H. Lin, E. Lee, K. Hestir, C. Leo, M. Huang et al., Discovery of a cytokine and its receptor by functional screening of the extracellular proteome, Science, vol.320, issue.5877, pp.807-811, 2008.

S. Nandi, M. Cioce, Y. G. Yeung, E. Nieves, L. Tesfa et al., Receptor-type protein-tyrosine phosphatase zeta is a functional receptor for interleukin-34, J Biol Chem, vol.288, issue.30, pp.21972-21986, 2013.

J. H. Baek, R. Zeng, J. Weinmann-menke, M. T. Valerius, Y. Wada et al., IL-34 mediates acute kidney injury and worsens subsequent chronic kidney disease, J Clin Invest, vol.125, issue.8, pp.3198-3214, 2015.
DOI : 10.1172/jci81166

URL : http://www.jci.org/articles/view/81166/files/pdf

A. I. Segaliny, R. Brion, E. Mortier, M. Maillasson, M. Cherel et al., Syndecan-1 regulates the biological activities of interleukin-34, Biochim Biophys Acta, vol.1853, issue.5, pp.1010-1021, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-01644783

S. Wei, S. Nandi, V. Chitu, Y. G. Yeung, W. Yu et al., Functional overlap but differential expression of CSF-1 and IL-34 in their CSF-1 receptormediated regulation of myeloid cells, J Leukoc Biol, vol.88, issue.3, pp.495-505, 2010.

A. I. Segaliny, R. Brion, B. Brulin, M. Maillasson, C. Charrier et al., IL-34 and M-CSF form a novel heteromeric cytokine and regulate the M-CSF receptor activation and localization, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-01644778

M. Baud'huin, R. R. Charrier, C. Riet, A. Moreau, A. Brion et al., Interleukin-34 is expressed by giant cell tumours of bone and plays a key role in RANKL-induced osteoclastogenesis, J Pathol, vol.221, issue.1, pp.77-86, 2010.

Y. Wang, K. J. Szretter, W. Vermi, S. Gilfillan, C. Rossini et al., IL-34 is a tissue-restricted ligand of CSF1R required for the development of Langerhans cells and microglia, Nat Immunol, vol.13, issue.8, pp.753-760, 2012.

M. Chemel, L. Goff, B. Brion, R. Cozic, C. Berreur et al., Interleukin 34 expression is associated with synovitis severity in rheumatoid arthritis patients, Ann Rheum Dis, vol.71, issue.1, pp.150-154, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00667486

V. Garceau, J. Smith, I. R. Paton, M. Davey, M. A. Fares et al., Pivotal Advance: Avian colonystimulating factor 1 (CSF-1), interleukin-34 (IL-34), and CSF-1 receptor genes and gene products, J Leukoc Biol, vol.87, issue.5, pp.753-764, 2010.
DOI : 10.1189/jlb.0909624

URL : https://jlb.onlinelibrary.wiley.com/doi/pdf/10.1189/jlb.0909624

C. J. Sherr, C. W. Rettenmier, R. Sacca, M. F. Roussel, A. T. Look et al., The c-fms proto-oncogene product is related to the receptor for the mononuclear phagocyte growth factor, CSF-1, Cell, vol.41, issue.3, pp.665-676, 1985.

X. Chen, H. Liu, P. J. Focia, A. H. Shim, and X. He, Structure of macrophage colony stimulating factor bound to FMS: diverse signaling assemblies of class III receptor tyrosine kinases, Proc Natl Acad Sci, vol.105, issue.47, pp.18267-18272, 2008.

W. Li and E. R. Stanley, Role of dimerization and modification of the CSF-1 receptor in its activation and internalization during the CSF-1 response, EMBO J, vol.10, issue.2, pp.277-288, 1991.

K. P. Macdonald, V. Rowe, H. M. Bofinger, R. Thomas, T. Sasmono et al., The colony-stimulating factor 1 receptor is expressed on dendritic cells during differentiation and regulates their expansion, J Immunol, vol.175, issue.3, pp.1399-1405, 2005.

R. T. Sasmono and E. Williams, Generation and characterization of MacGreen mice, the Cfs1r-EGFP transgenic mice, Methods Mol Biol, vol.844, pp.157-176, 2012.

F. Ginhoux, F. Tacke, V. Angeli, M. Bogunovic, M. Loubeau et al., Langerhans cells arise from monocytes in vivo, Nat Immunol, vol.7, issue.3, pp.265-273, 2006.

A. Takashima, D. Edelbaum, T. Kitajima, R. K. Shadduck, G. L. Gilmore et al., Colony-stimulating factor-1 secreted by fibroblasts promotes the growth of dendritic cell lines (XS series) derived from murine epidermis, J Immunol, vol.154, issue.10, pp.5128-5135, 1995.

A. H. Baker, S. A. Ridge, T. Hoy, P. G. Cachia, D. Culligan et al., Expression of the colony-stimulating factor 1 receptor in B lymphocytes, Oncogene, vol.8, issue.2, pp.371-378, 1993.

T. Inaba, T. Gotoda, H. Shimano, M. Shimada, K. Harada et al., Platelet-derived growth factor induces c-fms and scavenger receptor genes in vascular smooth muscle cells, J Biol Chem, vol.267, issue.18, pp.13107-13112, 1992.

W. Hofstetter, A. Wetterwald, M. C. Cecchini, R. Felix, H. Fleisch et al., Detection of transcripts for the receptor for macrophage colony-stimulating factor, c-fms, in murine osteoclasts, Proc Natl Acad Sci, vol.89, issue.20, pp.9637-9641, 1992.

E. P. Leonard, W. R. Cotton, and H. J. Keene, Morphological and histochemical observations on the lack of osteoclasis in the "tl" strain of rat, Proc Soc Exp Biol Med, vol.147, issue.3, pp.596-598, 1974.

X. M. Dai, G. R. Ryan, A. J. Hapel, M. G. Dominguez, R. G. Russell et al., Targeted disruption of the mouse colony-stimulating factor 1 receptor gene results in osteopetrosis, mononuclear phagocyte deficiency, increased primitive progenitor cell frequencies, and reproductive defects, Blood, vol.99, issue.1, pp.111-120, 2002.

E. K. Shin, S. H. Lee, S. H. Cho, S. Jung, S. H. Yoon et al., Association between colony-stimulating factor 1 receptor gene polymorphisms and asthma risk, Hum Genet, vol.128, issue.3, pp.293-302, 2010.
DOI : 10.1007/s00439-010-0850-3

URL : https://link.springer.com/content/pdf/10.1007%2Fs00439-010-0850-3.pdf

R. J. Alexander, A. Panja, E. Kaplan-liss, L. Mayer, and R. F. Raicht, Expression of growth factor receptor-encoded mRNA by colonic epithelial cells is altered in inflammatory bowel disease, Dig Dis Sci, vol.40, issue.3, pp.485-494, 1995.

M. D. Jose, L. Meur, Y. Atkins, R. C. Chadban, and S. J. , Blockade of macrophage colony-stimulating factor reduces macrophage proliferation and accumulation in renal allograft rejection, Am J Transplant, vol.3, issue.3, pp.294-300, 2003.

H. Ohno, Y. Uemura, H. Murooka, H. Takanashi, T. Tokieda et al., The orally-active and selective c-Fms tyrosine kinase inhibitor Ki20227 inhibits disease progression in a collagen-induced arthritis mouse model, Eur J Immunol, vol.38, issue.1, pp.283-291, 2008.

Y. Uemura, H. Ohno, Y. Ohzeki, H. Takanashi, H. Murooka et al., The selective M-CSF receptor tyrosine kinase inhibitor Ki20227 suppresses experimental autoimmune encephalomyelitis, J Neuroimmunol, vol.195, issue.1-2, pp.73-80, 2008.

K. P. Macdonald, J. S. Palmer, S. Cronau, E. Seppanen, S. Olver et al., An antibody against the colony-stimulating factor 1 receptor depletes the resident subset of monocytes and tissue-and tumor-associated macrophages but does not inhibit inflammation, Blood, vol.116, pp.3955-3963, 2010.

D. Hashimoto, A. Chow, M. Greter, Y. Saenger, W. H. Kwan et al., Pretransplant CSF-1 therapy expands recipient macrophages and ameliorates GVHD after allogeneic hematopoietic cell transplantation, J Exp Med, vol.208, issue.5, pp.1069-1082, 2011.
DOI : 10.1084/jem.20101709

URL : http://jem.rupress.org/content/208/5/1069.full.pdf

H. Liu, C. Leo, X. Chen, B. R. Wong, L. T. Williams et al., The mechanism of shared but distinct CSF-1R signaling by the non-homologous cytokines IL-34 and CSF-1, Biochim Biophys Acta, vol.1824, issue.7, pp.938-945, 2012.

T. Chihara, S. Suzu, R. Hassan, N. Chutiwitoonchai, M. Hiyoshi et al., IL-34 and M-CSF share the receptor Fms but are not identical in biological activity and signal activation, Cell Death Differ, vol.17, issue.12, pp.1917-1927, 2010.

S. Cohen, O. Y. Shoshana, E. Zelman-toister, N. Maharshak, I. Binsky-ehrenreich et al., The cytokine midkine and its receptor RPTPzeta regulate B cell survival in a pathway induced by CD74, Journal of immunology, vol.188, issue.1, pp.259-269, 2012.

Z. Diamantopoulou, P. Kitsou, S. Menashi, J. Courty, and P. Katsoris, Loss of receptor protein tyrosine phosphatase beta/zeta (RPTPbeta/zeta) promotes prostate cancer metastasis, J Biol Chem, vol.287, issue.48, pp.40339-40349, 2012.

Z. J. Feng, S. B. Gao, Y. Wu, X. F. Xu, X. Hua et al., Lung cancer cell migration is regulated via repressing growth factor PTN/RPTP beta/zeta signaling by menin, Oncogene, vol.29, issue.39, pp.5416-5426, 2010.
DOI : 10.1038/onc.2010.282

URL : https://www.nature.com/articles/onc2010282.pdf

S. Muller, P. Kunkel, K. Lamszus, U. Ulbricht, G. A. Lorente et al., A role for receptor tyrosine phosphatase zeta in glioma cell migration, Oncogene, vol.22, issue.43, pp.6661-6668, 2003.

U. Ulbricht, M. A. Brockmann, A. Aigner, C. Eckerich, S. Muller et al., Expression and function of the receptor protein tyrosine phosphatase zeta and its ligand pleiotrophin in human astrocytomas, J Neuropathol Exp Neurol, vol.62, issue.12, pp.1265-1275, 2003.

M. D. Sanchez-nino, A. B. Sanz, and A. Ortiz, Chronicity following ischaemia-reperfusion injury depends on tubularmacrophage crosstalk involving two tubular cell-derived CSF-1R activators: CSF-1 and IL-34, Nephrol Dial Transplant, vol.31, issue.9, pp.1409-1416, 2016.

N. Maeda, T. Nishiwaki, T. Shintani, H. Hamanaka, and M. Noda, 6B4 proteoglycan/phosphacan, an extracellular variant of receptor-like protein-tyrosine phosphatase zeta/RPTPbeta, binds pleiotrophin/heparin-binding growth-associated molecule (HB-GAM), J Biol Chem, vol.271, issue.35, pp.21446-21452, 1996.

E. Peles, M. Nativ, P. L. Campbell, T. Sakurai, R. Martinez et al., The carbonic anhydrase domain of receptor tyrosine phosphatase beta is a functional ligand for the axonal cell recognition molecule contactin, Cell, vol.82, issue.2, pp.251-260, 1995.

P. Milev, A. Chiba, M. Haring, H. Rauvala, M. Schachner et al., High affinity binding and overlapping localization of neurocan and phosphacan/proteintyrosine phosphatase-zeta/beta with tenascin-R, amphoterin, and the heparin-binding growth-associated molecule, J Biol Chem, vol.273, issue.12, pp.6998-7005, 1998.

M. Palaiologou, I. Delladetsima, and D. Tiniakos, CD138 (syndecan-1) expression in health and disease, Histol Histopathol, vol.29, issue.2, pp.177-189, 2014.

Y. Yang, S. Yaccoby, W. Liu, J. K. Langford, C. Y. Pumphrey et al., Soluble syndecan-1 promotes growth of myeloma tumors in vivo, Blood, vol.100, issue.2, pp.610-617, 2002.

P. Orecchia, R. Conte, E. Balza, A. Petretto, P. Mauri et al., A novel human anti-syndecan-1 antibody inhibits vascular maturation and tumour growth in melanoma, 2013.

, Eur J Cancer, vol.49, issue.8, pp.2022-2033

T. Goldmann, F. Otto, and E. Vollmer, A receptor-type protein tyrosine phosphatase PTP zeta is expressed in human cutaneous melanomas, Folia Histochem Cytobiol, vol.38, issue.1, pp.19-20, 2000.

J. R. Conejo, J. Kleeff, A. Koliopanos, K. Matsuda, Z. W. Zhu et al.,

H. Buchler and M. W. , Syndecan-1 expression is upregulated in pancreatic but not in other gastrointestinal cancers, Int J Cancer, vol.88, issue.1, pp.12-20, 2000.

R. Takazaki, Y. Shishido, R. Iwamoto, and E. Mekada, Suppression of the biological activities of the epidermal growth factor (EGF)-like domain by the heparin-binding domain of heparin-binding EGF-like Growth Factor, J Biol Chem, vol.279, issue.45, pp.47335-47343, 2004.

L. E. Kemp, B. Mulloy, and E. Gherardi, Signalling by HGF/ SF and Met: the role of heparan sulphate co-receptors, Biochem Soc Trans, vol.34, pp.414-417, 2006.

J. Dai and A. B. Rabie, VEGF: an essential mediator of both angiogenesis and endochondral ossification, J Dent Res, vol.86, issue.10, pp.937-950, 2007.

C. Niehrs, The complex world of WNT receptor signalling, Nat Rev Mol Cell Biol, vol.13, issue.12, pp.767-779, 2012.

C. C. Rider, Heparin/heparan sulphate binding in the TGF-beta cytokine superfamily, Biochem Soc Trans, vol.34, pp.458-460, 2006.

S. Bezie, E. Picarda, J. Ossart, L. Tesson, C. Usal et al., IL-34 is a Treg-specific cytokine and mediates transplant tolerance, J Clin Invest, vol.125, issue.10, pp.3952-3964, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-02148022

C. Guillonneau, M. Hill, F. X. Hubert, E. Chiffoleau, C. Herve et al., CD40Ig treatment results in allograft acceptance mediated by CD8CD45RC T cells, IFN-gamma, and indoleamine 2,3-dioxygenase, J Clin Invest, vol.117, issue.4, pp.1096-1106, 2007.

E. A. Bostrom and P. Lundberg, The newly discovered cytokine IL-34 is expressed in gingival fibroblasts, shows enhanced expression by pro-inflammatory cytokines, and stimulates osteoclast differentiation, PLoS One, vol.8, issue.12, p.81665, 2013.

Y. Yu, D. Yang, L. Qiu, H. Okamura, J. Guo et al., Tumor necrosis factor-alpha induces interleukin-34 expression through nuclear factorkappaB activation in MC3T3-E1 osteoblastic cells, Mol Med Rep, vol.10, issue.3, pp.1371-1376, 2014.

Z. Chen, K. Buki, J. Vaaraniemi, G. Gu, and H. K. Vaananen, The critical role of IL-34 in osteoclastogenesis, PLoS One, vol.6, issue.4, p.18689, 2011.

S. J. Moon, Y. S. Hong, J. H. Ju, S. K. Kwok, S. H. Park et al., Increased levels of interleukin 34 in serum and synovial fluid are associated with rheumatoid factor and anticyclic citrullinated peptide antibody titers in patients with rheumatoid arthritis, J Rheumatol, vol.40, issue.11, pp.1842-1849, 2013.

F. Zhang, R. Ding, P. Li, C. Ma, D. Song et al., Interleukin-34 in rheumatoid arthritis: potential role in clinical therapy, Int J Clin Exp Med, vol.8, issue.5, pp.7809-7815, 2015.

S. Yang, S. Jiang, Y. Wang, S. Tu, Z. Wang et al., Interleukin 34 upregulation contributes to the increment of micro-RNA 21 expression through STAT3 activation associated with disease activity in rheumatoid arthritis, J Rheumatol, vol.43, issue.7, pp.1312-1319, 2016.

S. J. Hwang, B. Choi, S. S. Kang, J. H. Chang, Y. G. Kim et al., Interleukin-34 produced by human fibroblast-like synovial cells in rheumatoid arthritis supports osteoclastogenesis, Arthritis Res Ther, vol.14, issue.1, p.14, 2012.

Y. Tian, H. Shen, L. Xia, and J. Lu, Elevated serum and synovial fluid levels of interleukin-34 in rheumatoid arthritis: possible association with disease progression via interleukin-17 production, J Interferon Cytokine Res, vol.33, issue.7, pp.398-401, 2013.

S. H. Chang, B. Y. Choi, J. Choi, J. J. Yoo, Y. J. Ha et al., Baseline serum interleukin-34 levels independently predict radiographic progression in patients with rheumatoid arthritis, Rheumatol Int, vol.35, issue.1, pp.71-79, 2015.

R. Ding, P. Li, D. Song, X. Zhang, and L. Bi, Predictors of response to TNF-alpha antagonist therapy in Chinese rheumatoid arthritis, Clin Rheumatol, vol.34, issue.7, pp.1203-1210, 2015.

Y. Nakamichi, N. Udagawa, and N. Takahashi, IL-34 and CSF-1: similarities and differences, J Bone Miner Metab, vol.31, issue.5, pp.486-495, 2013.
DOI : 10.1007/s00774-013-0476-3

E. L. Masteller and B. R. Wong, Targeting IL-34 in chronic inflammation, Drug Discov Today, vol.19, issue.8, pp.1212-1216, 2014.
DOI : 10.1016/j.drudis.2014.05.016

URL : https://doi.org/10.1016/j.drudis.2014.05.016

S. Garcia, L. M. Hartkamp, B. Malvar-fernandez, I. E. Van-es, H. Lin et al., Colonystimulating factor (CSF) 1 receptor blockade reduces inflammation in human and murine models of rheumatoid arthritis, Arthritis Res Therapy, vol.18, p.75, 2016.

F. Ciccia, A. R. Rodolico, V. Guggino, G. Raimondo, S. Guarnotta et al., IL-34 is overexpressed in the inflamed salivary glands of patients with Sjogren's syndrome and is associated with the local expansion of pro-inflammatory CD14(bright) CD16 + monocytes, Rheumatology, vol.52, issue.6, pp.1009-1017, 2013.

L. Zhou, H. Braat, K. N. Faber, G. Dijkstra, and M. P. Peppelenbosch, Monocytes and their pathophysiological role in Crohn's disease, Cell Mol Life Sci, vol.66, issue.2, pp.192-202, 2009.

E. Franze, I. Monteleone, M. L. Cupi, P. Mancia, F. Caprioli et al., Interleukin-34 sustains inflammatory pathways in the gut, Clin Sci, vol.129, issue.3, pp.271-280, 2015.

S. Zwicker, G. L. Martinez, M. Bosma, M. Gerling, R. Clark et al., Interleukin 34: a new modulator of human and experimental inflammatory bowel disease, Clin Sci, vol.129, issue.3, pp.281-290, 2015.

S. Tanida, K. Ozeki, T. Mizoshita, H. Tsukamoto, T. Katano et al., Managing refractory Crohn's disease: challenges and solutions, Clin Exp Gastroenterol, vol.8, pp.131-140, 2015.
DOI : 10.2147/ceg.s61868

URL : https://www.dovepress.com/getfile.php?fileID=24554

J. E. Below, E. R. Gamazon, J. V. Morrison, A. Konkashbaev, A. Pluzhnikov et al., , 2011.

, Genome-wide association and meta-analysis in populations from Starr County, Texas, and Mexico City identify type 2 diabetes susceptibility loci and enrichment for expression quantitative trait loci in top signals, Diabetologia, vol.54, issue.8, pp.2047-2055

E. J. Chang, S. K. Lee, Y. S. Song, Y. J. Jang, H. S. Park et al., IL-34 is associated with obesity, chronic inflammation, and insulin resistance, J Clin Endocrinol Metab, vol.99, issue.7, 2014.
DOI : 10.1210/jc.2013-4409

URL : https://academic.oup.com/jcem/article-pdf/99/7/E1263/17637988/jcem1263.pdf

K. Zorena, O. Jachimowicz-duda, and P. Waz, The cut-off value for interleukin 34 as an additional potential inflammatory biomarker for the prediction of the risk of diabetic complications, Biomarkers, vol.21, issue.3, pp.276-282, 2016.

P. Marchetti, Islet inflammation in type 2 diabetes, Diabetologia, vol.59, issue.4, pp.668-672, 2016.

P. M. Moraes-vieira, A. Castoldi, P. Aryal, K. Wellenstein, O. D. Peroni et al., Antigen presentation and T-cell activation are critical for RBP4-induced insulin resistance, Diabetes, vol.65, issue.5, pp.1317-1327, 2016.

D. Huynh, D. Akcora, J. Malaterre, C. K. Chan, X. M. Dai et al., CSF-1 receptordependent colon development, homeostasis and inflammatory stress response, PLoS One, vol.8, issue.2, p.56951, 2013.

Y. Wang and M. Colonna, Interkeukin-34, a cytokine crucial for the differentiation and maintenance of tissue resident macrophages and Langerhans cells, Eur J Immunol, vol.44, issue.6, pp.1575-1581, 2014.

F. K. Swirski, M. Nahrendorf, M. Etzrodt, M. Wildgruber, V. Cortez-retamozo et al., Identification of splenic reservoir monocytes and their deployment to inflammatory sites, Science, vol.325, issue.5940, pp.612-616, 2009.

A. I. Segaliny, A. Mohamadi, B. Dizier, A. Lokajczyk, R. Brion et al., Interleukin-34 promotes tumor progression and metastatic process in osteosarcoma through induction of angiogenesis and macrophage recruitment, Int J Cancer, vol.137, issue.1, pp.73-85, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-01644787

T. Chen, X. Wang, L. Guo, M. Wu, Z. Duan et al., Embryonic stem cells promoting macrophage survival and function are crucial for teratoma development, Front Immunol, vol.5, p.275, 2014.

D. G. Denardo, D. J. Brennan, E. Rexhepaj, B. Ruffell, S. L. Shiao et al., Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy, Cancer Discov, vol.1, issue.1, pp.54-67, 2011.

S. L. Zhou, Z. Q. Hu, Z. J. Zhou, Z. Dai, Z. Wang et al., miR-28-5p-IL-34-macrophage feedback loop modulates hepatocellular carcinoma metastasis, Hepatology, vol.63, issue.5, pp.1560-1575, 2016.

L. Preisser, C. Miot, L. Guillou-guillemette, H. Beaumont, E. Foucher et al., IL-34 and macrophage colonystimulating factor are overexpressed in hepatitis C virus fibrosis and induce profibrotic macrophages that promote collagen synthesis by hepatic stellate cells, Hepatology, vol.60, issue.6, pp.1879-1890, 2014.

G. Yu, Y. Bing, S. Zhu, W. Li, L. Xia et al., Activation of the interleukin-34 inflammatory pathway in response to influenza A virus infection, Am J Med Sci, vol.349, issue.2, pp.145-150, 2015.

L. Gerngross and T. Fischer, Evidence for cFMS signaling in HIV production by brain macrophages and microglia, J Neurovirol, vol.21, issue.3, pp.249-256, 2015.

R. Xu, H. F. Sun, D. W. Williams, A. V. Jones, A. Al-hussaini et al., IL-34 suppresses Candida albicans induced TNFalpha production in M1 macrophages by downregulating expression of Dectin-1 and TLR2, J Immunol Res, p.328146, 2015.

H. Esaki, D. A. Ewald, B. Ungar, M. Rozenblit, X. Zheng et al., Identification of novel immune and barrier genes in atopic dermatitis by means of laser capture microdissection, J Allergy Clin Immunol, vol.135, issue.1, pp.153-163, 2015.

M. Ohgidani, T. A. Kato, D. Setoyama, N. Sagata, R. Hashimoto et al., Direct induction of ramified microglia-like cells from human monocytes: dynamic microglial dysfunction in Nasu-Hakola disease, Sci Rep, vol.4, p.4957, 2014.

D. Ma, Y. Doi, J. S. Li, E. Sonobe, Y. Takeuchi et al., TGF-beta induced by interleukin-34-stimulated microglia regulates microglial proliferation and attenuates oligomeric amyloid beta neurotoxicity, Neurosci Lett, vol.529, issue.1, pp.86-91, 2012.

T. Mizuno, Y. Doi, H. Mizoguchi, J. S. Noda, M. Sonobe et al., Interleukin-34 selectively enhances the neuroprotective effects of microglia to attenuate oligomeric amyloid-beta neurotoxicity, Am J Pathol, vol.179, issue.4, pp.2016-2027, 2011.

S. Jin, Y. Sonobe, J. Kawanokuchi, H. Horiuchi, Y. Cheng et al., Interleukin-34 restores blood-brain barrier integrity by upregulating tight junction proteins in endothelial cells, PLoS One, vol.9, issue.12, p.115981, 2014.

J. Luo, F. Elwood, M. Britschgi, S. Villeda, H. Zhang et al., Colony-stimulating factor 1 receptor (CSF1R) signaling in injured neurons facilitates protection and survival, J Exp Med, vol.210, issue.1, pp.157-172, 2013.

M. Greter, I. Lelios, P. Pelczar, G. Hoeffel, J. Price et al., Stroma-derived interleukin-34 controls the development and maintenance of langerhans cells and the maintenance of microglia, Immunity, vol.37, issue.6, pp.1050-1060, 2012.

J. I. Kim and L. A. Turka, Transplant tolerance: a new role for IL-34, J Clin Invest, vol.125, issue.10, pp.3751-3753, 2015.

P. Conde, M. Rodriguez, W. Van-der-touw, A. Jimenez, M. Burns et al., DC-SIGN(+) macrophages control the induction of transplantation tolerance, Immunity, vol.42, issue.6, pp.1143-1158, 2015.

J. K. Edwards, Acute kidney injury: IL-34 promotes persistent ischaemia-induced AKI, Nat Rev Nephrol, vol.11, issue.9, p.504, 2015.
DOI : 10.1038/nrneph.2015.116

Y. Wang, J. Chang, B. Yao, A. Niu, E. Kelly et al., Proximal tubule-derived colony stimulating factor-1 mediates polarization of renal macrophages and dendritic cells, and recovery in acute kidney injury, Kidney Int, vol.88, issue.6, pp.1274-1282, 2015.

M. Z. Zhang, B. Yao, S. Yang, L. Jiang, S. Wang et al., CSF-1 signaling mediates recovery from acute kidney injury, J Clin Invest, vol.122, issue.12, pp.4519-4532, 2012.
DOI : 10.1172/jci60363

T. Masaoka, H. Shibata, R. Ohno, S. Katoh, M. Harada et al., Double-blind test of human urinary macrophage colony-stimulating factor for allogeneic and syngeneic bone marrow transplantation: effectiveness of treatment and 2-year follow-up for relapse of leukaemia, Br J Haematol, vol.76, issue.4, pp.501-505, 1990.

L. Grayfer and J. Robert, Divergent antiviral roles of amphibian (Xenopus laevis) macrophages elicited by colony-stimulating factor-1 and interleukin-34, J Leukoc Biol, vol.96, issue.6, pp.1143-1153, 2014.

L. Grayfer and J. Robert, Distinct functional roles of amphibian (Xenopus laevis) colony-stimulating factor-1-and interleukin-34-derived macrophages, J Leukoc Biol, vol.98, issue.4, pp.641-649, 2015.
DOI : 10.1189/jlb.4ab0315-117rr

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4763865/pdf

E. D. Foucher, S. Blanchard, L. Preisser, P. Descamps, N. Ifrah et al., IL-34-and M-CSF-induced macrophages switch memory T cells into Th17 cells via membrane IL-1alpha, Eur J Immunol, vol.45, issue.4, pp.1092-1102, 2015.
DOI : 10.1002/eji.201444606

URL : https://onlinelibrary.wiley.com/doi/pdf/10.1002/eji.201444606

J. A. Hutchinson, P. Riquelme, B. Sawitzki, S. Tomiuk, P. Miqueu et al., Cutting Edge: Immunological consequences and trafficking of human regulatory macrophages administered to renal transplant recipients, J Immunol, vol.187, issue.5, pp.2072-2078, 2011.

G. Liu, K. Duan, H. Ma, Z. Niu, J. Peng et al., An instructive role of donor macrophages in mixed chimeras in the induction of recipient CD4(+)Foxp3(+) Treg cells, Immunol Cell Biol, vol.89, issue.8, pp.827-835, 2011.

E. D. Foucher, S. Blanchard, L. Preisser, E. Garo, N. Ifrah et al., IL-34 induces the differentiation of human monocytes into immunosuppressive macrophages. antagonistic effects of GM-CSF and IFNgamma, PLoS One, vol.8, issue.2, p.56045, 2013.

X. L. Li, S. Menoret, S. Bezie, L. Caron, D. Chabannes et al., Mechanism and localization of CD8 regulatory T cells in a heart transplant model of tolerance, J Immunol, vol.185, issue.2, pp.823-833, 2010.

E. Picarda, S. Bezie, V. Venturi, K. Echasserieau, E. Merieau et al., , 2014.

, + Tregs and suppresses organ rejection, J Clin Invest, vol.124, issue.6, pp.2497-2512

P. Riquelme, S. Tomiuk, A. Kammler, F. Fandrich, H. J. Schlitt et al., IFN-gamma-induced iNOS expression in mouse regulatory macrophages prolongs allograft survival in fully immunocompetent recipients, Mol Ther, vol.21, issue.2, pp.409-422, 2013.

E. Rietkotter, A. Bleckmann, M. Bayerlova, K. Menck, H. N. Chuang et al., Anti-CSF-1 treatment is effective to prevent carcinoma invasion induced by monocyte-derived cells but scarcely by microglia, Oncotarget, vol.6, issue.17, pp.15482-15493, 2015.

Y. Wang, M. Bugatti, T. K. Ulland, W. Vermi, S. Gilfillan et al., Nonredundant roles of keratinocyte-derived IL-34 and neutrophil-derived CSF1 in langerhans cell renewal in the steady state and during inflammation, Eur J Immunol, vol.46, issue.3, pp.552-559, 2016.

H. Eda, J. Zhang, R. H. Keith, M. Michener, D. R. Beidler et al., Macrophage-colony stimulating factor and interleukin-34 induce chemokines in human whole blood, Cytokine, vol.52, issue.3, pp.215-220, 2010.

J. Felix, J. Elegheert, I. Gutsche, A. V. Shkumatov, Y. Wen et al., Human IL-34 and CSF-1 establish structurally similar extracellular assemblies with their common hematopoietic receptor, Structure, vol.21, issue.4, pp.528-539, 2013.

R. A. Barve, M. D. Zack, D. Weiss, R. H. Song, D. Beidler et al., Transcriptional profiling and pathway analysis of CSF-1 and IL-34 effects on human monocyte differentiation, Cytokine, vol.63, issue.1, pp.10-17, 2013.

F. Yamane, Y. Nishikawa, K. Matsui, M. Asakura, E. Iwasaki et al., CSF-1 receptor-mediated differentiation of a new type of monocytic cell with B cell-stimulating activity: its selective dependence on IL-34, J Leukoc Biol, vol.95, issue.1, pp.19-31, 2014.

S. Nandi, S. Gokhan, X. M. Dai, S. Wei, G. Enikolopov et al., The CSF-1 receptor ligands IL-34 and CSF-1 exhibit distinct developmental brain expression patterns and regulate neural progenitor cell maintenance and maturation, Dev biol, vol.367, issue.2, pp.100-113, 2012.
DOI : 10.1016/j.ydbio.2012.03.026

URL : https://doi.org/10.1016/j.ydbio.2012.03.026

M. Okubo, H. Yamanaka, K. Kobayashi, Y. Dai, H. Kanda et al., Macrophage-colony stimulating factor derived from injured primary afferent induces proliferation of spinal microglia and neuropathic pain in rats, PLoS One, vol.11, issue.4, p.153375, 2016.

S. Garcia, L. M. Hartkamp, B. Malvar-fernandez, I. E. Van-es, H. Lin et al., Colonystimulating factor (CSF) 1 receptor blockade reduces inflammation in human and murine models of rheumatoid arthritis, Arthr Res Ther, vol.18, p.75, 2015.

E. Franze, I. Marafini, D. Simone, V. Monteleone, I. Caprioli et al., Interleukin-34 induces cc-chemokine ligand 20 in gut epithelial cells, J Crohn's Colitis, vol.10, issue.1, pp.87-94, 2016.

H. Shoji, S. Yoshio, Y. Mano, E. Kumagai, M. Sugiyama et al., Interleukin-34 as a fibroblast-derived marker of liver fibrosis in patients with non-alcoholic fatty liver disease, 2016.

T. Wang, T. Kono, M. M. Monte, H. Kuse, M. M. Costa et al., Identification of IL-34 in teleost fish: differential expression of rainbow trout IL-34, MCSF1 and MCSF2, ligands of the MCSF receptor, Mol Immunol, vol.53, issue.4, pp.398-409, 2013.

L. Gerngross, G. Lehmicke, A. Belkadi, and T. Fischer, Role for cFMS in maintaining alternative macrophage polarization in SIV infection: implications for HIV neuropathogenesis, J Neuroinflammation, vol.12, p.58, 2015.

B. Wang, W. Xu, M. Tan, Y. Xiao, H. Yang et al., Integrative genomic analyses of a novel cytokine, interleukin-34 and its potential role in cancer prediction, Intern J Mol Med, vol.35, issue.1, pp.92-102, 2015.

B. E. Booker, R. S. Clark, S. T. Pellom, and S. E. Adunyah, Interleukin-34 induces monocytic-like differentiation in leukemia cell lines, Int J Biochem Mol Biol, vol.6, issue.1, pp.1-16, 2015.