E. Bigot-corbel, M. Gassin, I. Corre, L. Carrer, D. Delaroche et al., Hepatitis C virus (HCV) infection, monoclonal immunoglobulin specific for HCV core protein, and plasma-cell malignancy, Blood, vol.112, issue.10, pp.4357-4365, 2008.
DOI : 10.1182/blood-2008-07-167569

URL : http://www.bloodjournal.org/content/bloodjournal/112/10/4357.full.pdf

C. De-martel, J. Ferlay, S. Franceschi, J. Vignat, F. Bray et al., Global burden of cancers attributable to infections in 2008: a review and synthetic analysis, The Lancet Oncology, vol.13, issue.6, pp.607-622, 2012.
DOI : 10.1016/S1470-2045(12)70137-7

S. Hermouet, I. Corre, M. Gassin, E. Bigot-corbel, C. Sutton et al., Hepatitis C Virus, Human Herpesvirus 8, and the Development of Plasma-Cell Leukemia, New England Journal of Medicine, vol.348, issue.2, pp.178-187, 2003.
DOI : 10.1056/NEJM200301093480219

S. Rajkumar, R. Kyle, M. Plevak, J. Murray, and T. Therneau, Helicobacter pylori infection and monoclonal gammopathy of undetermined significance, British Journal of Haematology, vol.31, issue.3
DOI : 10.1056/NEJM199510123331508

URL : http://onlinelibrary.wiley.com/doi/10.1046/j.1365-2141.2002.03912.x/pdf

A. Saha and E. Robertson, Epstein-Barr Virus-Associated B-cell Lymphomas: Pathogenesis and Clinical Outcomes, Clinical Cancer Research, vol.17, issue.10, pp.3056-63, 2011.
DOI : 10.1158/1078-0432.CCR-10-2578

URL : http://clincancerres.aacrjournals.org/content/clincanres/17/10/3056.full.pdf

B. Weiss, J. Abadie, P. Verma, R. Howard, and W. Kuehl, A monoclonal gammopathy precedes multiple myeloma in most patients, Blood, vol.113, issue.22, pp.5418-5440, 2008.
DOI : 10.1182/blood-2008-12-195008

URL : http://www.bloodjournal.org/content/bloodjournal/113/22/5418.full.pdf

R. Wadhera and S. Rajkumar, Prevalence of Monoclonal Gammopathy of Undetermined Significance: A Systematic Review, Mayo Clinic Proceedings, vol.85, issue.10, pp.933-975, 2010.
DOI : 10.4065/mcp.2010.0337

R. Kyle, T. Therneau, S. Rajkumar, D. Larson, M. Plevak et al., Prevalence of Monoclonal Gammopathy of Undetermined Significance, New England Journal of Medicine, vol.354, issue.13, pp.1362-1371, 2006.
DOI : 10.1056/NEJMoa054494

S. Hermouet, E. Bigot-corbel, and B. Gardie, Pathogenesis of Myeloproliferative Neoplasms: Role and Mechanisms of Chronic Inflammation, Mediators of Inflammation, vol.348, issue.6239, 2015.
DOI : 10.4049/jimmunol.1003438

URL : https://hal.archives-ouvertes.fr/inserm-01402440

M. Boissinot, M. Vilaine, and S. Hermouet, The Hepatocyte Growth Factor (HGF)/Met Axis: A Neglected Target in the Treatment of Chronic Myeloproliferative Neoplasms?, Cancers, vol.79, issue.3, pp.1631-6910, 2014.
DOI : 10.3324/haematol.2014.107656

URL : https://hal.archives-ouvertes.fr/inserm-01401601

A. Bosseboeuf, D. Feron, A. Tallet, C. Rossi, C. Charlier et al., Monoclonal IgG in MGUS and multiple myeloma targets infectious pathogens, JCI Insight, vol.2, issue.19, 2017.
DOI : 10.1172/jci.insight.95367DS1

URL : https://hal.archives-ouvertes.fr/inserm-01628286

M. Dalziel, M. Crispin, C. Scanlan, N. Zitzmann, and R. Dwek, Emerging Principles for the Therapeutic Exploitation of Glycosylation, Science, vol.254, issue.6166, 2014.
DOI : 10.1016/j.jmb.2009.02.033

R. Parekh, I. Roitt, D. Isenberg, R. Dwek, B. Ansell et al., GALACTOSYLATION OF IgG ASSOCIATED OLIGOSACCHARIDES: REDUCTION IN PATIENTS WITH ADULT AND JUVENILE ONSET RHEUMATOID ARTHRITIS AND RELATION TO DISEASE ACTIVITY, The Lancet, vol.331, issue.8592, pp.966-975, 1988.
DOI : 10.1016/S0140-6736(88)91781-3

G. Rook, J. Steele, R. Brealey, A. Whyte, D. Isenberg et al., Changes in IgG glycoform levels are associated with remission of arthritis during pregnancy(91)90173-A 15 Immunoglobulin G galactosylation and sialylation are associated with pregnancy-induced improvement of rheumatoid arthritis and the postpartum flare: results from a large prospective cohort study, J Autoimmun Arthritis Res Ther, vol.4, issue.11, pp.779-94, 1186.

R. Anthony, F. Nimmerjahn, D. Ashline, V. Reinhold, J. Paulson et al., Recapitulation of IVIG Anti-Inflammatory Activity with a Recombinant IgG Fc, Science, vol.310, issue.5753, pp.373-379, 2008.
DOI : 10.1126/science.1118948

R. Anthony, F. Wermeling, M. Karlsson, and J. Ravetch, Identification of a receptor required for the anti-inflammatory activity of IVIG, Proceedings of the National Academy of Sciences, vol.8, issue.6, pp.19571-19579, 2008.
DOI : 10.1038/ni1470

Y. Kaneko, F. Nimmerjahn, and J. Ravetch, Anti-Inflammatory Activity of Immunoglobulin G Resulting from Fc Sialylation, Science, vol.313, issue.5787, pp.670-673, 2006.
DOI : 10.1126/science.1129594

I. Schwab, M. Biburger, G. Krönke, G. Schett, and F. Nimmerjahn, IVIg-mediated amelioration of ITP in mice is dependent on sialic acid and SIGNR1, European Journal of Immunology, vol.310, issue.4, pp.826-856, 2012.
DOI : 10.1126/science.1118948

M. Ackerman, M. Crispin, X. Yu, K. Baruah, A. Boesch et al., Natural variation in Fc glycosylation of HIV-specific antibodies impacts antiviral activity, Journal of Clinical Investigation, vol.123, issue.5, pp.2183-92, 2013.
DOI : 10.1172/JCI65708DS1

F. Nimmerjahn, Activating and inhibitory Fc??Rs in autoimmune disorders, Springer Seminars in Immunopathology, vol.107, issue.4, pp.305-324, 2006.
DOI : 10.4049/jimmunol.166.5.3499

F. Nimmerjahn and J. Ravetch, Divergent Immunoglobulin G Subclass Activity Through Selective Fc Receptor Binding, Science, vol.310, issue.5753, pp.1510-1512, 2005.
DOI : 10.1126/science.1118948

F. Nimmerjahn and J. Ravetch, Fc?? receptors as regulators of immune responses, Nature Reviews Immunology, vol.203, issue.1, pp.34-47, 2008.
DOI : 10.4049/jimmunol.172.11.7186

B. Scallon, S. Tam, S. Mccarthy, A. Cai, and T. Raju, Higher levels of sialylated Fc glycans in immunoglobulin G molecules can adversely impact functionality, Molecular Immunology, vol.44, issue.7, pp.1524-1558, 2007.
DOI : 10.1016/j.molimm.2006.09.005

I. Schwab, S. Mihai, M. Seeling, M. Kasperkiewicz, R. Ludwig et al., Broad requirement for terminal sialic acid residues and Fc??RIIB for the preventive and therapeutic activity of intravenous immunoglobulins in vivo, European Journal of Immunology, vol.16, issue.Suppl 1
DOI : 10.1016/S1074-7613(02)00275-3

R. Anthony, F. Wermeling, and J. Ravetch, Novel roles for the IgG Fc glycan, Annals of the New York Academy of Sciences, vol.293, issue.Suppl 1
DOI : 10.1126/science.1061692

R. Anthony, T. Kobayashi, F. Wermeling, and J. Ravetch, Intravenous gammaglobulin suppresses inflammation through a novel TH2 pathway, Nature, vol.182, issue.7354, pp.110-113, 2011.
DOI : 10.4049/jimmunol.0802870

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3694429/pdf

I. Quast, C. Keller, M. Maurer, J. Giddens, B. Tackenberg et al., Sialylation of IgG Fc domain impairs complement-dependent cytotoxicity, Journal of Clinical Investigation, vol.125, issue.11, pp.4160-70, 2015.
DOI : 10.1172/JCI82695DS1

N. Washburn, I. Schwab, D. Ortiz, N. Bhatnagar, J. Lansing et al., Controlled tetra-Fc sialylation of IVIg results in a drug candidate with consistent enhanced anti-inflammatory activity, Proceedings of the National Academy of Sciences, vol.1, issue.8592, pp.1297-306, 2015.
DOI : 10.1182/blood-2003-01-0023

P. Umaña, J. Jean-mairet, R. Moudry, H. Amstutz, and J. Bailey, Engineered glycoforms of an antineuroblastoma IgG1 with optimized antibody-dependent cellular cytotoxic activity, Nat Biotechnol, vol.17, pp.176-80106179, 1038.

J. Gasdaska, S. Sherwood, J. Regan, and L. Dickey, An afucosylated anti-CD20 monoclonal antibody with greater antibody-dependent cellular cytotoxicity and B-cell depletion and lower complement-dependent cytotoxicity than rituximab, Molecular Immunology, vol.50, issue.3, pp.134-175, 2012.
DOI : 10.1016/j.molimm.2012.01.001

R. Kapur, I. Kustiawan, A. Vestrheim, C. Koeleman, R. Visser et al., A prominent lack of IgG1-Fc fucosylation of platelet alloantibodies in pregnancy, Blood, vol.123, issue.4, pp.471-80, 2014.
DOI : 10.1182/blood-2013-09-527978

C. Oefner, A. Winkler, C. Hess, A. Lorenz, V. Holecska et al., Tolerance induction with T cell???dependent protein antigens induces regulatory sialylated IgGs, Journal of Allergy and Clinical Immunology, vol.129, issue.6, pp.1647-55, 2012.
DOI : 10.1016/j.jaci.2012.02.037

C. Hess, A. Winkler, A. Lorenz, V. Holecska, V. Blanchard et al., T cell???independent B cell activation induces immunosuppressive sialylated IgG antibodies, Journal of Clinical Investigation, vol.123, issue.9, pp.3788-96, 2013.
DOI : 10.1172/JCI65938DS1

URL : http://www.jci.org/articles/view/65938/files/pdf

M. Jones, D. Oswald, S. Joshi, S. Whiteheart, R. Orlando et al., B-cell???independent sialylation of IgG, Proceedings of the National Academy of Sciences, vol.7, issue.13, pp.7207-7219, 2016.
DOI : 10.1021/ac5020996

S. Fleming, S. Smith, D. Knowles, A. Skillen, and C. Self, Increased sialylation of oligosaccharides on IgG paraproteins--a potential new tumour marker in multiple myeloma, Journal of Clinical Pathology, vol.51, issue.11, pp.825-855, 1998.
DOI : 10.1136/jcp.51.11.825

T. Nishiura, S. Fujii, Y. Kanayama, A. Nishikawa, Y. Tomiyama et al., Carbohydrate analysis of immunoglobulin G myeloma proteins by lectin and

G. Ouest, R. Pays-de-la-loire, and J. Project, ); and by a grant from the Cancéropôle Grand Ouest and Région Centre, to EP The Cancéropôle Grand Ouest and Région Pays de la Loire financed the salary of AB (2015?2016). sUPPleMenTarY MaTerial The Supplementary Material for this article can be found online at http, pp.2015-2017, 2015.

S. Mittermayr, G. Lê, C. Clarke, M. Martín, S. Larkin et al., -Glycosylation in the Pathogenesis of Plasma Cell Disorders, Journal of Proteome Research, vol.16, issue.2, pp.748-62, 2017.
DOI : 10.1021/acs.jproteome.6b00768

D. Feron, C. Charlier, V. Gourain, L. Garderet, M. Coste-burel et al., Multiplexed infectious protein microarray immunoassay suitable for the study of the specificity of monoclonal immunoglobulins, Analytical Biochemistry, vol.433, issue.2, pp.202-211, 2013.
DOI : 10.1016/j.ab.2012.10.012

URL : https://hal.archives-ouvertes.fr/inserm-01401584

S. Malard-castagnet, E. Dugast, N. Degauque, A. Pallier, J. Soulillou et al., Sialylation of antibodies in kidney recipients with de novo donor specific antibody, with or without antibody mediated rejection, Human Immunology, vol.77, issue.11, pp.1076-83, 2016.
DOI : 10.1016/j.humimm.2015.10.021

A. Williamson, M. Salaman, and H. Kreth, MICROHETEROGENEITY AND ALLOMORPHISM OF PROTEINS, Annals of the New York Academy of Sciences, vol.16, issue.23, pp.210-234, 1973.
DOI : 10.1073/pnas.67.3.1398

F. Cornell, Isoelectric focusing, blotting and probing methods for detection and identification of monoclonal proteins, Clin Biochem Rev, vol.30, pp.123-153, 2009.

S. Nair, A. Branagan, J. Liu, C. Boddupalli, P. Mistry et al., Clonal Immunoglobulin against Lysolipids in the Origin of Myeloma, New England Journal of Medicine, vol.374, issue.6, pp.555-61, 2016.
DOI : 10.1056/NEJMoa1508808

T. Wang, J. Sewatanon, M. Memoli, J. Wrammert, S. Bournazos et al., IgG antibodies to dengue enhanced for Fc??RIIIA binding determine disease severity, Science, vol.16, issue.6323, pp.395-403, 2017.
DOI : 10.1186/s12879-015-1022-9

M. Nakano, R. Saldanha, A. Göbel, M. Kavallaris, and N. Packer, Identification of Glycan Structure Alterations on Cell Membrane Proteins in Desoxyepothilone B Resistant Leukemia Cells, Molecular & Cellular Proteomics, vol.5, issue.11, 2011.
DOI : 10.1016/0006-291X(80)90779-2

K. Hata, T. Tochigi, I. Sato, S. Kawamura, K. Shiozaki et al., Increased sialidase activity in serum of cancer patients: Identification of sialidase and inhibitor activities in human serum, Cancer Science, vol.16, issue.4, pp.383-392, 2015.
DOI : 10.1016/0167-5699(95)80032-8

T. Miyagi, T. Wada, K. Yamaguchi, K. Shiozaki, I. Sato et al., Human sialidase as a cancer marker, PROTEOMICS, vol.98, issue.16, pp.3303-3314, 2008.
DOI : 10.1111/j.1349-7006.1990.tb02692.x

C. Barrios, J. Zierer, I. Gudelj, J. ?tambuk, I. Ugrina et al., Glycosylation Profile of IgG in Moderate Kidney Dysfunction, Journal of the American Society of Nephrology, vol.27, issue.3, pp.933-974, 2016.
DOI : 10.1681/ASN.2015010109

A. Ahmed, J. Giddens, A. Pincetic, J. Lomino, J. Ravetch et al., Structural Characterization of Anti-Inflammatory Immunoglobulin G Fc Proteins, Journal of Molecular Biology, vol.426, issue.18, pp.3166-79, 2014.
DOI : 10.1016/j.jmb.2014.07.006

P. Sondermann, A. Pincetic, J. Maamary, K. Lammens, and J. Ravetch, General mechanism for modulating immunoglobulin effector function, Proceedings of the National Academy of Sciences, vol.287, issue.37, pp.9868-72, 2013.
DOI : 10.1074/jbc.C112.397059

URL : http://www.pnas.org/content/110/24/9868.full.pdf

B. Fiebiger, J. Maamary, A. Pincetic, and J. Ravetch, Protection in antibody- and T cell-mediated autoimmune diseases by antiinflammatory IgG Fcs requires type II FcRs, Proceedings of the National Academy of Sciences, vol.14, issue.10, pp.2385-94, 2015.
DOI : 10.1038/nprot.2006.285

C. Musolino, A. A. Profita, M. Alonci, A. Saitta, S. Russo et al., Reduced IL-33 plasma levels in multiple myeloma patients are associated with more advanced stage of disease, British Journal of Haematology, vol.86, issue.5, pp.709-719, 2013.
DOI : 10.2741/2335

M. Zheng, Z. Zhang, K. Bemis, A. Belch, L. Pilarski et al., The Systemic Cytokine Environment Is Permanently Altered in Multiple Myeloma, PLoS ONE, vol.20, issue.3, 2013.
DOI : 10.1371/journal.pone.0058504.t009

A. Jurczyszyn, J. Czepiel, G. Biesiada, J. Gdula-argasi?ska, D. Cibor et al., Play a Significant Role in the Progression of Multiple Myeloma, Journal of Cancer, vol.5, issue.7, pp.518-542, 2014.
DOI : 10.7150/jca.9266

C. Seidel, M. Børset, I. Turesson, N. Abildgaard, A. Sundan et al., Elevated serum concentrations of hepatocyte growth factor in patients with multiple myeloma. The Nordic Myeloma Study Group, Blood, vol.91, pp.806-818, 1998.

X. Wang, Q. Shi, N. Shah, C. Heijnen, E. Cohen et al., Inflammatory Markers and Development of Symptom Burden in Patients with Multiple Myeloma during Autologous Stem Cell Transplantation, Clinical Cancer Research, vol.20, issue.5, pp.1366-74, 2014.
DOI : 10.1158/1078-0432.CCR-13-2442

O. Hjertner, M. Torgersen, C. Seidel, H. Hjorth-hansen, A. Waage et al., Hepatocyte growth factor (HGF) induces interleukin-11 secretion from osteoblasts: a possible role for HGF in myeloma-associated osteolytic bone disease, Blood, vol.94, pp.3883-3891, 1999.

T. Diamond, S. Levy, A. Smith, P. Day, and A. Manoharan, Non-invasive markers of bone turnover and plasma cytokines differ in osteoporotic patients with multiple myeloma and monoclonal gammopathies of undetermined significance, Internal Medicine Journal, vol.15, issue.5, 2001.
DOI : 10.1046/j.1365-2141.1999.01651.x

M. Dong and G. Blobe, Role of transforming growth factor-beta in hematologic malignancies, Blood, vol.107, issue.12, pp.4589-9610, 2006.
DOI : 10.1182/blood-2005-10-4169

R. Aggarwal, I. Ghobrial, and G. Roodman, Chemokines in multiple myeloma, Experimental Hematology, vol.34, issue.10, pp.1289-95, 2006.
DOI : 10.1016/j.exphem.2006.06.017

T. Hashimoto, M. Abe, T. Oshima, H. Shibata, S. Ozaki et al., Ability of myeloma cells to secrete macrophage inflammatory protein (MIP)-1? and MIP-1? correlates with lytic bone lesions in patients with multiple myeloma

S. Rajkumar, T. Leong, P. Roche, R. Fonseca, A. Dispenzieri et al., Prognostic value of bone marrow angiogenesis in multiple myeloma, Clin Cancer Res, vol.6, pp.3111-3117, 2000.

O. Sezer, K. Niemöller, J. Eucker, J. C. Kaufmann, O. Zavrski et al., Bone marrow microvessel density is a prognostic factor for survival in patients with multiple myeloma, Annals of Hematology, vol.79, issue.10, pp.574-581, 2000.
DOI : 10.1007/s002770000236

A. Coluccia, T. Cirulli, P. Neri, D. Mangieri, M. Colanardi et al., Validation of PDGFR?? and c-Src tyrosine kinases as tumor/vessel targets in patients with multiple myeloma: preclinical efficacy of the novel, orally available inhibitor dasatinib, Blood, vol.112, issue.4, pp.1346-56, 2007.
DOI : 10.1182/blood-2007-10-116590

R. Pfeifle, T. Rothe, N. Ipseiz, H. Scherer, S. Culemann et al., Regulation of autoantibody activity by the IL-23???TH17 axis determines the onset of autoimmune disease, Nature Immunology, vol.278, issue.1, pp.104-117, 2016.
DOI : 10.1074/jbc.M304103200

T. Han and X. Wang, Leptin and its receptor in hematologic malignancies, Int J Clin Exp Med, vol.8, pp.19840-19849, 2015.

G. Pamuk, M. Demir, F. Harmandar, Y. Yesil, B. Turgut et al., Leptin and resistin levels in serum of patients with hematologic malignancies: correlation with clinical characteristics, Exp Oncol, vol.28, pp.241-245, 2006.

J. Reseland, S. Reppe, O. Olstad, H. Hjorth-hansen, A. Brenne et al., Abnormal adipokine levels and leptin-induced changes in gene expression profiles in multiple myeloma, European Journal of Haematology, vol.99, issue.5, 2009.
DOI : 10.4049/jimmunol.171.10.5091

M. Fujimori, E. Franca, T. Morais, V. Fiorin, L. De-abreu et al., Cytokine and adipokine are biofactors can act in blood and colostrum of obese mothers, BioFactors, vol.52, issue.2, pp.243-50, 2017.
DOI : 10.2337/diabetes.52.7.1779

D. Lullo, G. Marcatti, M. Heltai, S. Brunetto, E. Tresoldi et al., Th22 cells increase in poor prognosis multiple myeloma and promote tumor cell growth and survival, OncoImmunology, vol.6, issue.5, 2015.
DOI : 10.1111/j.1365-2559.2006.02375.x