. J. Horwood, R. J. Thomas, D. K. Hards et al., Localization of RANKL (receptor activator of NF?B ligand) mRNA and protein in skeletal and extraskeletal tissues, Bone, vol.25, pp.525-534, 1999.

T. Wada, T. Nakashima, N. Hiroshi, and J. M. Penninger, RANKL-RANK signaling in osteoclastogenesis and bone disease, Trends Mol Med, vol.12, pp.17-25, 2006.

, The RANK/RANKL/OPG triad

D. J. Hadjidakis, I. I. Androulakis, and . Bone-remodeling, Ann N Y Acad Sci, vol.1092, pp.385-396, 2006.

E. Jimi, S. Hirata, K. Osawa, M. Terashita, C. Kitamura et al., The Current and Future Therapies of Bone Regeneration to Repair Bone Defects, Int J Dent, vol.2012, p.148261, 2012.

D. L. Lacey, E. Timms, H. Tan, M. J. Kelley, C. R. Dunstan et al., Osteoprotegerin Ligand Is a Cytokine that Regulates Osteoclast Differentiation and Activation, Cell, vol.93, pp.165-176, 1998.

M. C. Walsh and Y. Choi, Biology of the RANKL-RANK-OPG system in immunity, bone, and beyond, Inflammation, vol.5, p.511, 2014.

L. E. Theill, W. J. Boyle, and J. M. Penninger, RANK-L AND RANK: T Cells, Bone Loss, and Mammalian Evolution, Annu Rev Immunol, vol.20, pp.795-823, 2002.

W. C. Dougall, M. Glaccum, K. Charrier, K. Rohrbach, K. Brasel et al., RANK is essential for osteoclast and lymph node development, Genes Dev, vol.13, pp.2412-2424, 1999.

Y. Kong, H. Yoshida, I. Sarosi, H. Tan, E. Timms et al., OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis, Nature, vol.397, pp.315-323, 1999.

N. Bucay, I. Sarosi, C. R. Dunstan, S. Morony, J. Tarpley et al., osteoprotegerin-deficient mice develop early onset osteoporosis and arterial calcification, Genes Dev, vol.12, pp.1260-1268, 1998.

A. Mizuno, N. Amizuka, K. Irie, A. Murakami, N. Fujise et al., Severe Osteoporosis in Mice Lacking Osteoclastogenesis Inhibitory Factor/Osteoprotegerin, Biochem Biophys Res Commun, vol.247, pp.610-615, 1998.

T. Nakashima, Y. Kobayashi, S. Yamasaki, A. Kawakami, K. Eguchi et al., Protein Expression and Functional Difference of Membrane-Bound and Soluble Receptor Activator of NF-?B Ligand: Modulation of the Expression by Osteotropic Factors and Cytokines, Biochem Biophys Res Commun, vol.275, pp.768-775, 2000.

H. Kitaura, K. Kimura, M. Ishida, H. Kohara, M. Yoshimatsu et al., Immunological Reaction in TNF-α-Mediated Osteoclast Formation and Bone Resorption In Vitro and In Vivo, J Immunol Res, 2013.

M. A. Lomaga, W. Yeh, I. Sarosi, G. S. Duncan, C. Furlonger et al., TRAF6 deficiency results in osteopetrosis and defective interleukin-1, CD40, and LPS signaling, Genes Dev, vol.13, pp.1015-1024, 1999.

G. Franzoso, L. Carlson, L. Xing, L. Poljak, E. W. Shores et al., Requirement for NF-?B in osteoclast and B-cell development, Genes Dev, vol.11, p.3482, 1997.

E. Dejardin, The alternative NF-?B pathway from biochemistry to biology: Pitfalls and promises for future drug development, Biochem Pharmacol, vol.72, pp.1161-1179, 2006.

R. Zeng, R. Faccio, and D. V. Novack, Alternative NF-?B Regulates RANKL-Induced Osteoclast Differentiation and Mitochondrial Biogenesis via Independent Mechanisms, J Bone Miner Res, vol.30, pp.2287-2299, 2015.

J. David, K. Sabapathy, O. Hoffmann, M. H. Idarraga, and E. F. Wagner, JNK1 modulates osteoclastogenesis through both c-Jun phosphorylation-dependent and -independent mechanisms, J Cell Sci, vol.115, pp.4317-4325, 2002.

B. R. Wong, D. Besser, N. Kim, J. R. Arron, M. Vologodskaia et al., TNF Family Member, Activates Akt/PKB through a Signaling Complex Involving TRAF6 and c-Src, vol.4, pp.1041-1049, 1999.

, Chapter 2: Biology of the RANK/RANKL/OPG triad

J. C. Crockett, D. J. Mellis, D. I. Scott, and M. H. Helfrich, New knowledge on critical osteoclast formation and activation pathways from study of rare genetic diseases of osteoclasts: focus on the RANK/RANKL axis, Osteoporos Int, vol.22, pp.1-20, 2010.

T. J. Yun, M. D. Tallquist, A. Aicher, K. L. Rafferty, A. J. Marshall et al., Crucial Regulator of Bone Metabolism, Also Regulates B Cell Development and Function, J Immunol, vol.166, pp.1482-1491, 2001.

T. J. Yun, P. M. Chaudhary, G. L. Shu, J. K. Frazer, M. K. Ewings et al., OPG/FDCR-1, a TNF Receptor Family Member, Is Expressed in Lymphoid Cells and Is Up-Regulated by Ligating CD40, J Immunol, vol.161, pp.6113-6121, 1998.

T. Perlot and J. M. Penninger, Development and Function of Murine B Cells Lacking RANK, J Immunol, vol.188, pp.1201-1205, 2012.

N. A. Roberts, A. J. White, W. E. Jenkinson, G. Turchinovich, K. Nakamura et al., Rank Signaling Links the Development of Invariant ?? T Cell Progenitors and Aire+ Medullary Epithelium, Immunity, vol.36, pp.427-437, 2012.

B. R. Wong, R. Josien, S. Y. Lee, B. Sauter, H. Li et al., TRANCE (Tumor Necrosis Factor [TNF]-related Activation-induced Cytokine), a New TNF Family Member Predominantly Expressed in T cells, Is a Dendritic Cell-specific Survival Factor, J Exp Med, vol.186, pp.2075-2080, 1997.

E. Williamson, J. M. Bilsborough, and J. L. Viney, Regulation of Mucosal Dendritic Cell Function by Receptor Activator of NF-?B (RANK)/RANK Ligand Interactions: Impact on Tolerance Induction, J Immunol, vol.169, pp.3606-3612, 2002.

R. Josien, B. R. Wong, H. Li, R. M. Steinman, Y. Choi et al., Is Differentially Expressed on T Cell Subsets and Induces Cytokine Production in Dendritic Cells, J Immunol, vol.162, pp.2562-2568, 1999.

L. Zhong, A. Granelli-piperno, Y. Choi, and R. M. Steinman, Recombinant adenovirus is an efficient and non-perturbing genetic vector for human dendritic cells, Eur J Immunol, vol.29, pp.964-972, 1999.

R. Josien, H. Li, E. Ingulli, S. Sarma, R. Wong et al., Trance, a Tumor Necrosis Factor Family Member, Enhances the Longevity and Adjuvant Properties of Dendritic Cells in Vivo, J Exp Med, vol.191, pp.495-502, 2000.

M. F. Bachmann, B. R. Wong, R. Josien, R. M. Steinman, A. Oxenius et al., TRANCE, a Tumor Necrosis Factor Family Member Critical for CD40 Ligand-independent T Helper Cell Activation, J Exp Med, vol.189, pp.1025-1031, 1999.

J. Barbaroux, M. Beleut, C. Brisken, C. G. Mueller, and R. W. Groves, Epidermal Receptor Activator of NF-?B Ligand Controls Langerhans Cells Numbers and Proliferation, J Immunol, vol.181, pp.1103-1108, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00300531

M. Stolina, D. Dwyer, M. S. Ominsky, T. Corbin, G. Van et al., Continuous RANKL Inhibition in Osteoprotegerin Transgenic Mice and Rats Suppresses Bone Resorption without Impairing Lymphorganogenesis or Functional Immune Responses, J Immunol, vol.179, pp.7497-7505, 2007.

, Chapter 2: Biology of the RANK/RANKL/OPG triad

S. W. Rossi, M. Kim, A. Leibbrandt, S. M. Parnell, W. E. Jenkinson et al., RANK signals from CD4+3? inducer cells regulate development of Aire-expressing epithelial cells in the thymic medulla, J Exp Med, vol.204, pp.1267-1272, 2007.

T. Akiyama, S. Maeda, S. Yamane, K. Ogino, M. Kasai et al., Dependence of Self-Tolerance on TRAF6-Directed Development of Thymic Stroma, Science, vol.308, pp.248-251, 2005.

L. Burkly, C. Hession, L. Ogata, C. Reilly, L. A. Marconl et al., Expression of relB is required for the development of thymic medulla and dendritic cells, Nature, vol.373, pp.531-536, 1995.

F. Weih, D. Carrasco, S. K. Durham, D. S. Barton, C. A. Rizzo et al., Multiorgan inflammation and hematopoietic abnormalities in mice with a targeted disruption of RelB, a member of the NF-?B/Rel family, Cell, vol.80, pp.90416-90422, 1995.

D. C. Otero, D. P. Baker, and M. David, IRF7-Dependent IFN-? Production in Response to RANKL Promotes Medullary Thymic Epithelial Cell Development, J Immunol, vol.190, pp.3289-3298, 2013.

S. Sakaguchi, Naturally arising Foxp3-expressing CD25+CD4+ regulatory T cells in immunological tolerance to self and non-self, Nat Immunol, vol.6, pp.345-352, 2005.

K. Loser, A. Mehling, S. Loeser, J. Apelt, A. Kuhn et al., Epidermal RANKL controls regulatory T-cell numbers via activation of dendritic cells, Nat Med, vol.12, pp.1372-1379, 2006.

E. A. Green, Y. Choi, and R. A. Flavell, Pancreatic Lymph Node-Derived CD4+CD25+ Treg Cells: Highly Potent Regulators of Diabetes that Require TRANCE-RANK Signals, Immunity, vol.16, pp.183-191, 2002.

T. Totsuka, T. Kanai, Y. Nemoto, T. Tomita, R. Okamoto et al., RANK-RANKL Signaling Pathway Is Critically Involved in the Function of CD4+CD25+ Regulatory T Cells in Chronic Colitis, J Immunol, vol.182, pp.6079-6087, 2009.

M. L. Cheng and L. Fong, Effects of RANKL-Targeted Therapy in Immunity and Cancer, Front Oncol, vol.3, 2014.

E. M. Gravallese, C. Manning, A. Tsay, A. Naito, C. Pan et al., Synovial tissue in rheumatoid arthritis is a source of osteoclast differentiation factor, Arthritis Rheum, vol.43, pp.250-258, 2000.

H. Takayanagi, H. Iizuka, T. Juji, T. Nakagawa, A. Yamamoto et al., Involvement of receptor activator of nuclear factor ?B ligand/osteoclast differentiation factor in osteoclastogenesis from synoviocytes in rheumatoid arthritis, Arthritis Rheum, vol.43, pp.259-269, 2000.

K. Okamoto and H. Takayanagi, Regulation of bone by the adaptive immune system in arthritis, Arthritis Res Ther, vol.13, p.219, 2011.

J. Kikuta, Y. Wada, T. Kowada, Z. Wang, G. Sun-wada et al., Dynamic visualization of RANKL and Th17-mediated osteoclast function, J Clin Invest, vol.123, pp.866-873, 2013.

, Chapter 2: Biology of the RANK/RANKL/OPG triad

R. Zhao, N. Chen, X. Zhou, P. Miao, C. Hu et al., Exogenous IFN-beta regulates the RANKL-c-Fos-IFN-beta signaling pathway in the collagen antibody-induced arthritis model, J Transl Med, vol.12, 2014.

M. Onal, J. Xiong, X. Chen, J. D. Thostenson, M. Almeida et al., Receptor Activator of Nuclear Factor ?B Ligand (RANKL) Protein Expression by B Lymphocytes Contributes to Ovariectomy-induced Bone Loss, J Biol Chem, vol.287, pp.29851-29860, 2012.

L. Yeo, H. Lom, M. Juarez, M. Snow, C. D. Buckley et al., Expression of FcRL4 defines a proinflammatory, RANKL-producing B cell subset in rheumatoid arthritis, Ann Rheum Dis, vol.74, pp.928-935, 2015.

L. Yeo, K. Toellner, M. Salmon, A. Filer, C. D. Buckley et al., Cytokine mRNA profiling identifies B cells as a major source of RANKL in rheumatoid arthritis, Ann Rheum Dis, vol.70, pp.2022-2028, 2011.

J. E. Fata, Y. Kong, J. Li, T. Sasaki, J. Irie-sasaki et al., The Osteoclast Differentiation Factor Osteoprotegerin-Ligand Is Essential for Mammary Gland Development, Cell, vol.103, pp.41-50, 2000.

N. Kim, H. Kim, K. Kwon, M. Kim, Y. Cho et al., Receptor Activator of NF-?B Ligand Regulates the Proliferation of Mammary Epithelial Cells via Id2, Mol Cell Biol, vol.26, pp.1002-1013, 2006.

Y. Cao, G. Bonizzi, T. N. Seagroves, F. R. Greten, R. Johnson et al., IKK? Provides an Essential Link between RANK Signaling and Cyclin D1 Expression during Mammary Gland Development, Cell, vol.107, pp.763-775, 2001.

F. Toberer, J. Sykora, D. Göttel, V. Ruland, W. Hartschuh et al., Tissue microarray analysis of RANKL in cutaneous lupus erythematosus and psoriasis, Exp Dermatol, vol.20, pp.600-602, 2011.

V. Duheron, E. Hess, M. Duval, M. Decossas, B. Castaneda et al., Receptor activator of NF-?B (RANK) stimulates the proliferation of epithelial cells of the epidermo-pilosebaceous unit, Proc Natl Acad Sci U S A, vol.108, pp.5342-5347, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00579674

N. A. Mabbott, D. S. Donaldson, H. Ohno, I. R. Williams, and M. A. Microfold, M) cells: important immunosurveillance posts in the intestinal epithelium, Mucosal Immunol, vol.6, pp.666-677, 2013.

R. T. Taylor, S. R. Patel, L. E. Butler, B. R. Lake, J. G. Newberry et al., Lymphotoxin-Independent Expression of TNF-Related Activation-Induced Cytokine by Stromal Cells in Cryptopatches, Isolated Lymphoid Follicles, and Peyer's Patches, J Immunol, vol.178, pp.5659-5667, 2007.

K. A. Knoop, B. R. Butler, N. Kumar, R. D. Newberry, and I. R. Williams, Distinct Developmental Requirements for Isolated Lymphoid Follicle Formation in the Small and Large Intestine: RANKL Is Essential Only in the Small Intestine, Am J Pathol, vol.179, pp.1861-1871, 2011.

. Lau-w-de, P. Kujala, K. Schneeberger, S. Middendorp, V. Li et al., Peyer's Patch M Cells Derived from Lgr5+ Stem Cells Require SpiB and Are Induced by RankL in Cultured "Miniguts, Mol Cell Biol, vol.32, pp.3639-3647, 2012.

, Biology of the RANK/RANKL/OPG triad 85. figure M cells, 2017.

Y. Kim, Y. Kim, Y. M. Lee, H. Kim, J. D. Kim et al., TNF-related Activation-induced Cytokine (TRANCE) Induces Angiogenesis through the Activation of Src and Phospholipase C (PLC) in Human Endothelial Cells, J Biol Chem, vol.277, pp.6799-6805, 2002.

C. and P. , Regulation of Vascular Calcification by Osteoclast Regulatory Factors RANKL and Osteoprotegerin, Circ Res, vol.95, pp.1046-1057, 2004.

H. Kim, H. S. Shin, H. J. Kwak, K. Y. Ahn, J. Kim et al., RANKL regulates endothelial cell survival through the phosphatidylinositol 3?-kinase/Akt signal transduction pathway, FASEB J, 2003.

L. B. Pritzker, M. Scatena, and C. M. Giachelli, The Role of Osteoprotegerin and Tumor Necrosis Factorrelated Apoptosis-inducing Ligand in Human Microvascular Endothelial Cell Survival, Mol Biol Cell, vol.15, pp.2834-2841, 2004.

U. M. Malyankar, M. Scatena, K. L. Suchland, T. J. Yun, E. A. Clark et al., Osteoprotegerin Is an ?v?3-induced, NF-?B-dependent Survival Factor for Endothelial Cells, J Biol Chem, vol.275, pp.20959-20962, 2000.

M. Kobayashi-sakamoto, E. Isogai, K. Hirose, and I. Chiba, Role of ?v integrin in osteoprotegerininduced endothelial cell migration and proliferation, Microvasc Res, vol.76, pp.139-144, 2008.

J. Min, Y. Kim, S. W. Kim, M. Kwon, Y. Kong et al., TNF-Related Activation-Induced Cytokine Enhances Leukocyte Adhesiveness: Induction of ICAM-1 and VCAM-1 via TNF ReceptorAssociated Factor and Protein Kinase C-Dependent NF-?B Activation in Endothelial Cells, J Immunol, vol.175, pp.531-540, 2005.

E. González-suárez and A. Sanz-moreno, RANK as a therapeutic target in cancer, FEBS J, vol.283, pp.2018-2033, 2016.

D. Schramek, A. Leibbrandt, V. Sigl, L. Kenner, J. A. Pospisilik et al., Osteoclast differentiation factor RANKL controls development of progestin-driven mammary cancer, Nature, vol.468, pp.98-102, 2010.

E. Gonzalez-suarez, A. P. Jacob, J. Jones, R. Miller, M. P. Roudier-meyer et al., RANK ligand mediates progestin-induced mammary epithelial proliferation and carcinogenesis, Nature, vol.468, pp.103-107, 2010.

W. Tan, W. Zhang, A. Strasner, S. Grivennikov, J. Q. Cheng et al., Tumour-infiltrating regulatory T cells stimulate mammary cancer metastasis through RANKL-RANK signalling, Nature, vol.470, pp.548-553, 2011.

M. Palafox, I. Ferrer, P. Pellegrini, S. Vila, S. Hernandez-ortega et al., RANK Induces Epithelial-Mesenchymal Transition and Stemness in Human Mammary Epithelial Cells and Promotes Tumorigenesis and Metastasis, Cancer Res, vol.72, pp.2879-2888, 2012.

, Chapter 2: Biology of the RANK/RANKL/OPG triad

W. Zhang, W. Tan, X. Wu, M. Poustovoitov, A. Strasner et al., A NIK-IKK? Module Expands ErbB2-Induced Tumor-Initiating Cells by Stimulating Nuclear Export of p27/Kip1, Cancer Cell, vol.23, pp.647-659, 2013.

K. Mori, L. Goff, B. Berreur, M. Riet, A. Moreau et al., Human osteosarcoma cells express functional receptor activator of nuclear factor-kappa B, J Pathol, vol.211, pp.555-562, 2007.

V. Kupas, C. Weishaupt, D. Siepmann, M. Kaserer, M. Eickelmann et al., RANK Is Expressed in Metastatic Melanoma and Highly Upregulated on Melanoma-Initiating Cells, J Invest Dermatol, vol.131, pp.944-955, 2011.

G. Chen, K. Sircar, A. Aprikian, A. Potti, D. Goltzman et al., Expression of RANKL/RANK/OPG in primary and metastatic human prostate cancer as markers of disease stage and functional regulation, Cancer, vol.107, pp.289-298, 2006.

A. Hameed, J. J. Brady, P. Dowling, M. Clynes, O. 'gorman et al., Bone Disease in Multiple Myeloma: Pathophysiology and Management, Cancer Growth Metastasis, vol.2014, pp.33-42, 2014.

R. J. Thomas, T. A. Guise, J. J. Yin, J. Elliott, N. J. Horwood et al., Breast Cancer Cells Interact with Osteoblasts to Support Osteoclast Formation, Endocrinology, vol.140, pp.4451-4458, 1999.

J. Zhang, J. Dai, Y. Qi, D. Lin, P. Smith et al., Osteoprotegerin inhibits prostate cancer-induced osteoclastogenesis and prevents prostate tumor growth in the bone, J Clin Invest, vol.107, pp.1235-1244, 2001.

A. Lipton and C. Goessl, Clinical development of anti-RANKL therapies for treatment and prevention of bone metastasis, Bone, vol.48, pp.96-99, 2011.

I. S. Khan, M. L. Mouchess, M. Zhu, B. Conley, K. J. Fasano et al., Enhancement of an antitumor immune response by transient blockade of central T cell tolerance, J Exp Med, vol.211, pp.761-768, 2014.

C. Wiethe, K. Dittmar, T. Doan, W. Lindenmaier, and R. Tindle, Enhanced Effector and Memory CTL Responses Generated by Incorporation of Receptor Activator of NF-?B (RANK)/RANK Ligand Costimulatory Molecules into Dendritic Cell Immunogens Expressing a Human Tumor-Specific Antigen, J Immunol, vol.171, pp.4121-4130, 2003.

X. Ma, Y. Liu, Y. Zhang, X. Yu, W. Wang et al., Jolkinolide B inhibits RANKL-induced osteoclastogenesis by suppressing the activation NF-?B and MAPK signaling pathways, Biochem Biophys Res Commun, vol.445, pp.282-288, 2014.

Y. Chen, J. Sun, C. Dou, N. Li, F. Kang et al., Alliin Attenuated RANKL-Induced Osteoclastogenesis by Scavenging Reactive Oxygen Species through Inhibiting Nox1, Int J Mol Sci, vol.17, 2016.

L. Li, M. Sapkota, S. Kim, and Y. Soh, Herbacetin inhibits RANKL-mediated osteoclastogenesis in vitro and prevents inflammatory bone loss in vivo, Eur J Pharmacol, vol.777, pp.17-25, 2016.

, Biology of the RANK/RANKL/OPG triad inhibiting NF-?B activation and down regulating inflammatory cytokines, Chem Biol Interact, vol.2, pp.467-479, 2013.

X. Cheng, M. Kinosaki, M. Takami, Y. Choi, H. Zhang et al., Disabling of Receptor Activator of Nuclear Factor-?B (RANK) Receptor Complex by Novel Osteoprotegerin-like Peptidomimetics Restores Bone Loss in Vivo, J Biol Chem, vol.279, pp.8269-8277, 2004.

G. Kato, Y. Shimizu, Y. Arai, N. Suzuki, Y. Sugamori et al., The inhibitory effects of a RANKL-binding peptide on articular and periarticular bone loss in a murine model of collageninduced arthritis: a bone histomorphometric study, Arthritis Res Ther, vol.17, p.251, 2015.

D. J. Heath, K. Vanderkerken, X. Cheng, O. Gallagher, M. Prideaux et al., An Osteoprotegerin-like Peptidomimetic Inhibits Osteoclastic Bone Resorption and Osteolytic Bone Disease in Myeloma, Cancer Res, vol.67, pp.202-208, 2007.

J. Hur, A. Ghosh, K. Kim, H. M. Ta, H. Kim et al., Design of a RANK-Mimetic Peptide Inhibitor of Osteoclastogenesis with Enhanced RANKL-Binding Affinity, Mol Cells, vol.39, pp.316-321, 2016.

H. M. Ta, G. Nguyen, H. M. Jin, J. Choi, H. Park et al., Structure-based development of a receptor activator of nuclear factor-?B ligand (RANKL) inhibitor peptide and molecular basis for osteopetrosis, Proc Natl Acad Sci, vol.107, pp.20281-20286, 2010.

K. Aoki, H. Saito, C. Itzstein, M. Ishiguro, T. Shibata et al., A TNF receptor loop peptide mimic blocks RANK ligand-induced signaling, bone resorption, and bone loss, J Clin Invest, vol.116, pp.1525-1534, 2006.

M. Newa, M. Lam, K. H. Bhandari, B. Xu, and M. R. Doschak, Expression, Characterization, and Evaluation of a RANK-Binding Single Chain Fraction Variable: An Osteoclast Targeting Drug Delivery Strategy, Mol Pharm, vol.11, pp.81-89, 2014.

L. L. Green, Antibody engineering via genetic engineering of the mouse: XenoMouse strains are a vehicle for the facile generation of therapeutic human monoclonal antibodies, J Immunol Methods, vol.231, pp.11-23, 1999.

A. E. Kearns, S. Khosla, and P. J. Kostenuik, Receptor Activator of Nuclear Factor ?B Ligand and Osteoprotegerin Regulation of Bone Remodeling in Health and Disease, Endocr Rev, vol.29, pp.155-192, 2008.

P. J. Kostenuik, H. Q. Nguyen, J. Mccabe, K. S. Warmington, C. Kurahara et al., Denosumab, a Fully Human Monoclonal Antibody to RANKL, Inhibits Bone Resorption and Increases BMD in Knock-In Mice That Express Chimeric (Murine/Human) RANKL, J Bone Miner Res, vol.24, pp.182-195, 2009.

E. M. Schwarz and C. T. Ritchlin, Clinical development of anti-RANKL therapy, Arthritis Res Ther, vol.9, p.7, 2007.

M. R. Mcclung, E. M. Lewiecki, S. B. Cohen, M. A. Bolognese, G. C. Woodson et al., Denosumab in Postmenopausal Women with Low Bone Mineral Density, N Engl J Med, vol.354, pp.821-831, 2006.

, Chapter 2: Biology of the RANK/RANKL/OPG triad

G. Asellio, Qua sententiae anatomicae multae, vel perperam receptae convelluntur, vel parum perceptae illustrantur. Lugduni Batavorum, pp.1581-1626

U. H. Von-andrian and T. R. Mempel, Homing and cellular traffic in lymph nodes, Nat Rev Immunol, vol.3, pp.867-878, 2003.

G. Oliver, Lymphatic vasculature development, Nat Rev Immunol, vol.4, pp.35-45, 2004.

G. Jurisic and M. Detmar, Lymphatic endothelium in health and disease, Cell Tissue Res, vol.335, pp.97-108, 2009.

J. Y. Jang, Y. J. Koh, S. Lee, J. Lee, K. H. Kim et al., Conditional ablation of LYVE-1+ cells unveils defensive roles of lymphatic vessels in intestine and lymph nodes, Blood, vol.122, pp.2151-2161, 2013.

L. N. Cueni and M. Detmar, New Insights into the Molecular Control of the Lymphatic Vascular System and its Role in Disease, J Invest Dermatol, vol.126, pp.2167-2177, 2006.

P. Baluk, J. Fuxe, H. Hashizume, T. Romano, E. Lashnits et al., Functionally specialized junctions between endothelial cells of lymphatic vessels, J Exp Med, vol.204, pp.2349-2362, 2007.

A. W. Lund, T. R. Medler, S. A. Leachman, and L. M. Coussens, Lymphatic Vessels, Inflammation, and Immunity in Skin Cancer, Cancer Discov, vol.6, pp.22-35, 2016.

H. Pflicke and M. Sixt, Preformed portals facilitate dendritic cell entry into afferent lymphatic vessels, J Exp Med, vol.206, pp.2925-2935, 2009.

M. Weber, R. Hauschild, J. Schwarz, C. Moussion, . Vries-i-de et al., Interstitial Dendritic Cell Guidance by Haptotactic Chemokine Gradients, Science, vol.339, pp.328-332, 2013.

E. Russo, A. Teijeira, K. Vaahtomeri, A. Willrodt, J. S. Bloch et al., Intralymphatic CCL21 Promotes Tissue Egress of Dendritic Cells through Afferent Lymphatic Vessels, Cell Rep, vol.14, pp.1723-1734, 2016.

S. K. Bromley, S. Yan, M. Tomura, O. Kanagawa, and A. D. Luster, Recirculating Memory T Cells Are a Unique Subset of CD4+ T Cells with a Distinct Phenotype and Migratory Pattern, J Immunol, vol.190, pp.970-976, 2013.

C. Beauvillain, P. Cunin, A. Doni, M. Scotet, S. Jaillon et al., CCR7 is involved in the migration of neutrophils to lymph nodes, Blood, vol.117, pp.1196-1204, 2011.

K. Kabashima, N. Shiraishi, K. Sugita, T. Mori, A. Onoue et al., CXCL12-CXCR4 Engagement Is Required for Migration of Cutaneous Dendritic Cells, Am J Pathol, vol.171, pp.1249-1257, 2007.

H. Takamatsu, N. Takegahara, Y. Nakagawa, M. Tomura, M. Taniguchi et al., Semaphorins guide the entry of dendritic cells into the lymphatics by activating myosin II, Nat Immunol, vol.11, pp.594-600, 2010.

M. H. Ulvmar and T. Mäkinen, Heterogeneity in the lymphatic vascular system and its origin, Cardiovasc Res, vol.111, pp.310-321, 2016.

G. Jurisic and M. Detmar, Lymphatic endothelium in health and disease, Cell Tissue Res, vol.335, pp.97-108, 2009.

E. Bazigou and T. Makinen, Flow control in our vessels: vascular valves make sure there is no way back, Cell Mol Life Sci, vol.70, pp.1055-1066, 2013.

E. Bazigou, S. Xie, C. Chen, A. Weston, N. Miura et al., Integrin-?9 Is Required for Fibronectin Matrix Assembly during Lymphatic Valve Morphogenesis, Dev Cell, vol.17, pp.175-186, 2009.

C. Norrmén, K. I. Ivanov, J. Cheng, N. Zangger, M. Delorenzi et al., FOXC2 controls formation and maturation of lymphatic collecting vessels through cooperation with NFATc1, J Cell Biol, vol.185, pp.439-457, 2009.

J. Kazenwadel, K. L. Betterman, C. Chong, P. H. Stokes, Y. K. Lee et al., GATA2 is required for lymphatic vessel valve development and maintenance, J Clin Invest, vol.125, pp.2979-2994, 2015.

J. T. Wigle and G. Oliver, Prox1 Function Is Required for the Development of the Murine Lymphatic System, Cell, vol.98, pp.769-778, 1999.

J. L. Astarita, S. E. Acton, and S. J. Turley, Podoplanin: emerging functions in development, the immune system, and cancer, Front Immunol, vol.3, 2012.

L. N. Cueni, L. Chen, H. Zhang, D. Marino, R. Huggenberger et al., Podoplanin-Fc reduces lymphatic vessel formation in vitro and in vivo and causes disseminated intravascular coagulation when transgenically expressed in the skin, Blood, vol.116, pp.4376-4384, 2010.

C. Bénézech, S. Nayar, B. A. Finney, D. R. Withers, K. Lowe et al., CLEC-2 is required for development and maintenance of lymph nodes, Blood, vol.123, pp.3200-3207, 2014.

S. E. Acton, J. L. Astarita, D. Malhotra, V. Lukacs-kornek, B. Franz et al., Podoplanin-Rich Stromal Networks Induce Dendritic Cell Motility via Activation of the C-type Lectin Receptor CLEC-2, Immunity, vol.37, pp.276-289, 2012.

P. R. Hess, D. R. Rawnsley, Z. Jakus, Y. Yang, D. T. Sweet et al., Platelets mediate lymphovenous hemostasis to maintain blood-lymphatic separation throughout life, J Clin Invest, vol.124, pp.273-284, 2014.

S. Banerji, J. Ni, S. Wang, S. Clasper, J. Su et al., LYVE-1, a New Homologue of the CD44 Glycoprotein, Is a Lymph-specific Receptor for Hyaluronan, J Cell Biol, vol.144, pp.789-801, 1999.

T. Mäkinen, R. H. Adams, J. Bailey, Q. Lu, A. Ziemiecki et al., PDZ interaction site in ephrinB2 is required for the remodeling of lymphatic vasculature, Genes Dev, vol.19, pp.397-410, 2005.

N. L. Harvey and E. J. Gordon, Deciphering the roles of macrophages in developmental and inflammation stimulated lymphangiogenesis, Vasc Cell, vol.4, p.15, 2012.

S. Ran and K. E. Montgomery, Macrophage-Mediated Lymphangiogenesis: The Emerging Role of Macrophages as Lymphatic Endothelial Progenitors, Cancers, vol.4, pp.618-657, 2012.

I. L. Grigorova, S. R. Schwab, T. G. Phan, T. Pham, T. Okada et al., Cortical sinus probing, S1P1-dependent entry and flow-based capture of egressing T cells, Nat Immunol, vol.10, pp.58-65, 2009.

M. Furuya, S. B. Kirschbaum, A. Paulovich, B. U. Pauli, H. Zhang et al., Lymphatic endothelial mCLCA1 is a ligand for leukocyte LFA-1 and Mac-1, J Immunol Baltim Md, vol.185, pp.5769-5777, 1950.

R. Prevo, S. Banerji, D. Ferguson, S. Clasper, and D. G. Jackson, Mouse LYVE-1 Is an Endocytic Receptor for Hyaluronan in Lymphatic Endothelium, J Biol Chem, vol.276, pp.19420-19430, 2001.

D. Malhotra, A. L. Fletcher, J. Astarita, V. Lukacs-kornek, P. Tayalia et al., Transcriptional profiling of stroma from inflamed and resting lymph nodes defines immunological hallmarks, Nat Immunol, vol.13, pp.499-510, 2012.

J. Lefkovits, E. F. Plow, and E. J. Topol, Platelet Glycoprotein IIb/IIIa Receptors in Cardiovascular Medicine, N Engl J Med, vol.332, pp.1553-1559, 1995.

L. Liu and G. Shi, CD31: beyond a marker for endothelial cells, Cardiovasc Res, vol.94, pp.3-5, 2012.

W. Cl, Normal Structure, Function, and Histology of Lymph Nodes, Toxicol Pathol, vol.34, pp.409-424, 2006.

S. Standring and . Gray, Anatomy: The Anatomical Basis of Clinical Practice, vol.3, 2015.

T. Junt, E. Scandella, and B. Ludewig, Form follows function: lymphoid tissue microarchitecture in antimicrobial immune defence, Nat Rev Immunol, vol.8, pp.764-775, 2008.

T. Katakai, Marginal reticular cells: a stromal subset directly descended from the lymphoid tissue organizer, Front Immunol, vol.3, 2012.

T. Katakai, H. Suto, M. Sugai, H. Gonda, A. Togawa et al., Organizer-Like Reticular Stromal Cell Layer Common to Adult Secondary Lymphoid Organs, J Immunol, vol.181, pp.6189-6200, 2008.

S. N. Mueller and R. N. Germain, Stromal cell contributions to the homeostasis and functionality of the immune system, Nat Rev Immunol, vol.9, pp.618-629, 2009.

S. J. Turley, A. L. Fletcher, and K. G. Elpek, The stromal and haematopoietic antigen-presenting cells that reside in secondary lymphoid organs, Nat Rev Immunol, vol.10, pp.813-825, 2010.

J. J. Koning and R. E. Mebius, Interdependence of stromal and immune cells for lymph node function, Trends Immunol, vol.33, pp.264-270, 2012.

A. Link, T. K. Vogt, S. Favre, M. R. Britschgi, H. Acha-orbea et al., Fibroblastic reticular cells in lymph nodes regulate the homeostasis of naive T cells, Nat Immunol, vol.8, pp.1255-1265, 2007.

J. E. Gretz, C. C. Norbury, A. O. Anderson, A. Proudfoot, and S. Shaw, Lymph-Borne Chemokines and Other Low Molecular Weight Molecules Reach High Endothelial Venules via Specialized Conduits While a Functional Barrier Limits Access to the Lymphocyte Microenvironments in Lymph Node Cortex, J Exp Med, vol.192, pp.1425-1440, 2000.

P. Rantakari, K. Auvinen, N. Jäppinen, M. Kapraali, J. Valtonen et al., The endothelial protein PLVAP in lymphatics controls the entry of lymphocytes and antigens into lymph nodes, Nat Immunol, vol.16, pp.386-396, 2015.

O. Tal, H. Y. Lim, I. Gurevich, M. I. Shipony, Z. Ng et al., DC mobilization from the skin requires docking to immobilized CCL21 on lymphatic endothelium and intralymphatic crawling, J Exp Med, vol.208, pp.2141-2153, 2011.

A. Braun, T. Worbs, G. L. Moschovakis, S. Halle, K. Hoffmann et al., Afferent lymph-derived T cells and DCs use different chemokine receptor CCR7-dependent routes for entry into the lymph node and intranodal migration, Nat Immunol, vol.12, pp.879-887, 2011.

M. H. Ulvmar, K. Werth, A. Braun, P. Kelay, E. Hub et al., The atypical chemokine receptor CCRL1 shapes functional CCL21 gradients in lymph nodes, Nat Immunol, vol.15, pp.623-630, 2014.

C. Qu, E. W. Edwards, F. Tacke, V. Angeli, J. Llodrá et al., Role of CCR8 and Other Chemokine Pathways in the Migration of Monocyte-derived Dendritic Cells to Lymph Nodes, J Exp Med, vol.200, pp.1231-1241, 2004.

I. Iftakhar-e-khuda, R. Fair-mäkelä, A. Kukkonen-macchi, K. Elima, M. Karikoski et al., Geneexpression profiling of different arms of lymphatic vasculature identifies candidates for manipulation of cell traffic, Proc Natl Acad Sci, vol.113, pp.10643-10648, 2016.

S. J. Rouhani, J. D. Eccles, E. F. Tewalt, and V. H. Engelhard, Regulation of T-cell Tolerance by Lymphatic Endothelial Cells, J Clin Cell Immunol, vol.5, 2014.

T. Pham, P. Baluk, Y. Xu, I. Grigorova, A. J. Bankovich et al., Lymphatic endothelial cell sphingosine kinase activity is required for lymphocyte egress and lymphatic patterning, J Exp Med, vol.207, pp.17-27, 2010.

R. Pappu, S. R. Schwab, I. Cornelissen, J. P. Pereira, J. B. Regard et al., Promotion of Lymphocyte Egress into Blood and Lymph by Distinct Sources of Sphingosine-1-Phosphate, Science, vol.316, pp.295-298, 2007.

M. Matloubian, C. G. Lo, G. Cinamon, M. J. Lesneski, Y. Xu et al., Lymphocyte egress from thymus and peripheral lymphoid organs is dependent on S1P receptor 1, Nature, vol.427, pp.355-360, 2004.

K. Ito, J. Morimoto, A. Kihara, Y. Matsui, D. Kurotaki et al., Integrin ?9 on lymphatic endothelial cells regulates lymphocyte egress, Proc Natl Acad Sci, vol.111, pp.3080-3085, 2014.

H. Irjala, E. Johansson, R. Grenman, K. Alanen, M. Salmi et al., Mannose Receptor Is a Novel Ligand for L-Selectin and Mediates Lymphocyte Binding to Lymphatic Endothelium, J Exp Med, vol.194, pp.1033-1042, 2001.

H. Irjala, K. Elima, E. Johansson, M. Merinen, K. Kontula et al., The same endothelial receptor controls lymphocyte traffic both in vascular and lymphatic vessels, Eur J Immunol, vol.33, pp.815-824, 2003.

A. W. Lund, F. V. Duraes, S. Hirosue, V. R. Raghavan, C. Nembrini et al., VEGF-C Promotes Immune Tolerance in B16 Melanomas and Cross-Presentation of Tumor Antigen by Lymph Node Lymphatics, Cell Rep, vol.1, pp.191-199, 2012.

J. N. Cohen, C. J. Guidi, E. F. Tewalt, H. Qiao, S. J. Rouhani et al., Lymph node-resident lymphatic endothelial cells mediate peripheral tolerance via Aire-independent direct antigen presentation, J Exp Med, vol.207, pp.681-688, 2010.

E. F. Tewalt, J. N. Cohen, S. J. Rouhani, C. J. Guidi, H. Qiao et al., Lymphatic endothelial cells induce tolerance via PD-L1 and lack of costimulation leading to high-level PD-1 expression on CD8 T cells, Blood, vol.120, pp.4772-4782, 2012.

S. Amatschek, E. Kriehuber, W. Bauer, B. Reininger, P. Meraner et al., Blood and lymphatic endothelial cell-specific differentiation programs are stringently controlled by the tissue environment, Blood, vol.109, pp.4777-4785, 2007.

C. H. Tripp, B. Haid, V. Flacher, M. Sixt, H. Peter et al., The lymph vessel network in mouse skin visualised with antibodies against the hyaluronan receptor LYVE-1, Immunobiology, vol.213, pp.715-728, 2008.

S. J. Rouhani, J. D. Eccles, P. Riccardi, J. D. Peske, E. F. Tewalt et al., Roles of lymphatic endothelial cells expressing peripheral tissue antigens in CD4 T-cell tolerance induction, Nat Commun, vol.6, p.6771, 2015.

M. Nörder, M. G. Gutierrez, S. Zicari, E. Cervi, A. Caruso et al., Lymph node-derived lymphatic endothelial cells express functional costimulatory molecules and impair dendritic cell-induced allogenic Tcell proliferation, FASEB J, vol.26, pp.2835-2846, 2012.

J. N. Cohen, E. F. Tewalt, S. J. Rouhani, E. L. Buonomo, A. N. Bruce et al., Tolerogenic Properties of Lymphatic Endothelial Cells Are Controlled by the Lymph Node Microenvironment, PLoS ONE, vol.9, p.87740, 2014.

E. F. Tewalt, J. N. Cohen, S. J. Rouhani, and V. H. Engelhard, Lymphatic endothelial cells -key players in regulation of tolerance and immunity, Front Immunol, vol.3, 2012.

C. Koble and B. Kyewski, The thymic medulla: a unique microenvironment for intercellular self-antigen transfer, J Exp Med, vol.206, pp.1505-1513, 2009.

B. A. Tamburini, M. A. Burchill, and R. M. Kedl, Antigen capture and archiving by lymphatic endothelial cells following vaccination or viral infection, Nat Commun, vol.5, p.3989, 2014.

C. M. Card, S. S. Yu, and M. A. Swartz, Emerging roles of lymphatic endothelium in regulating adaptive immunity, J Clin Invest, vol.124, pp.943-952, 2014.

A. Aspelund, T. Tammela, S. Antila, H. Nurmi, V. Leppänen et al., The Schlemm's canal is a VEGF-C/VEGFR-3-responsive lymphatic-like vessel, J Clin Invest, vol.124, pp.3975-3986, 2014.

D. Park, J. Lee, I. Park, D. Choi, S. Lee et al., Lymphatic regulator PROX1 determines Schlemm's canal integrity and identity, J Clin Invest, vol.124, pp.3960-3974, 2014.

A. Aspelund, S. Antila, S. T. Proulx, T. V. Karlsen, S. Karaman et al., A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules, J Exp Med, vol.212, pp.991-999, 2015.

A. Louveau, I. Smirnov, T. J. Keyes, J. D. Eccles, S. J. Rouhani et al., Structural and functional features of central nervous system lymphatic vessels, Nature, vol.523, pp.337-341, 2015.

D. E. Schraufnagel, Lung lymphatic anatomy and correlates, Pathophysiology, vol.17, pp.337-343, 2010.

J. Bernier-latmani, C. Cisarovsky, C. S. Demir, M. Bruand, M. Jaquet et al., DLL4 promotes continuous adult intestinal lacteal regeneration and dietary fat transport, J Clin Invest, vol.125, pp.4572-4586, 2015.

O. Pabst and A. M. Mowat, Oral tolerance to food protein, Mucosal Immunol, vol.5, pp.232-239, 2012.

A. Pegu, S. Qin, B. Junecko, R. E. Nisato, M. S. Pepper et al., Human Lymphatic Endothelial Cells Express Multiple Functional TLRs, J Immunol, vol.180, pp.3399-3405, 2008.

E. Garrafa, L. Imberti, G. Tiberio, A. Prandini, S. M. Giulini et al., Heterogeneous expression of toll-like receptors in lymphatic endothelial cells derived from different tissues, Immunol Cell Biol, vol.89, pp.475-481, 2011.

R. P. Kataru, H. Kim, C. Jang, D. K. Choi, B. I. Koh et al., T Lymphocytes Negatively Regulate Lymph Node Lymphatic Vessel Formation, Immunity, vol.34, pp.96-107, 2011.

Y. Sawa, T. Ueki, M. Hata, K. Iwasawa, E. Tsuruga et al., LPS-induced IL-6, IL-8, VCAM-1, and ICAM-1 Expression in Human Lymphatic Endothelium, J Histochem Cytochem, vol.56, pp.97-109, 2008.

K. M. Lee, C. S. Mckimmie, D. S. Gilchrist, K. J. Pallas, R. J. Nibbs et al., D6 facilitates cellular migration and fluid flow to lymph nodes by suppressing lymphatic congestion, Blood, vol.118, pp.6220-6229, 2011.

G. J. Graham, D6 and the atypical chemokine receptor family: Novel regulators of immune and inflammatory processes, Eur J Immunol, vol.39, pp.342-351, 2009.

O. Bonavita, M. Poeta, V. Setten, E. Massara, M. Bonecchi et al., ACKR2: An Atypical Chemokine Receptor Regulating Lymphatic Biology, Front Immunol, vol.7, 2017.

K. E. Kim, Y. Koh, J. Jang, C. Han, J. Kataru et al., Role of CD11b+ Macrophages in Intraperitoneal Lipopolysaccharide-Induced Aberrant Lymphangiogenesis and Lymphatic Function in the Diaphragm, Am J Pathol, vol.175, pp.1733-1745, 2009.

M. J. Flister, A. Wilber, K. L. Hall, C. Iwata, K. Miyazono et al., Inflammation induces lymphangiogenesis through up-regulation of VEGFR-3 mediated by NF-?B and Prox1, Blood, vol.115, pp.418-429, 2010.

R. H. Mounzer, O. S. Svendsen, P. Baluk, C. M. Bergman, T. P. Padera et al., Lymphotoxin-alpha contributes to lymphangiogenesis, Blood, vol.116, pp.2173-2182, 2010.

C. D. Buckley, F. Barone, S. Nayar, C. Bénézech, and J. Caamaño, Stromal cells in chronic inflammation and tertiary lymphoid organ formation, Annu Rev Immunol, vol.33, pp.715-745, 2015.

R. Ji, Macrophages are important mediators of either tumor-or inflammation-induced lymphangiogenesis, Cell Mol Life Sci, vol.69, pp.897-914, 2011.

H. Kim, R. P. Kataru, and G. Y. Koh, Inflammation-associated lymphangiogenesis: a double-edged sword?, J Clin Invest, vol.124, pp.936-942, 2014.

D. O. Miteva, J. M. Rutkowski, J. B. Dixon, W. Kilarski, J. D. Shields et al., Transmural Flow Modulates Cell and Fluid Transport Functions of Lymphatic Endothelium, Circ Res, vol.106, pp.920-931, 2010.

C. Eich, I. De-vries, P. C. Linssen, A. De-boer, J. B. Boezeman et al., The lymphoid chemokine CCL21 triggers LFA-1 adhesive properties on human dendritic cells, Immunol Cell Biol, vol.89, pp.458-465, 2011.

A. Teijeira, M. C. Hunter, E. Russo, S. T. Proulx, T. Frei et al., Cell Migration from Inflamed Skin to Draining Lymph Nodes Requires Intralymphatic Crawling Supported by ICAM-1/LFA-1 Interactions, Cell Rep, vol.18, pp.857-865, 2017.

K. W. Tan, K. P. Yeo, F. Wong, H. Y. Lim, K. L. Khoo et al., Expansion of Cortical and Medullary Sinuses Restrains Lymph Node Hypertrophy during Prolonged Inflammation, J Immunol, vol.188, pp.4065-4080, 2012.

L. Onder, P. Narang, E. Scandella, Q. Chai, M. Iolyeva et al., IL-7-producing stromal cells are critical for lymph node remodeling, Blood, vol.120, pp.4675-4683, 2012.

M. Iolyeva, D. Aebischer, S. T. Proulx, A. Willrodt, T. Ecoiffier et al., Interleukin-7 is produced by afferent lymphatic vessels and supports lymphatic drainage, Blood, vol.122, pp.2271-2281, 2013.

S. Podgrabinska, O. Kamalu, L. Mayer, M. Shimaoka, H. Snoeck et al., Regulated release of nitric oxide by nonhematopoietic stroma controls expansion of the activated T cell pool in lymph nodes, J Immunol Baltim Md, vol.183, pp.1096-1104, 1950.

S. Nayar, J. Campos, M. M. Chung, L. Navarro-núñez, M. Chachlani et al., Bimodal Expansion of the Lymphatic Vessels Is Regulated by the Sequential Expression of IL-7 and Lymphotoxin ?1?2 in Newly Formed Tertiary Lymphoid Structures, J Immunol Author Choice, vol.197, pp.1957-1967, 2016.

M. K. Gatumu, K. Skarstein, A. Papandile, J. L. Browning, R. A. Fava et al., Blockade of lymphotoxin-beta receptor signaling reduces aspects of Sjögren's syndrome in salivary glands of non-obese diabetic mice, Arthritis Res Ther, vol.11, 2009.

Y. Lee, R. K. Chin, P. Christiansen, Y. Sun, A. V. Tumanov et al., Recruitment and Activation of Naive T Cells in the Islets by Lymphotoxin ? Receptor-Dependent Tertiary Lymphoid Structure, Immunity, vol.25, pp.499-509, 2006.

C. Remouchamps, L. Boutaffala, C. Ganeff, and E. Dejardin, Biology and signal transduction pathways of the Lymphotoxin-??/LT?R system, Cytokine Growth Factor Rev, vol.22, pp.301-310, 2011.

R. C. Ji, Characteristics of lymphatic endothelial cells in physiological and pathological conditions, Histol Histopathol, vol.20, pp.155-175, 2005.

W. D. Melrose, Lymphatic filariasis: new insights into an old disease, Int J Parasitol, vol.32, pp.947-960, 2002.

Y. Saito, H. Nakagami, Y. Kaneda, R. Morishita, T. Lymphedema et al.,

, BioMed Res Int, 2013.

S. A. Stacker, S. P. Williams, T. Karnezis, R. Shayan, S. B. Fox et al., Lymphangiogenesis and lymphatic vessel remodelling in cancer, Nat Rev Cancer, vol.14, pp.159-172, 2014.

M. Caunt, J. Mak, W. Liang, S. Stawicki, Q. Pan et al., Blocking Neuropilin-2 Function Inhibits Tumor Cell Metastasis, Cancer Cell, vol.13, pp.331-342, 2008.

T. Karnezis, R. Shayan, C. Caesar, S. Roufail, N. C. Harris et al., VEGF-D Promotes Tumor Metastasis by Regulating Prostaglandins Produced by the Collecting Lymphatic Endothelium, Cancer Cell, vol.21, pp.181-195, 2012.

M. Oka, C. Iwata, H. I. Suzuki, K. Kiyono, Y. Morishita et al., Inhibition of endogenous TGF-? signaling enhances lymphangiogenesis, Blood, vol.111, pp.4571-4579, 2008.

B. Garmy-susini, C. J. Avraamides, M. C. Schmid, P. Foubert, L. G. Ellies et al., Integrin ?4?1 signaling is required for lymphangiogenesis and tumor metastasis, Cancer Res, vol.70, pp.3042-3051, 2010.

S. Clasper, D. Royston, D. Baban, Y. Cao, S. Ewers et al., A Novel Gene Expression Profile in Lymphatics Associated with Tumor Growth and Nodal Metastasis, Cancer Res, vol.68, pp.7293-7303, 2008.

L. C. Dieterich, K. Ikenberg, T. Cetintas, K. Kapaklikaya, C. Hutmacher et al., Tumor-Associated Lymphatic Vessels Upregulate PDL1 to Inhibit T-Cell Activation, Front Immunol, vol.8, 2017.

E. Hess, V. Duheron, M. Decossas, F. Lézot, A. Berdal et al., RANKL Induces Organized Lymph Node Growth by Stromal Cell Proliferation, J Immunol, vol.188, pp.1245-1254, 2012.
URL : https://hal.archives-ouvertes.fr/pasteur-00782725

A. I. Tauber, Metchnikoff and the phagocytosis theory, Nat Rev Mol Cell Biol, vol.4, pp.897-901, 2003.

R. Van-furth, Z. A. Cohn, J. G. Hirsch, J. H. Humphrey, W. G. Spector et al., The mononuclear phagocyte system: a new classification of macrophages, monocytes, and their precursor cells, Bull World Health Organ, vol.46, pp.845-852, 1972.

E. E. Gray and J. G. Cyster, Lymph node macrophages, J Innate Immun, vol.4, pp.424-436, 2012.

S. Gordon, The macrophage: Past, present and future, Eur J Immunol, vol.37, pp.9-17, 2007.

S. Gordon and P. R. Taylor, Monocyte and macrophage heterogeneity, Nat Rev Immunol, vol.5, pp.953-964, 2005.

J. W. Pollard, Trophic macrophages in development and disease, Nat Rev Immunol, vol.9, pp.259-270, 2009.

, CD169 + macrophages

A. O'neill, T. K. Van-den-berg, and G. Mullen, Sialoadhesin -a macrophage-restricted marker of immunoregulation and inflammation, Immunology, vol.138, pp.198-207, 2013.

P. L. Delputte, H. V. Gorp, H. W. Favoreel, I. Hoebeke, I. Delrue et al., Porcine Sialoadhesin (CD169/Siglec-1) Is an Endocytic Receptor that Allows Targeted Delivery of Toxins and Antigens to Macrophages, PLOS ONE, vol.6, 2011.

W. C. Chen, N. Kawasaki, C. M. Nycholat, S. Han, J. Pilotte et al., Antigen Delivery to Macrophages Using Liposomal Nanoparticles Targeting Sialoadhesin/CD169, PLOS ONE, vol.7, p.39039, 2012.

S. Gordon, J. Hamann, H. Lin, and M. Stacey, F4/80 and the related adhesion-GPCRs, Eur J Immunol, vol.41, pp.2472-2476, 2011.

T. Geijtenbeek, P. C. Groot, M. A. Nolte, S. J. Vliet, . Van et al., Marginal zone macrophages express a murine homologue of DC-SIGN that captures blood-borne antigens in vivo, Blood, vol.100, pp.2908-2916, 2002.

Y. Chen, T. Pikkarainen, O. Elomaa, R. Soininen, T. Kodama et al., Defective Microarchitecture of the Spleen Marginal Zone and Impaired Response to a Thymus-Independent Type 2 Antigen in Mice Lacking Scavenger Receptors MARCO and SR-A, J Immunol, vol.175, pp.8173-8180, 2005.

S. Mukhopadhyay, Y. Chen, M. Sankala, L. Peiser, T. Pikkarainen et al., an innate activation marker of macrophages, is a class A scavenger receptor for Neisseria meningitidis, Eur J Immunol, vol.36, pp.940-949, 2006.

M. Karlsson, R. Guinamard, S. Bolland, M. Sankala, R. M. Steinman et al., Macrophages Control the Retention and Trafficking of B Lymphocytes in the Splenic Marginal Zone, J Exp Med, vol.198, pp.333-340, 2003.

S. Gordon, A. Plüddemann, and S. Mukhopadhyay, Sinusoidal Immunity: Macrophages at the Lymphohematopoietic Interface, Cold Spring Harb Perspect Biol, vol.7, p.16378, 2015.

S. L. Clark, The reticulum of lymph nodes in mice studied with the electron microscope, Am J Anat, vol.110, pp.217-257, 1962.

S. Fossum, The architecture of rat lymph nodes. IV. Distribution of ferritin and colloidal carbon in the draining lymph nodes after foot-pad injection, Scand J Immunol, vol.12, pp.433-441, 1980.

T. G. Phan, J. A. Green, E. E. Gray, Y. Xu, and J. G. Cyster, Immune complex relay by subcapsular sinus macrophages and noncognate B cells drives antibody affinity maturation, Nat Immunol, vol.10, pp.786-793, 2009.

L. Martínez-pomares, M. Kosco-vilbois, E. Darley, P. Tree, S. Herren et al., Fc chimeric protein containing the cysteine-rich domain of the murine mannose receptor binds to macrophages from splenic marginal zone and lymph node subcapsular sinus and to germinal centers, J Exp Med, vol.184, pp.1927-1937, 1996.

T. Junt, E. A. Moseman, M. Iannacone, S. Massberg, P. A. Lang et al., Subcapsular sinus macrophages in lymph nodes clear lymph-borne viruses and present them to antiviral B cells, Nature, vol.450, pp.110-114, 2007.

Y. R. Carrasco and F. D. Batista, B Cells Acquire Particulate Antigen in a Macrophage-Rich Area at the Boundary between the Follicle and the Subcapsular Sinus of the Lymph Node, Immunity, vol.27, pp.160-171, 2007.

, CD169 + macrophages

L. Martinez-pomares and S. Gordon, CD169+ macrophages at the crossroads of antigen presentation, Trends Immunol, vol.33, pp.66-70, 2012.

Y. R. Carrasco, S. J. Fleire, T. Cameron, M. L. Dustin, and F. D. Batista, LFA-1/ICAM-1 Interaction Lowers the Threshold of B Cell Activation by Facilitating B Cell Adhesion and Synapse Formation, Immunity, vol.20, pp.589-599, 2004.

G. Nossal, G. L. Ada, C. M. Austin, and J. Pye, Antigens in immunity, Immunology, vol.9, pp.349-357, 1965.

H. W. Steer and R. A. Foot, Changes in the medulla of the parathymic lymph nodes of the rat during acute gastrointestinal inflammation, J Anat, vol.152, pp.23-36, 1987.

M. Kuka and M. Iannacone, The role of lymph node sinus macrophages in host defense, Ann N Y Acad Sci, vol.1319, pp.38-46, 2014.

F. G. Delemarre, N. Kors, G. Kraal, and N. Van-rooijen, Repopulation of macrophages in popliteal lymph nodes of mice after liposome-mediated depletion, J Leukoc Biol, vol.47, pp.251-257, 1990.

J. Haan, . Den, and L. Martinez-pomares, Macrophage heterogeneity in lymphoid tissues, Semin Immunopathol, vol.35, pp.541-552, 2013.

R. E. Mebius and G. Kraal, Structure and function of the spleen, Nat Rev Immunol, vol.5, pp.606-616, 2005.

H. Borges-da-silva, R. Fonseca, R. M. Pereira, A. Cassado-a-dos, J. M. Álvarez et al., Splenic Macrophage Subsets and Their Function during Blood-Borne Infections, Front Immunol, vol.6, 2015.

L. C. Davies, S. J. Jenkins, J. E. Allen, and P. R. Taylor, Tissue-resident macrophages, Nat Immunol, vol.14, pp.986-995, 2013.

A. Eloranta, Splenic Marginal Metallophilic Macrophages and Marginal Zone Macrophages are the Major Interferon-?/? Producers in Mice upon Intravenous Challenge with Herpes Simplex Virus, Scand J Immunol, vol.49, pp.391-394, 1999.

N. Kawasaki, J. L. Vela, C. M. Nycholat, C. Rademacher, A. Khurana et al., Targeted delivery of lipid antigen to macrophages via the CD169/sialoadhesin endocytic pathway induces robust invariant natural killer T cell activation, Proc Natl Acad Sci, vol.110, pp.7826-7831, 2013.

G. Kraal and M. Janse, Marginal metallophilic cells of the mouse spleen identified by a monoclonal antibody, Immunology, vol.58, pp.665-669, 1986.

J. Den-haan and G. Kraal, Innate Immune Functions of Macrophage Subpopulations in the Spleen, J Innate Immun, vol.4, pp.437-445, 2012.

Y. Kang, S. Yamazaki, T. Iyoda, M. Pack, S. A. Bruening et al., SIGN-R1, a novel C-type lectin expressed by marginal zone macrophages in spleen, mediates uptake of the polysaccharide dextran, Int Immunol, vol.15, pp.177-186, 2003.

O. Elomaa, M. Kangas, C. Sahlberg, J. Tuukkanen, R. Sormunen et al., Cloning of a novel bacteriabinding receptor structurally related to scavenger receptors and expressed in a subset of macrophages, Cell, vol.80, pp.90514-90520, 1995.

S. Gordon, A. Plüddemann, M. Estrada, and F. , Macrophage heterogeneity in tissues: phenotypic diversity and functions, Immunol Rev, vol.262, pp.36-55, 2014.

, CD169 + macrophages

M. Kohyama, W. Ise, B. T. Edelson, P. R. Wilker, K. Hildner et al., Role for Spi-C in the development of red pulp macrophages and splenic iron homeostasis, Nature, vol.457, pp.318-321, 2009.

D. A. Hume, A. P. Robinson, G. G. Macpherson, and S. Gordon, The mononuclear phagocyte system of the mouse defined by immunohistochemical localization of antigen F4/80. Relationship between macrophages, Langerhans cells, reticular cells, and dendritic cells in lymphoid and hematopoietic organs, J Exp Med, vol.158, pp.1522-1536, 1983.

I. H. Hiemstra, M. R. Beijer, H. Veninga, K. Vrijland, E. Borg et al., The identification and developmental requirements of colonic CD169+ macrophages, Immunology, vol.142, pp.269-278, 2014.

K. Asano, N. Takahashi, M. Ushiki, M. Monya, F. Aihara et al., Intestinal CD169+ macrophages initiate mucosal inflammation by secreting CCL8 that recruits inflammatory monocytes, Nat Commun, vol.6, p.7802, 2015.

P. Aichele, J. Zinke, L. Grode, R. A. Schwendener, S. Kaufmann et al., Macrophages of the Splenic Marginal Zone Are Essential for Trapping of Blood-Borne Particulate Antigen but Dispensable for Induction of Specific T Cell Responses, J Immunol, vol.171, pp.1148-1155, 2003.

P. Gupta, S. M. Lai, J. Sheng, P. Tetlak, A. Balachander et al., Tissue-Resident CD169+ Macrophages Form a Crucial Front Line against Plasmodium Infection, Cell Rep, vol.16, pp.1749-1761, 2016.

P. Seiler, P. Aichele, B. Odermatt, H. Hengartner, R. M. Zinkernagel et al., Crucial role of marginal zone macrophages and marginal zone metallophils in the clearance of lymphocytic choriomeningitis virus infection, Eur J Immunol, vol.27, pp.2626-2633, 1997.

H. E. Farrell, N. Davis-poynter, K. Bruce, C. Lawler, L. Dolken et al., Lymph Node Macrophages Restrict Murine Cytomegalovirus Dissemination, J Virol, vol.89, pp.7147-7158, 2015.

M. Iannacone, E. A. Moseman, E. Tonti, L. Bosurgi, T. Junt et al., Subcapsular sinus macrophages prevent CNS invasion on peripheral infection with a neurotropic virus, Nature, vol.465, pp.1079-1083, 2010.

E. A. Moseman, M. Iannacone, L. Bosurgi, E. Tonti, N. Chevrier et al., Cell Maintenance of Subcapsular Sinus Macrophages Protects against a Fatal Viral Infection Independent of Adaptive Immunity, Immunity, vol.36, pp.415-426, 2012.

Z. Garcia, F. Lemaître, M. L. Rooijen-n-van,-albert, Y. Levy, and O. Schwartz, Subcapsular sinus macrophages promote NK cell accumulation and activation in response to lymph-borne viral particles, Blood, vol.120, pp.4744-4750, 2012.
URL : https://hal.archives-ouvertes.fr/pasteur-01110078

P. Barral, P. Polzella, A. Bruckbauer, N. Van-rooijen, G. S. Besra et al., CD169+ macrophages present lipid antigens to mediate early activation of iNKT cells in lymph nodes, Nat Immunol, vol.11, pp.303-312, 2010.

J. L. Coombes, S. Han, N. Van-rooijen, D. H. Raulet, and E. A. Robey, Infection-Induced Regulation of Natural Killer Cells by Macrophages and Collagen at the Lymph Node Subcapsular Sinus, Cell Rep, vol.2, pp.124-135, 2012.

W. Kastenmüller, P. Torabi-parizi, N. Subramanian, T. Lämmermann, and R. N. Germain, A Spatially-Organized Multicellular Innate Immune Response in Lymph Nodes Limits Systemic Pathogen Spread, Cell, vol.150, pp.1235-1248, 2012.

, CD169 + macrophages

A. L. Desbien, N. D. Cauwelaert, S. J. Reed, H. R. Bailor, H. Liang et al., IL-18 and Subcapsular Lymph Node Macrophages are Essential for Enhanced B Cell Responses with TLR4 Agonist Adjuvants, J Immunol, vol.197, pp.4351-4359, 2016.

S. Detienne, I. Welsby, C. Collignon, S. Wouters, M. Coccia et al., Central Role of CD169+ Lymph Node Resident Macrophages in the Adjuvanticity of the QS-21 Component of AS01, Sci Rep, vol.6, p.39475, 2016.

T. G. Phan, I. Grigorova, T. Okada, and J. G. Cyster, Subcapsular encounter and complement-dependent transport of immune complexes by lymph node B cells, Nat Immunol, vol.8, pp.992-1000, 2007.

L. Martinez-pomares and S. Gordon, Antigen Presentation the Macrophage Way, Cell, vol.131, pp.641-643, 2007.

C. Berney, S. Herren, C. A. Power, S. Gordon, L. Martinez-pomares et al., A Member of the Dendritic Cell Family That Enters B Cell Follicles and Stimulates Primary Antibody Responses Identified by a Mannose Receptor Fusion Protein, J Exp Med, vol.190, pp.851-860, 1999.

M. Gaya, A. Castello, B. Montaner, N. Rogers, . Sousa-cr-e et al., Inflammation-induced disruption of SCS macrophages impairs B cell responses to secondary infection, Science, vol.347, pp.667-672, 2015.

T. Chtanova, S. Han, M. Schaeffer, G. G. Van-dooren, P. Herzmark et al., Dynamics of T cell, antigen presenting cell, and pathogen interactions during recall responses in the lymph node, Immunity, vol.31, pp.342-355, 2009.

C. A. Bernhard, C. Ried, S. Kochanek, and T. Brocker, CD169+ macrophages are sufficient for priming of CTLs with specificities left out by cross-priming dendritic cells, Proc Natl Acad Sci, vol.112, pp.5461-5466, 2015.

R. Backer, T. Schwandt, M. Greuter, M. Oosting, F. Jüngerkes et al., Effective collaboration between marginal metallophilic macrophages and CD8+ dendritic cells in the generation of cytotoxic T cells, Proc Natl Acad Sci, vol.107, pp.216-221, 2010.

J. Bouaziz, K. Yanaba, G. M. Venturi, Y. Wang, R. M. Tisch et al., Therapeutic B cell depletion impairs adaptive and autoreactive CD4+ T cell activation in mice, Proc Natl Acad Sci, vol.104, pp.20878-20883, 2007.

J. K. Hu, T. Kagari, J. M. Clingan, and M. Matloubian, Expression of chemokine receptor CXCR3 on T cells affects the balance between effector and memory CD8 T-cell generation, Proc Natl Acad Sci, vol.108, 2011.

E. K. Persson, A. M. Agnarson, H. Lambert, N. Hitziger, H. Yagita et al., Death Receptor Ligation or Exposure to Perforin Trigger Rapid Egress of the Intracellular Parasite Toxoplasma gondii, J Immunol, vol.179, pp.8357-8365, 2007.

L. Beattie, C. R. Engwerda, M. Wykes, and M. F. Good, CD8+ T Lymphocyte-Mediated Loss of Marginal Metallophilic Macrophages following Infection with Plasmodium chabaudi chabaudi AS, J Immunol, vol.177, pp.2518-2526, 2006.

H. D. Hickman, K. Takeda, C. N. Skon, F. R. Murray, S. E. Hensley et al., Direct priming of antiviral CD8+ T cells in the peripheral interfollicular region of lymph nodes, Nat Immunol, vol.9, pp.155-165, 2008.

K. M. Hsu, J. R. Pratt, W. J. Akers, S. I. Achilefu, and W. M. Yokoyama, Murine cytomegalovirus displays selective infection of cells within hours after systemic administration, J Gen Virol, vol.90, pp.33-43, 2009.

, CD169 + macrophages

N. Honke, N. Shaabani, G. Cadeddu, U. R. Sorg, D. Zhang et al., Enforced viral replication activates adaptive immunity and is essential for the control of a cytopathic virus, Nat Immunol, vol.13, pp.51-57, 2012.

B. Ludewig and L. Cervantes-barragan, CD169+ macrophages take the bullet, Nat Immunol, vol.13, pp.13-14, 2012.

T. Chtanova, M. Schaeffer, S. Han, G. G. Van-dooren, M. Nollmann et al., Dynamics of Neutrophil Migration in Lymph Nodes during Infection, Immunity, vol.29, pp.487-496, 2008.

H. Hemmi, J. Idoyaga, K. Suda, N. Suda, K. Kennedy et al., A New Triggering Receptor Expressed on Myeloid Cells (Trem) Family Member, Trem-Like 4, Binds to Dead Cells and Is a DNAX Activation Protein 12-Linked Marker for Subsets of Mouse Macrophages and Dendritic Cells, J Immunol, vol.182, pp.1278-1286, 2009.

G. J. Freeman, J. M. Casasnovas, D. T. Umetsu, and R. H. Dekruyff, TIM genes: a family of cell surface phosphatidylserine receptors that regulate innate and adaptive immunity, Immunol Rev, vol.235, pp.172-189, 2010.

L. A. Albacker, S. Yu, D. Bedoret, W. Lee, S. E. Umetsu et al., TIM-4, expressed by medullary macrophages, regulates respiratory tolerance by mediating phagocytosis of antigen-specific T cells, Mucosal Immunol, vol.6, pp.580-590, 2013.

B. Ravishankar, R. Shinde, H. Liu, K. Chaudhary, J. Bradley et al., Marginal zone CD169+ macrophages coordinate apoptotic cell-driven cellular recruitment and tolerance, Proc Natl Acad Sci, vol.111, pp.4215-4220, 2014.

K. Asano, A. Nabeyama, Y. Miyake, C. Qiu, A. Kurita et al., CD169-Positive Macrophages Dominate Antitumor Immunity by Crosspresenting Dead Cell-Associated Antigens, Immunity, vol.34, pp.85-95, 2011.

D. L. Bratton and P. M. Henson, Apoptotic Cell Recognition: Will the Real Phosphatidylserine Receptor(s) Please Stand up?, Curr Biol, vol.18, pp.76-79, 2008.

F. Pucci, C. Garris, C. P. Lai, A. Newton, C. Pfirschke et al., SCS macrophages suppress melanoma by restricting tumor-derived vesicle-B cell interactions, Science, 2016.

K. Ohnishi, Y. Komohara, Y. Saito, Y. Miyamoto, M. Watanabe et al., CD169-positive macrophages in regional lymph nodes are associated with a favorable prognosis in patients with colorectal carcinoma, Cancer Sci, vol.104, pp.1237-1244, 2013.

K. Ohnishi, M. Yamaguchi, C. Erdenebaatar, F. Saito, H. Tashiro et al., Prognostic significance of CD169-positive lymph node sinus macrophages in patients with endometrial carcinoma, Cancer Sci, 2016.

,

T. Shiota, Y. Miyasato, K. Ohnishi, M. Yamamoto-ibusuki, Y. Yamamoto et al., The Clinical Significance of CD169-Positive Lymph Node Macrophage in Patients with Breast Cancer, PLOS ONE, vol.11, 2016.

M. D. Witmer-pack, D. A. Hughes, G. Schuler, L. Lawson, A. Mcwilliam et al., Identification of macrophages and dendritic cells in the osteopetrotic (op/op) mouse, J Cell Sci, vol.104, pp.1021-1029, 1993.

M. G. Cecchini, M. G. Dominguez, S. Mocci, A. Wetterwald, R. Felix et al., Role of colony stimulating factor-1 in the establishment and regulation of tissue macrophages during postnatal development of the mouse, Development, vol.120, pp.1357-1372, 1994.

, CD169 + macrophages

G. R. Ryan, X. Dai, M. G. Dominguez, W. Tong, F. Chuan et al., Rescue of the colony-stimulating factor 1 (CSF-1)-nullizygous mouse (Csf1 op /Csf1 op ) phenotype with a CSF-1 transgene and identification of sites of local CSF-1 synthesis, Blood, vol.98, pp.74-84, 2001.

P. D. Rennert, J. L. Browning, R. Mebius, F. Mackay, and P. S. Hochman, Surface lymphotoxin alpha/beta complex is required for the development of peripheral lymphoid organs, J Exp Med, vol.184, pp.1999-2006, 1996.

A. V. Tumanov, S. I. Grivennikov, A. N. Shakhov, S. A. Rybtsov, E. P. Koroleva et al., Dissecting the role of lymphotoxin in lymphoid organs by conditional targeting, Immunol Rev, vol.195, pp.106-116, 2003.

L. Poljak, L. Carlson, K. Cunningham, M. H. Kosco-vilbois, and U. Siebenlist, Distinct Activities of p52/NF-?B Required for Proper Secondary Lymphoid Organ Microarchitecture: Functions Enhanced by Bcl-3, J Immunol, vol.163, pp.6581-6588, 1999.

R. Ettinger, R. Mebius, J. L. Browning, S. A. Michie, and G. Tuijl-s-van,-kraal, Effects of tumor necrosis factor and lymphotoxin on peripheral lymphoid tissue development, Int Immunol, vol.10, pp.727-741, 1998.

M. Pasparakis, S. Kousteni, J. Peschon, and G. Kollias, Tumor Necrosis Factor and the p55TNF Receptor Are Required for Optimal Development of the Marginal Sinus and for Migration of Follicular Dendritic Cell Precursors into Splenic Follicles, Cell Immunol, vol.201, pp.33-41, 2000.

A. Guillen, J. A. Gallardo, G. Diaz, M. De-la-rosa, J. V. Hernandez et al., The nuclear receptor LXR? controls the functional specialization of splenic macrophages, Nat Immunol, vol.14, pp.831-839, 2013.

M. J. Pittet and R. Weissleder, Intravital Imaging. Cell, vol.147, pp.983-991, 2011.

. Furth-r-van and M. Dulk, Dual origin of mouse spleen macrophages, J Exp Med, vol.160, pp.1273-1283, 1984.

J. Westermann, S. Ronneberg, F. J. Fritz, and R. Pabst, Proliferation of macrophage subpopulations in the adult rat: comparison of various lymphoid organs, J Leukoc Biol, vol.46, pp.263-269, 1989.

N. Van-rooijen, N. Kors, and G. Kraal, Macrophage subset repopulation in the spleen: differential kinetics after liposome-mediated elimination, J Leukoc Biol, vol.45, pp.97-104, 1989.

Y. Miyake, K. Asano, H. Kaise, M. Uemura, M. Nakayama et al., Critical role of macrophages in the marginal zone in the suppression of immune responses to apoptotic cell-associated antigens, J Clin Invest, vol.117, pp.2268-2278, 2007.

R. E. Mebius, G. Martens, J. Brevé, F. Delemarre, and G. Kraal, Is early repopulation of macrophage-depleted lymph node independent of blood monocyte immigration?, Eur J Immunol, vol.21, pp.3041-3044, 1991.

A. Chow, M. Huggins, J. Ahmed, D. Hashimoto, D. Lucas et al., CD169+ macrophages provide a niche promoting erythropoiesis under homeostasis and stress, Nat Med, vol.19, pp.429-436, 2013.

R. N. Jacobsen, C. E. Forristal, L. J. Raggatt, B. Nowlan, V. Barbier et al., Mobilization with granulocyte colony-stimulating factor blocks medullar erythropoiesis by depleting F4/80+VCAM1+CD169+ER-HR3+Ly6G+ erythroid island macrophages in the mouse, Exp Hematol, vol.42, pp.547-561, 2014.

, CD169 + macrophages

M. Albiero, N. Poncina, S. Ciciliot, R. Cappellari, L. Menegazzo et al., Bone Marrow Macrophages Contribute to Diabetic Stem Cell Mobilopathy by Producing Oncostatin M, Diabetes, vol.64, pp.2957-2968, 2015.

A. Chow, D. Lucas, A. Hidalgo, S. Méndez-ferrer, D. Hashimoto et al., Bone marrow CD169+ macrophages promote the retention of hematopoietic stem and progenitor cells in the mesenchymal stem cell niche, J Exp Med, vol.208, pp.261-271, 2011.

Y. Ikezumi, T. Suzuki, S. Hayafuji, S. Okubo, D. J. Nikolic-paterson et al., The sialoadhesin (CD169) expressing a macrophage subset in human proliferative glomerulonephritis, Nephrol Dial Transplant, vol.20, pp.2704-2713, 2005.

K. Karasawa, K. Asano, S. Moriyama, M. Ushiki, M. Monya et al., Vascular-Resident CD169-Positive Monocytes and Macrophages Control Neutrophil Accumulation in the Kidney with Ischemia-Reperfusion Injury, J Am Soc Nephrol JASN, vol.26, pp.896-906, 2015.

A. Hartnell, J. Steel, H. Turley, M. Jones, D. G. Jackson et al., Characterization of human sialoadhesin, a sialic acid binding receptor expressed by resident and inflammatory macrophage populations, Blood, vol.97, pp.288-296, 2001.

M. B. Graeber, W. J. Streit, R. Kiefer, S. W. Schoen, and G. W. Kreutzberg, New expression of myelomonocytic antigens by microglia and perivascular cells following lethal motor neuron injury, J Neuroimmunol, vol.27, pp.121-132, 1990.

M. Gijbels, M. Van-der-cammen, L. Van-der-laan, J. J. Emeis, L. M. Havekes et al., Progression and regression of atherosclerosis in APOE3-Leiden transgenic mice: an immunohistochemical study, Atherosclerosis, vol.143, pp.15-25, 1999.

Y. Zhang, J. Li, Z. Jiang, L. Li, Y. Wu et al., CD169 identifies an anti-tumour macrophage subpopulation in human hepatocellular carcinoma, J Pathol, vol.239, pp.231-241, 2016.

B. S. Steiniger, Human spleen microanatomy: why mice do not suffice, Immunology, vol.145, pp.334-346, 2015.

J. Martens, J. Kzhyshkowska, M. Falkowski-hansen, K. Schledzewski, A. Gratchev et al., Differential expression of a gene signature for scavenger/lectin receptors by endothelial cells and macrophages in human lymph node sinuses, the primary sites of regional metastasis, J Pathol, vol.208, pp.574-589, 2006.

D. M. Anderson, E. Maraskovsky, W. L. Billingsley, W. C. Dougall, M. E. Tometsko et al., Nature, vol.390, pp.175-179, 1997.

B. R. Wong, R. Josien, S. Y. Lee, B. Sauter, H. Li et al., J. Exp. Med, vol.186, pp.2075-2080, 1997.

W. S. Simonet, D. L. Lacey, C. R. Dunstan, M. Kelley, M. Chang et al., Cell, vol.89, pp.309-319, 1997.

M. C. Walsh and Y. Choi, Inflammation, vol.5, p.511, 2014.

K. Okamoto and H. Takayanagi, Arthritis Res. Ther, vol.13, p.219, 2011.

E. González-suárez, A. Sanz-moreno, and F. J. , , vol.283, pp.2018-2033, 2016.

X. Ma, Y. Liu, Y. Zhang, X. Yu, W. Wang et al., Biochem. Biophys. Res. Commun, vol.445, pp.282-288, 2014.

Y. Chen, J. Sun, C. Dou, N. Li, F. Kang et al., Int. J. Mol. Sci, p.17, 2016.

Y. Xiu, H. Xu, C. Zhao, J. Li, Y. Morita et al., J. Clin. Invest, vol.124, pp.297-310, 2014.

C. Lee, F. Liu, C. Chen, T. Chen, D. Chang et al., Eur. J. Med. Chem, vol.98, pp.115-126, 2015.

B. Aggarwal, B. Darnay, and S. Singh, Inhibitors of Receptor Activator of NF-kappaB and Uses Thereof, pp.20040167072-1, 2004.

H. Kim, H. K. Choi, J. H. Shin, K. H. Kim, J. Y. Huh et al., J. Clin. Invest, vol.119, pp.813-825, 2009.

G. Kato, Y. Shimizu, Y. Arai, N. Suzuki, Y. Sugamori et al., Arthritis Res. Ther, vol.17, p.251, 2015.

J. Hur, A. Ghosh, K. Kim, H. M. Ta, H. Kim et al., Mol. Cells, vol.39, pp.316-321, 2016.

D. L. Lacey, W. J. Boyle, W. S. Simonet, P. J. Kostenuik, W. C. Dougall et al., Nat. Rev. Drug Discov, vol.11, pp.401-419, 2012.

G. Porcu, E. Serone, V. D. Nardis, D. D. Giandomenico, G. Lucisano et al., PLOS ONE, vol.10, p.144550, 2015.

C. Corbel, B. Zhang, A. L. Parc, B. Baratte, P. Colas et al., Chem. Biol, vol.22, pp.472-482, 2015.

M. Chypre, J. Seaman, O. G. Cordeiro, L. Willen, K. A. Knoop et al., Immunol. Lett, vol.171, pp.5-14, 2016.

T. Ciucci, L. Ibáñez, A. Boucoiran, E. Birgy-barelli, J. Pène et al., Gut, vol.64, pp.1072-1081, 2015.

P. J. Kostenuik, H. Q. Nguyen, J. Mccabe, K. S. Warmington, C. Kurahara et al., J. Bone Miner. Res, vol.24, pp.182-195, 2009.

F. Toberer, J. Sykora, D. Göttel, V. Ruland, W. Hartschuh et al., Exp. Dermatol, vol.20, pp.600-602, 2011.

V. I. Group, Am. J. Ophthalmol, vol.131, pp.541-560, 2001.

U. Schmidt-erfurth and T. Hasan, Surv. Ophthalmol, vol.45, pp.195-214, 2000.

P. Schneider, L. Willen, and C. R. Smulski, , pp.103-125, 2014.

L. K. Swee, K. Ingold-salamin, A. Tardivel, L. Willen, O. Gaide et al., J. Biol. Chem, vol.284, pp.27567-27576, 2009.

, RANK ?CD11c and RANK ?LysM mice were bred and kept in specific pathogen-free conditions. All experiments were carried out in conformity with the animal bioethics legislation and institutional guidelines. To generate mice with conditional RANKL deficiency in marginal reticular cells, Ly5.1 (CD45.1) and RANKL ?Ccl19

, RANKLf/f (B6.129-Tnfsf11tm1.1Caob/J) mice, vol.19

, CD11c Cre;RANK flox/flox (RANK ?CD11c ) mice and LysM Cre;RANK flox/flox (RANK ?LysM ) mice were kindly provided by Toby

. France, To inhibit lymphotoxin, RANKL and TNF pathway, mice received 20 µg of LT?R-muIgG1 (LT?R-Fc , kindly provided by Biogen

, ?b-actin (CD45.2+) and from Ly5.1 (RANK +/+ ;CD45.1+) mice

. Briefly, Collagenase D (Roche,1mg/ml), DNase I (Roche, 0.1mg/ml) under agitation at 37°C for 1h. The cell suspension was then filtered at 100µm, RPMI + 2% FCS + Dispase II (Roche, 1mg/ml)

O. G. Cordeiro, M. Chypre, N. Brouard, S. Rauber, F. Alloush et al., Integrin-Alpha IIb Identifies Murine Lymph Node Lymphatic Endothelial Cells Responsive to RANKL, PLOS ONE, vol.11, 2016.

Z. Garcia, F. Lemaître, M. L. Rooijen-n-van,-albert, Y. Levy, and O. Schwartz, Subcapsular sinus macrophages promote NK cell accumulation and activation in response to lymph-borne viral particles, Blood, vol.120, pp.4744-4750, 2012.
URL : https://hal.archives-ouvertes.fr/pasteur-01110078

M. Iannacone, E. A. Moseman, E. Tonti, L. Bosurgi, T. Junt et al., Subcapsular sinus macrophages prevent CNS invasion on peripheral infection with a neurotropic virus, Nature, vol.465, pp.1079-1083, 2010.

E. A. Moseman, M. Iannacone, L. Bosurgi, E. Tonti, N. Chevrier et al., Cell Maintenance of Subcapsular Sinus Macrophages Protects against a Fatal Viral Infection Independent of Adaptive Immunity, Immunity, vol.36, pp.415-426, 2012.

J. L. Coombes, S. Han, N. Van-rooijen, D. H. Raulet, and E. A. Robey, Infection-Induced Regulation of Natural Killer Cells by Macrophages and Collagen at the Lymph Node Subcapsular Sinus, Cell Rep, vol.2, pp.124-135, 2012.

Y. R. Carrasco and F. D. Batista, B Cells Acquire Particulate Antigen in a Macrophage-Rich Area at the Boundary between the Follicle and the Subcapsular Sinus of the Lymph Node, Immunity, vol.27, pp.160-171, 2007.

T. G. Phan, I. Grigorova, T. Okada, and J. G. Cyster, Subcapsular encounter and complement-dependent transport of immune complexes by lymph node B cells, Nat Immunol, vol.8, pp.992-1000, 2007.

S. F. Gonzalez, V. Lukacs-kornek, M. P. Kuligowski, L. A. Pitcher, S. E. Degn et al., Capture of influenza by medullary dendritic cells via SIGN-R1 is essential for humoral immunity in draining lymph nodes, Nat Immunol, vol.11, pp.427-434, 2010.

T. G. Phan, J. A. Green, E. E. Gray, Y. Xu, and J. G. Cyster, Immune complex relay by subcapsular sinus macrophages and noncognate B cells drives antibody affinity maturation, Nat Immunol, vol.10, pp.786-793, 2009.

W. C. Dougall, M. Glaccum, K. Charrier, K. Rohrbach, K. Brasel et al., RANK is essential for osteoclast and lymph node development, Genes Dev, vol.13, pp.2412-2424, 1999.

Y. Kong, H. Yoshida, I. Sarosi, H. Tan, E. Timms et al., OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis, Nature, vol.397, pp.315-323, 1999.

M. C. Walsh and Y. Choi, Biology of the RANKL-RANK-OPG system in immunity, bone, and beyond, Inflammation, vol.5, p.511, 2014.

J. Barbaroux, M. Beleut, C. Brisken, C. G. Mueller, and R. W. Groves, Epidermal Receptor Activator of NF-?B Ligand Controls Langerhans Cells Numbers and Proliferation, J Immunol, vol.181, pp.1103-1108, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00300531

C. G. Mueller and E. Hess, Emerging Functions of RANKL in Lymphoid Tissues, Front Immunol, vol.3, 2012.

T. Katakai, H. Suto, M. Sugai, H. Gonda, A. Togawa et al., Organizer-Like Reticular Stromal Cell Layer Common to Adult Secondary Lymphoid Organs, J Immunol, vol.181, pp.6189-6200, 2008.

M. Jarjour, A. Jorquera, I. Mondor, S. Wienert, P. Narang et al., Fate mapping reveals origin and dynamics of lymph node follicular dendritic cells, J Exp Med, vol.211, pp.1109-1122, 2014.

N. Kim, P. R. Odgren, D. Kim, S. C. Marks, and Y. Choi, Diverse roles of the tumor necrosis factor family member TRANCE in skeletal physiology revealed by TRANCE deficiency and partial rescue by a lymphocyteexpressed TRANCE transgene, Proc Natl Acad Sci U S A, vol.97, pp.10905-10910, 2000.

Q. Chai, L. Onder, E. Scandella, C. Gil-cruz, C. Perez-shibayama et al., Maturation of Lymph Node Fibroblastic Reticular Cells from Myofibroblastic Precursors Is Critical for Antiviral Immunity, Immunity, vol.38, pp.1013-1024, 2013.

J. Xiong, M. Onal, R. L. Jilka, R. S. Weinstein, S. C. Manolagas et al., Matrix-embedded cells control osteoclast formation, Nat Med, vol.17, pp.1235-1241, 2011.

S. Kamijo, A. Nakajima, K. Ikeda, K. Aoki, K. Ohya et al., Amelioration of bone loss in collagen-induced arthritis by neutralizing anti-RANKL monoclonal antibody, Biochem Biophys Res Commun, vol.347, pp.124-132, 2006.

A. L. Fletcher, D. Malhotra, S. E. Acton, V. Lukacs-kornek, A. Bellemare-pelletier et al., Reproducible Isolation of Lymph Node Stromal Cells Reveals Site-Dependent Differences in Fibroblastic Reticular Cells, Front Immunol, vol.2, 2011.

A. Link, T. K. Vogt, S. Favre, M. R. Britschgi, H. Acha-orbea et al., Fibroblastic reticular cells in lymph nodes regulate the homeostasis of naive T cells, Nat Immunol, vol.8, pp.1255-1265, 2007.

R. Ettinger, R. Mebius, J. L. Browning, S. A. Michie, and G. Tuijl-s-van,-kraal, Effects of tumor necrosis factor and lymphotoxin on peripheral lymphoid tissue development, Int Immunol, vol.10, pp.727-741, 1998.

M. Pasparakis, S. Kousteni, J. Peschon, and G. Kollias, Tumor Necrosis Factor and the p55TNF Receptor Are Required for Optimal Development of the Marginal Sinus and for Migration of Follicular Dendritic Cell Precursors into Splenic Follicles, Cell Immunol, vol.201, pp.33-41, 2000.

C. Bénézech, A. White, E. Mader, K. Serre, S. Parnell et al., Ontogeny of Stromal Organizer Cells during Lymph Node Development, J Immunol, vol.184, pp.4521-4530, 2010.

E. Hess, V. Duheron, M. Decossas, F. Lézot, A. Berdal et al., RANKL Induces Organized Lymph Node Growth by Stromal Cell Proliferation, J Immunol, vol.188, pp.1245-1254, 2012.
URL : https://hal.archives-ouvertes.fr/pasteur-00782725

F. Moalli, S. T. Proulx, R. Schwendener, M. Detmar, C. Schlapbach et al., Intravital and Whole-Organ Imaging Reveals Capture of Melanoma-Derived Antigen by Lymph Node Subcapsular Macrophages Leading to Widespread Deposition on, Follicular Dendritic Cells. Front Immunol, vol.6, 2015.

L. E. Theill, W. J. Boyle, and J. M. Penninger, RANK-L AND RANK: T Cells, Bone Loss, and Mammalian Evolution, Annu Rev Immunol, vol.20, pp.795-823, 2002.

C. L. Abram, G. L. Roberge, Y. Hu, and C. A. Lowell, Comparative analysis of the efficiency and specificity of myeloidCre deleting strains using ROSA-EYFP reporter mice, J Immunol Methods, vol.408, pp.89-100, 2014.

C. Jakubzick, M. Bogunovic, A. J. Bonito, E. L. Kuan, M. Merad et al., Lymph-migrating, tissue-derived dendritic cells are minor constituents within steady-state lymph nodes, J Exp Med, vol.205, pp.2839-2850, 2008.

K. Asano, A. Nabeyama, Y. Miyake, C. Qiu, A. Kurita et al., CD169-Positive Macrophages Dominate Antitumor Immunity by Crosspresenting Dead Cell-Associated Antigens, Immunity, vol.34, pp.85-95, 2011.

F. Pucci, C. Garris, C. P. Lai, A. Newton, C. Pfirschke et al., SCS macrophages suppress melanoma by restricting tumor-derived vesicle-B cell interactions, Science, 2016.

I. H. Hiemstra, M. R. Beijer, H. Veninga, K. Vrijland, E. Borg et al., The identification and developmental requirements of colonic CD169+ macrophages, Immunology, vol.142, pp.269-278, 2014.

A. Chow, D. Lucas, A. Hidalgo, S. Méndez-ferrer, D. Hashimoto et al., Bone marrow CD169+ macrophages promote the retention of hematopoietic stem and progenitor cells in the mesenchymal stem cell niche, J Exp Med, vol.208, pp.261-271, 2011.

A. Chow, M. Huggins, J. Ahmed, D. Hashimoto, D. Lucas et al., CD169+ macrophages provide a niche promoting erythropoiesis under homeostasis and stress, Nat Med, vol.19, pp.429-436, 2013.

D. L. Lacey, W. J. Boyle, W. S. Simonet, P. J. Kostenuik, W. C. Dougall et al., Bench to bedside: elucidation of the OPG-RANK-RANKL pathway and the development of denosumab, Nat Rev Drug Discov, vol.11, pp.401-419, 2012.

E. Coste, I. R. Greig, P. Mollat, L. Rose, M. Gray et al., Identification of small molecule inhibitors of RANKL and TNF signalling as anti-inflammatory and antiresorptive agents in mice, Ann Rheum Dis, vol.74, pp.220-226, 2015.

B. Aggarwal, B. Darnay, and S. Singh, Inhibitors of receptor activator of NF-kappaB and uses thereof

. Us20040167072-a1, , 2004.

V. Naidu, D. Babu, K. R. Thwin, M. M. Satish, R. L. Kumar et al., RANKL targeted peptides inhibit osteoclastogenesis and attenuate adjuvant induced arthritis by inhibiting NF-?B activation and down regulating inflammatory cytokines, Chem Biol Interact, vol.203, pp.467-479, 2013.

X. Cheng, M. Kinosaki, M. Takami, Y. Choi, H. Zhang et al., Disabling of Receptor Activator of Nuclear Factor-?B (RANK) Receptor Complex by Novel Osteoprotegerin-like Peptidomimetics Restores Bone Loss in Vivo, J Biol Chem, vol.279, pp.8269-8277, 2004.

U. Schmidt-erfurth and T. Hasan, Mechanisms of Action of Photodynamic Therapy with Verteporfin for the Treatment of Age-Related Macular Degeneration, Surv Ophthalmol, vol.45, pp.195-214, 2000.

E. Donohue, A. Tovey, A. W. Vogl, S. Arns, E. Sternberg et al., Inhibition of Autophagosome Formation by the Benzoporphyrin Derivative Verteporfin, J Biol Chem, vol.286, 2011.

E. Donohue, A. Thomas, N. Maurer, I. Manisali, M. Zeisser-labouebe et al., The Autophagy Inhibitor Verteporfin Moderately Enhances the Antitumor Activity of Gemcitabine in a Pancreatic Ductal Adenocarcinoma Model, J Cancer, vol.4, pp.585-596, 2013.

J. Feng, J. Gou, J. Jia, T. Yi, T. Cui et al., Verteporfin, a suppressor of YAP-TEAD complex, presents promising antitumor properties on ovarian cancer, OncoTargets Ther, vol.9, pp.5371-5381, 2016.

H. Zhang, S. K. Ramakrishnan, D. Triner, B. Centofanti, D. Maitra et al., Tumor-selective proteotoxicity of verteporfin inhibits colon cancer progression independently of YAP1, Sci Signal, vol.8, pp.98-98, 2015.

T. G. Phan, J. A. Green, E. E. Gray, Y. Xu, and J. G. Cyster, Immune complex relay by subcapsular sinus macrophages and noncognate B cells drives antibody affinity maturation, Nat Immunol, vol.10, pp.786-793, 2009.

T. G. Phan, I. Grigorova, T. Okada, and J. G. Cyster, Subcapsular encounter and complement-dependent transport of immune complexes by lymph node B cells, Nat Immunol, vol.8, pp.992-1000, 2007.

T. Junt, E. A. Moseman, M. Iannacone, S. Massberg, P. A. Lang et al., Subcapsular sinus macrophages in lymph nodes clear lymph-borne viruses and present them to antiviral B cells, Nature, vol.450, pp.110-114, 2007.

Y. R. Carrasco and F. D. Batista, B Cells Acquire Particulate Antigen in a Macrophage-Rich Area at the Boundary between the Follicle and the Subcapsular Sinus of the Lymph Node, Immunity, vol.27, pp.160-171, 2007.

M. Iannacone, E. A. Moseman, E. Tonti, L. Bosurgi, T. Junt et al., Subcapsular sinus macrophages prevent CNS invasion on peripheral infection with a neurotropic virus, Nature, vol.465, pp.1079-1083, 2010.

G. Nossal, G. L. Ada, C. M. Austin, and J. Pye, Antigens in immunity, Immunology, vol.9, pp.349-357, 1965.

S. Fossum, The architecture of rat lymph nodes. IV. Distribution of ferritin and colloidal carbon in the draining lymph nodes after foot-pad injection, Scand J Immunol, vol.12, pp.433-441, 1980.

H. W. Steer and R. A. Foot, Changes in the medulla of the parathymic lymph nodes of the rat during acute gastrointestinal inflammation, J Anat, vol.152, pp.23-36, 1987.

E. A. Moseman, M. Iannacone, L. Bosurgi, E. Tonti, N. Chevrier et al., Cell Maintenance of Subcapsular Sinus Macrophages Protects against a Fatal Viral Infection Independent of Adaptive Immunity, Immunity, vol.36, pp.415-426, 2012.

E. Hess, V. Duheron, M. Decossas, F. Lézot, A. Berdal et al., RANKL Induces Organized Lymph Node Growth by Stromal Cell Proliferation, J Immunol, vol.188, pp.1245-1254, 2012.
URL : https://hal.archives-ouvertes.fr/pasteur-00782725

J. N. Cohen, E. F. Tewalt, S. J. Rouhani, E. L. Buonomo, A. N. Bruce et al., Tolerogenic Properties of Lymphatic Endothelial Cells Are Controlled by the Lymph Node Microenvironment, PLoS ONE, vol.9, p.87740, 2014.

D. Rios, M. B. Wood, J. Li, B. Chassaing, A. T. Gewirtz et al., Antigen sampling by intestinal M cells is the principal pathway initiating mucosal IgA production to commensal enteric bacteria, Mucosal Immunol, vol.9, pp.907-916, 2016.

D. Malhotra, A. L. Fletcher, J. Astarita, V. Lukacs-kornek, P. Tayalia et al., Transcriptional profiling of stroma from inflamed and resting lymph nodes defines immunological hallmarks, Nat Immunol, vol.13, pp.499-510, 2012.

S. Fiorentini, A. Luganini, V. Dell'oste, B. Lorusso, E. Cervi et al., Human cytomegalovirus productively infects lymphatic endothelial cells and induces a secretome that promotes angiogenesis and lymphangiogenesis through interleukin-6 and granulocyte-macrophage colony-stimulating factor, J Gen Virol, vol.92, pp.650-660, 2011.

M. G. Cecchini, M. G. Dominguez, S. Mocci, A. Wetterwald, R. Felix et al., Role of colony stimulating factor-1 in the establishment and regulation of tissue macrophages during postnatal development of the mouse, Development, vol.120, pp.1357-1372, 1994.

H. Rempel, C. Calosing, B. Sun, and L. Pulliam, Sialoadhesin Expressed on IFN-Induced Monocytes Binds HIV-1 and Enhances Infectivity, PLOS ONE, vol.3, p.1967, 2008.

M. R. York, T. Nagai, A. J. Mangini, R. Lemaire, J. M. Van-seventer et al., A macrophage marker, siglec-1, is increased on circulating monocytes in patients with systemic sclerosis and induced by type i interferons and toll-like receptor agonists, Arthritis Rheum, vol.56, pp.1010-1020, 2007.

. Berg-tk-van-den, C. R. Die-i-van,-lavalette, . De, E. A. Döpp, L. D. Smit et al., Regulation of sialoadhesin expression on rat macrophages. Induction by glucocorticoids and enhancement by IFN-beta, IFN-gamma, IL-4, and lipopolysaccharide, J Immunol, vol.157, pp.3130-3138, 1996.

N. A. Mabbott, D. S. Donaldson, H. Ohno, I. R. Williams, and M. A. Microfold, M) cells: important immunosurveillance posts in the intestinal epithelium, Mucosal Immunol, vol.6, pp.666-677, 2013.

M. M. Guerrini, K. Okamoto, N. Komatsu, S. Sawa, L. Danks et al., Inhibition of the TNF Family Cytokine RANKL Prevents Autoimmune Inflammation in the Central Nervous System, Immunity, vol.43, pp.1174-1185, 2015.

I. H. Hiemstra, M. R. Beijer, H. Veninga, K. Vrijland, E. Borg et al., The identification and developmental requirements of colonic CD169+ macrophages, Immunology, vol.142, pp.269-278, 2014.

K. Asano, N. Takahashi, M. Ushiki, M. Monya, F. Aihara et al., Intestinal CD169+ macrophages initiate mucosal inflammation by secreting CCL8 that recruits inflammatory monocytes, Nat Commun, vol.6, p.7802, 2015.

K. Asano, A. Nabeyama, Y. Miyake, C. Qiu, A. Kurita et al., CD169-Positive Macrophages Dominate Antitumor Immunity by Crosspresenting Dead Cell-Associated Antigens, Immunity, vol.34, pp.85-95, 2011.

Y. Kim, Y. Kim, Y. M. Lee, H. Kim, J. D. Kim et al., TNF-related Activation-induced Cytokine (TRANCE) Induces Angiogenesis through the Activation of Src and Phospholipase C (PLC) in Human Endothelial Cells, J Biol Chem, vol.277, pp.6799-6805, 2002.

S. A. Stacker, S. P. Williams, T. Karnezis, R. Shayan, S. B. Fox et al., Lymphangiogenesis and lymphatic vessel remodelling in cancer, Nat Rev Cancer, vol.14, pp.159-172, 2014.

E. L. Berg, L. M. Mcevoy, C. Berlin, R. F. Bargatze, and E. C. Butcher, L-selectin-mediated lymphocyte rolling on MAdCAM-1, Nature, vol.366, pp.695-698, 1993.

C. Berlin, E. L. Berg, M. J. Briskin, D. P. Andrew, P. J. Kilshaw et al., ?4?7 integrin mediates lymphocyte binding to the mucosal vascular addressin MAdCAM-1, Poster : Bristish Society for Immunology summer school, vol.74, pp.185-195, 1993.