Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Apr 17.
Published in final edited form as: Inflamm Allergy Drug Targets. 2009 Mar;8(1):80–86. doi: 10.2174/187152809787582525

Role of Fc Receptors as a Therapeutic Target

Atsuhiro Masuda 1,4, Masaru Yoshida 1,*, Hideyuki Shiomi 1, Yoshinori Morita 1, Hiromu Kutsumi 1, Hideto Inokuchi 1, Shigeto Mizuno 4, Akira Nakamura 2, Toshiyuki Takai 2, Richard S Blumberg 3, Takeshi Azuma 1
PMCID: PMC3990358  NIHMSID: NIHMS570587  PMID: 19275696

Abstract

It has been forty years since the discovery of Fc Receptors and their function. Fc Receptors include the IgG receptors (FcγR), high-affinity IgE receptor (FcεRI), IgA and IgA/IgM receptors, and neonatal Fc receptor for IgG (FcRn). In particular, the FcγRs have been well known to play an important role in many biologic processes including those associated with the response to infection and cancer as well as in the pathogenesis of immune-mediated diseases. Both positive and negative regulatory function has ascribed to Fc receptors and FcγRs in particular which serve to establish a threshold for immune cell activation. In other cases, Fc receptors such as FcRn possess a novel structure and function by playing a major role in the transport of IgG across polarized epithelial barriers at mucosal surfaces and in the regulation of IgG half-life. These diverse functions highlight the potential effectiveness of targeting Fc receptors for therapeutic purposes. This review summarizes new information available in the therapeutic applications of this biology.

Keywords: Fc receptors, FcRn, IgG, FcγR

INTRODUCTION

Fc receptors, the receptors for the Fc portion of immunoglobulins, play an essential role in antibody-dependent immune responses [1]. Fc receptors are detected on many types of hematopoietic cells including macrophages, neutrophils, dendritic cells, eosinophils, basophils, mast cells, and NK cells [2]. Plasma cells produce five classes of antibodies, IgA, IgD, IgE, IgG and IgM. Fc receptors with an Ig superfamily related structure exist that correspond to each of these classes of immunoglobulins. They include the IgG receptors (FcγR), high-affinity IgE receptor (FcεRI), IgA receptor and polymeric immunoglobulin receptor for IgA and IgM. The second category of Fc receptors is the neonatal Fc receptor for IgG (FcRn), which is a unique FcR that has three major functions with respect to IgG; IgG transport across epithelial barriers, protection of IgG from catabolism and antigen presentation. This review focuses on the functions of the common Fc receptors (FcγRs) and FcRn and summarizes the application of this information to the therapy of human diseases.

THE FAMILY OF FCγ RECEPTORS AND THEIR FUNCTION

Fcγ receptors include four different classes of receptors in mice that are known as FcγRI, FcγRIIB, FcγRIII, FcγRIV. Functionally, these receptors are classified in two types of Fc receptors; those that are activating as opposed to those that are inhibitory. These receptors transmit their signals via immunoreceptor tyrosine-based activation motifs (ITAMs) or immunoreceptor tyrosine-based inhibitory motifs (ITIMs), respectively [3]. Activating FcγRs that possess ITAMs include FcγRI, FcγRIII and FcγRIV. Ligation of these receptors leads to activation of downsteam-signalling pathways. In contrast, the inhibitory FcγR, FcγRIIB, is a unique FcγR that directs an inhibitory program via ITIMs. The composite expression of activating and inhibitory FcγRs regulates the immune response by establishing a threshold for immune cell activation. In many murine models, the aberrant expression of FcγRs can result in uncontrolled immune responses and the initiation of autoimmune diseases [46]. Mice which are deficient in the Fcγ-chain, a subunit that is common to the FcγRI, FcγRIII, FcγRIV, FcεRI and FcαRI receptors exhibit an inability to activate all such FcRs. This results in abrogated or heavily impaired immune complex (IC)-mediated immune responses, such as antibody-dependent cell mediated cytotoxicity (ADCC), release of inflammatory mediators and cytokines, and phagocytosis of ICs [7, 8]. The inhibitory receptor FcγRIIB is the most broadly expressed FcγR, and is present on all leukocytes with the exception of NK cells and T cells. There are two different isoforms of FcγRIIb that are named FcγRIIB-1 and FcγRIIB-2. FcγRIIB-1 is specifically expressed on B cells and negatively regulates B cell activation. In comparison, FcγRIIB-2 is widely expressed on cell types that express FcγRIIB and functions in the inhibition of dendritic cells (DC) and macrophages as manifest by diminished antigen uptake, antigen presentation and cellular activation. Mice deficient in FcγRIIB exhibit spontaneous glomerulonephritis and an enhancement of many types of autoimmune responses. It is believed that the lack of FcγRIIB leads to a breakdown in immunologic tolerance. In humans, the FcγR system is more complex, as exemplified by the existence of the high-affinity IgG receptor FcγRI (FcγRIA, FcγRIB, FcγRIC) and low-affinity IgG receptors FcγRII (FcγRIIA, FcγRIIB and FcγRIIC) and FcγRIII (FcγRIIIA and FcγRIIIB) and the presence of several allelic FcγR variants [9]. FcγRI and FcγRIIB are structurally and functionally similar between human and mice. With the exception of human FcγRIIA and FcγRIIC, activating FcγR’s typically consist of a ligand-binding FcγR α-chain and a signal-transducing γ-chain dimer, which carries immunoreceptor tyrosine based activating motifs (ITAMs). In addition, humans have a glycosylphosphatidylinositol (GPI)-linked receptor that is exclusively expressed by neutrophils, called FcγRIIIB. Moreover, a variety of human FcγR alleles with altered functionality exist. Specifically, FcγRIIA131H and the FcγRIIIA158V have a higher affinity for certain IgG subclasses compared to their allelic counterparts. The FcγRIIB232T variant is unable to associate with lipid rafts and is therefore strongly impaired in its negative regulatory activity. There are many differences between the Fcγ receptors of mice and those of humans. However, observations in mouse have in general mirrored those of human systems.

FC RECEPTORS AND INFECTION

There are many reports describing the role of activating Fc receptors in defending against infection [1012]. We recently reported, for example, on the role of FcγRs in the colonic inflammation induced by infection with Citrobacter rodentium [13]. C. rodentium, a murine model pathogen for enteropathogenic Escherichia coli, specifically colonizes the epithelium of the colon utilizing attaching and effacement structures to adhere to the luminal surface of intestinal epithelial cells and as such cause mucosal inflammation. CD4+ T cells, B cells and IgG, but not secretory IgA or IgM, have been shown to play a critical role in eradicating this pathogen. Therefore, C. rodentium has served as an appropriate model to assess the role of IgG and FcγRs in defending against infections. FcRγ-chain deficient mice, which disables activating FcγRs, are more susceptible to C. rodentium induced colitis. This occurs through a decrease in the efficiency of FcγR-mediated endocytosis and associated maturation of DCs. As a consequence, in the absence of the FcRγ chain, the activation of antigen specific T cells is significantly diminished. Moreover, in the absence of FcγRs, phagocytosis by macrophages is significantly impaired. Therefore, activating FcγRs play an important role in defending against C. rodentium infection supporting a critical role for IgG and the importance of FcγRs in the control of this model of infection. Consistent with this, mice that are deficient in the inhibitory receptor, FcγRIIB, exhibit significantly less inflammation of the distal colon during C. rodentium infection (MY and AM, unpublished observations). Macrophages from FcγRIIB deficient mice display increased phagocytic function in comparison to those obtained from wild type mice. These observations with FcγRIIB mice suggest that targeting this receptor can be envisioned as a means to enhance the function of activating FcγR in the treatment of infectious diseases.

FC RECEPTORS AND CANCER

FcγRs play an important role in determining the therapeutic activity of monoclonal IgG antibodies (mAbs) by their ability to activate the cytotoxic activity of FcγR-positive cells such as NK cells, monocytes, macrophages and neutrophils and by increasing antigen presentation by DC when ligated by the Fc portion of therapeutic antibodies [1417]. Recent studies in Fc receptor-deficient nude mice show that the anti-tumor effects of mAbs such as those directed at-CD20 (Rituximab) and HER2 (Herceptin) require the presence of the signal transducing Fcγ chain that is involved in the activation of FcγRI and FcγRIII receptors that are expressed on monocytes, macrophages, and NK cells [18]. In the B16 metastatic tumor model, FcγR [19] and FcγRIV in particular [20] have been shown to play a significant role in the therapeutic activity of the TA99 antibody specific for the gp75 tumor antigen.

FcγRIIB has been examined in lymphoid tissues and B cell lymphomas suggesting an important role in these contexts. FcγRIIB expression has been described in reactive lymphoid follicles. Mantle cells of secondary follicles exhibit strong plasma membrane expression of FcγRIIB in contrast to the absence of detectable expression in germinal centers and specifically on follicular dendritic cells, tingible bodies macrophages or lymphoid cells. In the interfollicular region of lymph nodes, immunoblasts are negative for FcγRIIB expression in comparison to plasma cells which commonly exhibit strong membrane expression of this receptor [21]. A number of lymphomas however express FcγRIIB [21]. A recent study in mice examined an anti-human FcγRIIB antibody (2B6) in a xenograft model of FcγRIIB expressing lymphomas. The activity of this antibody is Fc-dependent and triggers ADCC. Despite its inhibitory properties, the expression of FcγRIIB on lymphomas led to eradication of the Daudi cell line as a human lymphoma tumor model in a manner that was as effective as targeting CD20 [22]. The outcome of 2B6 infusion in humans with lymphoma remains to be established. Therefore, the ectopic expression of FcγRIIB in human DLBCL (diffuse large cell lymphoma) and follicular lymphomas should be considered as a therapeutic opportunity.

FC RECEPTORS AND AUTOIMMUNE DISEASES

Autoimmune diseases are a complex group of diseases that depend on both genetic and environmental factors for their development. One group of environmental factors that are particularly important are those related to infections and the possibility that they may initiate abnormal immune responses [23, 24]. Consistent with a potential pathogenic origin of autoimmune diseases, recent studies have revealed links between innate immune responses to pathogens and auto-immunity in diseases such as systemic lupus erythematosus [25, 26]. Innate immune responses initiated by pathogens are moreover further linked to adaptive immune responses by a variety of mechanisms. For example, after the activation of innate immune cells such as macrophages, DC, B cells and NK cells by pathogens, the antigen presenting processes that are initiated are important to subsequent T cell activation and B cell maturation including class switching of B cells from IgM to IgG. IgG antibodies including those that are directed at autoantigens play a central pathogenic role in autoimmune diseases. Sequestration of antigen in immune complexes on the surface of follicular dendritic cells may promote class switching, selection of high affinity B cells and control of antibody responses via co-ligation of FcγRIIa and FcγRIIb [27]. Alternations of FcγRs may be linked to autoimmunity in three ways: 1. failure to clear immune complexes from the circulation and from specific sites such as synovial joints and kidneys, 2. hyper-responsiveness to circulating immune complexes through interaction with activating FcγRs and 3. lack of control of antibody production leading to immune complex formation [28].

Anti-inflammatory drugs that inhibit FcγRIIa function are a potentially novel means to block autoimmune disease development early on before the activation of the inflammatory cascade. It has recently shown for example that small molecules and antibody fragments that were designed to bind to the human FcγRIIa could inhibit collagen II induced arthritis in the FcγRIIa transgenic mice [29, 30]. These latter mice develop a spontaneous destructive arthritic disease that is characterized by erythema, swelling and joint ankylosis in up to 50% of animals over 25 weeks of age [31]. The FcγRIIa specific small molecules suppressed disease in the FcγRIIa transgenic mice longer than that achieved with methotrexate, a treatment widely used for rheumatoid arthritis. Synthetic FcR mimetics have also been used to block the function of FcγRII in vitro [32] and the modulation of FcγRIIa and FcγRIIb function in humans [33]. Thus, FcγRII offers a valid target for the treatment of patients with auto-immune disease.

FCRN, THE NEONATAL FC RECEPTOR FOR IGG

FcRn was originally functionally identified in suckling rats as the receptor involved in the transport of IgG (derived from milk) across the intestinal epithelium into the bloodstream [3436]. More recently, it has been shown that FcRn not only delivers IgG across the maternofetal barrier during gestation [37, 38 ] but is also responsible for the maintenance of serum IgG levels [3942]. The gene encoding rat FcRn was first isolated by Simister and Mostov in 1989 [43]. FcRn is structurally related to MHC class I molecules and consists of a heterodimer that is composed of a glycosylated heavy (α) chain that is associated noncovalently with the β2-microglobulin (β2m) light chain [43]. The structural similarity to MHC class I molecules has been confirmed by solution of the X-ray crystallographic structure of the extracellular domains of FcRn together with IgG [44]. FcRn binding of IgG requires three critical amino acids within the CH3–CH2 domain interface of the Fc fragment (Ile253, His310, and His435) in humans and rodents [45]. FcRn homologues have been identified in rat [43], mouse [46], human [47], cow [48], pig [49], and sheep [50]. Although FcRn was originally described as being developmentally regulated in rodent intestine in that its functional expression at birth was notably downregulated within the intestinal epithelium at the time of weaning [39], it has recently been demonstrated that FcRn is also expressed in many human and non-human adult tissues and cell types including hepatocytes [51], endothelial cells [52], a variety of epithelial cell subtypes [5358], monocytes, macrophages, and dendritic cells but not other hematopoetic cell lineages [59]. In various species, these findings have predicted that the functions of FcRn extend to adult mammalian (including human) life.

FcRn has been linked to four major cell biologic pathways: IgG transcytosis across polarized epithelia such as epithelial cells of the placenta [47, 60, 61], intestine [53] and lung [62, 63]; the protection of IgG from catabolism in the circulation [64]; the protection of albumin from catabolism in the circulation as FcRn serves as the albumin receptor [65], and; immune complex mediated antigen presentation in dendritic cells [66]. These functions are driven by the characteristic binding features of IgG to FcRn, allowing for an “on–off” relationship between the receptor (FcRn) and cargo (IgG). IgG binding to FcRn is strongly pH dependent with high-affinity binding at acidic pH (pH<6.5) and weak to no binding at or above neutral pH (pH>7.0), which is consistent with the presence of histidine residues within IgG that are involved in FcRn binding [67]. Using β2m-deficient mice that lack FcRn function [41, 42, 68] and more recently FcRn-deficient mice [69], several groups have shown that FcRn expression is associated with all of these aforementioned functions. At the cellular level, it has been hypothesized that FcRn binds IgG either on the cell surface as driven by the acidic properties of certain cell surfaces (e.g., apical cell surface of epithelia due potentially to the presence of the Na+–H+ exchanger) [70] or in an acidic compartment such as early endosomes [55]. This binding between FcRn and IgG directs monomeric IgG away from degradation in either late endosomes and/or lysosomes with either subsequent recycling into the extracellular milieu (protection function) [71] or to the opposite side of a polarized epithelial cell (transfer function) [7274]. Such a process is important in protecting IgG from catabolism and shuttling IgG across polarized epithelial barriers in the process known as transcytosis. In contrast, binding of FcRn to polymeric IgG antigen-antibody complexes results in the mobilization of these to the lysosomes and enhancement of antigen presentation [66].

FCRN AND INFECTION

The bidirectional transport of IgG confers a unique ability upon FcRn to to be able to retrieve intestinal luminal antigens as a complex with IgG and to deposit these into the intestinal mucosa where the antigen/IgG complexes can be captured by DCs for subsequent presentation to CD4+ T cells either locally or within regional lymphoid structures [75]. These properties of FcRn define a unique mechanism by which absorptive epithelia, which cover the majority of the surface of the intestines, can specifically acquire and transport antigens into the lamina propria and regional lymphoid structures. Consistent with this, recent studies have indicated that intestinal bacterial antigens are required to direct the maturation of immune responses [76] and that such immune responses are induced throughout the intestine rather than within restricted regions such as Peyer’s patches [77,78]. Therefore, these recent observations have raised a potential possibility that epithelial cell-mediated sampling of luminal bacterial antigens throughout the intestinal surface contributes to the regulation of mucosal and systemic immune responses. It is interesting to note that FcRn−/− mice exhibit more body-weight loss and higher bacterial concentrations in the feces at 21 days after C. rodentium infection compared to FcRn−/+ mice [79]. Consistent with these clinical changes, FcRn−/− C57BL/6 mice that were infected with C. rodentium exhibit increased cellular infiltration with mononuclear cells and neutrophils and significantly increased epithelial injury in comparison to that observed in FcRn−/+ mice. These results indicate that FcRn−/− mice, which show an absence of transporting IgG and bacterial antigen/IgG complexes, are more susceptible to C. rodentium-induced colitis. Both innate and acquired immune responses are involved in the pathogenesis of infectious colitis [80, 81]. It has been hypothesized that FcRn also plays a role in infection-induced acquired immune responses by delivering bacteria-derived antigens coupled to specific IgG into mucosal immune cells in addition to potential local, immune protective effects of this receptor in the epithelium. To show this, a genetically engineered C. rodentium strain (C. rodentium-OVA) that constitutively expresses an OVA fragment containing the OT-II and DO11.10 peptides was examined in a transgenic animal expressing mouse FcRn and mouse β2m within the epithelium under the control of an IFABP. It was shown that FcRn-mediated delivery of IgG was required for the effective induction of immune responses to an epithelial pathogen in vivo by the ability to deliver epithelia-associated antigens to host immune cells in the mesenteric lymph nodes as defined by the detection of multiple cell divisions in CFSE-labeled transgenic, OVA-specific T cells [79]. These studies indicate that epithelial FcRn can induce effective CD4+ T cell responses systemically to pathogen-derived antigens associated with the lumen and/or intestinal epithelium when they are retrieved as antigen/IgG complexes.

FCRN AS A THERAPUTIC TARGET

These properties of FcRn have led to the concept of targeting FcRn for therapeutic purposes. Such opportunities would include extension of the serum half-life of therapeutic antibodies and Fc-fusion proteins to improve their efficacy or conversely decreasing the serum level of pathogenic antibodies by inhibiting FcRn function. The latter would be important in antibody mediated diseases such as myasthenia gravis, bullous pemphigoid, idiopathic thrombocytopenic purpura (ITP) and systemic lupus erythematosus (SLE). Alternatively, the transport functions of FcRn have lent themselves to enabling drug delivery across mucosal surfaces. Fc-fusion proteins of erythropoietin (EPO-Fc) have been shown to be able to be delivered across the epithelia of the lungs of mice [58], monkeys [63] and humans [62] in a pathway that is mediated specifically by FcRn since mutation of the critical isoleucine 252, histidine 433 and histidine 435 residues abrogates this transport. The clinical trials with EPO-Fc [62] in humans in particular proves that the FcRn is active at mucosal surfaces in adult human life.

The remainder of this discussion will focus on the manipulation of therapeutic and pathogenic antibodies within the circulation. The half-life of IgG depends on its concentration in the circulation in an inverse manner due to the saturability of the FcRn receptor [82, 83]. The level of FcRn expression controls the serum concentration of IgG such that excess IgG antibodies do not bind to FcRn and enter a degradative pathway. This leads to a shortening of the serum IgG half-life. High-dose intravenous immunoglobulin (IVIG) treatment [84, 85] is thought to exert an immunomodulatory effect by numerous mechanisms, including engagement of the inhibitory FcγRIIb receptor and by FcRn saturation. In mouse models such as bullous pemphigoid, ITP and auto-immune arthritis, IVIG treatment is effective because of the decrease of the pathogenic antibodies. The therapeutic effect for IVIG is maintained in FcγRIIb-deficient mice and its absence in FcRn deficient mice is strong evidence that an important mechanism of action of IVIG is its ability to compromise FcRn function [86, 87].

Another approach to reduce serum pathogenic antibodies is to block the FcRn–IgG interaction using FcRn-specific monoclonal antibodies. A monoclonal antibody against β2m has been reported to block the ability of FcRn to bind IgG in vitro [88] and in vivo [89]. β2m is the common light chain for all MHC class I and many MHC class Ib molecules besides FcRn. Therefore therapy using β2m specific monoclonal antibodies may incur unexpected side effects. Indeed, a monoclonal antibody directed against the FcRn heavy chain has been shown to reduce disease symptoms in rats with experimentally induced myasthenia gravis [90]. Such FcRn specific antibodies and other targeting modalities may be used as therapeutic agents in such contexts in the future. These alternative methods of targeting and inhibiting FcRn include the creation through genetic engineering of antibodies with increased binding to FcRn at both neutral and acidic pH (so-called “Abdegs”) [91, 92] or cyclic peptides with similar properties [93].

CONCLUSION

The FcγRs and FcRn are revealing themselves to be clinically useful targets in the treatment of infection, cancer and autoimmune diseases. These approaches include the blockade of inhibitory FcγRs for enhancing immune responses to infectious pathogens and cancer, blocking activating FcγRs for the treatment of autoimmune diseases and targeting the ectopic expression of particular FcγRs such as occurs in lymphomas for the elimination of tumors. The manipulation of FcRn interactions with antibodies allows for the development of designer antibodies that have particular pH dependent binding characteristics lends themselves to enhancing therapeutic antibody half-life or promoting the degradation of pathogenic antibodies. Finally, co-opting FcRn transport function is a means to enable the mucosal delivery of therapeutic proteins.

Acknowledgments

M.Y. was supported by grants from Nagao Memorial Fund, Grant-in-Aid for Scientific Research, for Scientific Research in Priority Areas ‘Membrane Traffic’ from Ministry of Education, Culture, Sports, Science and Technology of Japan, and Foundation of Advancement of International Science, A.M. was supported by grants from Research Leader Program of Kobe University School of Medicine and Sinryokukai Fund. R.S.B. was supported by grants from the National Institutes of Health (RO1 DK53056, DK51362, DK44319).

References

  • 1.Ravetch JV, Kinet JP. Fc receptors. Annu Rev Immunol. 1991;9:457–92. doi: 10.1146/annurev.iy.09.040191.002325. [DOI] [PubMed] [Google Scholar]
  • 2.Qiu WQ, de Bruin D, Brownstein BH, Pearse R, Ravetch JV. Organization of the human and mouse low-affinity Fc gamma R genes: duplication and recombination. Science. 1990;248:732–5. doi: 10.1126/science.2139735. [DOI] [PubMed] [Google Scholar]
  • 3.Ravetch JV, Lanier LL. Immune inhibitory receptors. Science. 2000;290:84–9. doi: 10.1126/science.290.5489.84. [DOI] [PubMed] [Google Scholar]
  • 4.Bolland S, Ravetch JV. Spontaneous autoimmune disease in Fc(gamma)RIIB-deficient mice results from strain-specific epistasis. Immunity. 2000;13:277–85. doi: 10.1016/s1074-7613(00)00027-3. [DOI] [PubMed] [Google Scholar]
  • 5.Ioan-Facsinay A, de Kimpe SJ, Hellwig SM, van Lent PL, Hofhuis FM, van Ojik HH, Sedlik C, da Silveira SA, Gerber J, de Jong YF, Roozendaal R, Aarden LA, van den Berg WB, Saito T, Mosser D, Amigorena S, Izui S, van Ommen GJ, van Vugt M, van de Winkel JG, Verbeek JS. FcgammaRI (CD64) contributes substantially to severity of arthritis, hypersensitivity responses, and protection from bacterial infection. Immunity. 2002;16:391–402. doi: 10.1016/s1074-7613(02)00294-7. [DOI] [PubMed] [Google Scholar]
  • 6.Sylvestre DL, Ravetch JV. Fc receptors initiate the Arthus reaction: redefining the inflammatory cascade. Science. 1994;265:1095–8. doi: 10.1126/science.8066448. [DOI] [PubMed] [Google Scholar]
  • 7.Nimmerjahn F, Ravetch JV. Fcgamma receptors: old friends and new family members. Immunity. 2006;24:19–28. doi: 10.1016/j.immuni.2005.11.010. [DOI] [PubMed] [Google Scholar]
  • 8.Takai T, Li M, Sylvestre D, Clynes R, Ravetch JV. FcR gamma chain deletion results in pleiotrophic effector cell defects. Cell. 1994;76:519–29. doi: 10.1016/0092-8674(94)90115-5. [DOI] [PubMed] [Google Scholar]
  • 9.Takai T. Roles of Fc receptors in autoimmunity. Nat Rev Immunol. 2002;2:580–92. doi: 10.1038/nri856. [DOI] [PubMed] [Google Scholar]
  • 10.Moore T, Ananaba GA, Bolier J, Bowers S, Belay T, Eko FO, Igietseme JU. Fc receptor regulation of protective immunity against Chlamydia trachomatis. Immunology. 2002;105:213–21. doi: 10.1046/j.0019-2805.2001.01354.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Yoneto T, Waki S, Takai T, Tagawa Y, Iwakura Y, Mizuguchi J, Nariuchi H, Yoshimoto T. A critical role of Fc receptor-mediated antibody-dependent phagocytosis in the host resistance to blood-stage Plasmodium berghei XAT infection. J Immunol. 2001;166:6236–41. doi: 10.4049/jimmunol.166.10.6236. [DOI] [PubMed] [Google Scholar]
  • 12.Onah DN, Uchiyama F, Nagakui Y, Ono M, Takai T, Nawa Y. Mucosal defense against gastrointestinal nematodes: responses of mucosal mast cells and mouse mast cell protease 1 during primary strongyloides venezuelensis infection in FcRgamma-knockout mice. Infect Immun. 2000;68:4968–71. doi: 10.1128/iai.68.9.4968-4971.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Masuda A, Yoshida M, Shiomi H, Ikezawa S, Takagawa T, Tanaka H, Chinzei R, Ishida T, Morita Y, Kutsumi H, Inokuchi H, Wang S, Kobayashi K, Mizuno S, Nakamura A, Takai T, Blumberg RS, Azuma T. Fcgamma receptor regulation of Citrobacter rodentium infection. Infect Immun. 2008;76:1728–37. doi: 10.1128/IAI.01493-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.White CA, Weaver RL, Grillo-Lopez AJ. Antibody-targeted immunotherapy for treatment of malignancy. Annu Rev Med. 2001;52:125–45. doi: 10.1146/annurev.med.52.1.125. [DOI] [PubMed] [Google Scholar]
  • 15.Carter PJ. Potent antibody therapeutics by design. Nat Rev Immunol. 2006;6:343–57. doi: 10.1038/nri1837. [DOI] [PubMed] [Google Scholar]
  • 16.Akiyama K, Ebihara S, Yada A, Matsumura K, Aiba S, Nukiwa T, Takai T. Targeting apoptotic tumor cells to Fc gamma R provides efficient and versatile vaccination against tumors by dendritic cells. J Immunol. 2003;170:1641–8. doi: 10.4049/jimmunol.170.4.1641. [DOI] [PubMed] [Google Scholar]
  • 17.Rafiq K, Bergtold A, Clynes R. Immune complex-mediated antigen presentation induces tumor immunity. J Clin Invest. 2002;110:71–9. doi: 10.1172/JCI15640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Clynes R, Takechi Y, Moroi Y, Houghton A, Ravetch JV. Fc receptors are required in passive and active immunity to melanoma. Proc Natl Acad Sci USA. 1998;95:652–6. doi: 10.1073/pnas.95.2.652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Bevaart L, Jansen MJ, van Vugt MJ, Verbeek JS, van de Winkel JG, Leusen JH. The high-affinity IgG receptor, FcgammaRI, plays a central role in antibody therapy of experimental melanoma. Cancer Res. 2006;66:1261–4. doi: 10.1158/0008-5472.CAN-05-2856. [DOI] [PubMed] [Google Scholar]
  • 20.Nimmerjahn F, Ravetch JV. Divergent immunoglobuling subclass activity through selective Fc receptor binding. Science. 2005;310:1510–2. doi: 10.1126/science.1118948. [DOI] [PubMed] [Google Scholar]
  • 21.Camilleri-Broet S, Cassard L, Broet P, Delmer A, Le Touneau A, Diebold J, Fridman WH, Molina TJ, Sautes-Fridman C. FcgammaRIIB is differentially expressed during B cell maturation and in B-cell lymphomas. Br J Haematol. 2004;124:55–62. doi: 10.1046/j.1365-2141.2003.04737.x. [DOI] [PubMed] [Google Scholar]
  • 22.Rankin CT, Veri MC, Gorlatov S, Tuaillon N, Burke S, Huang L, Inzunza HD, Li H, Thomas S, Johnson S, Stavenhagen J, Koenig S, Bonvini E. CD32B, the human inhibitory Fc-gamma receptor IIB, as a target for monoclonal antibody therapy of B-cell lymphoma. Blood. 2006;108:2384–91. doi: 10.1182/blood-2006-05-020602. [DOI] [PubMed] [Google Scholar]
  • 23.Anderson MS, Venanzi ES, Klein L, Chen Z, Berzins SP, Turley SJ, von Boehmer H, Bronson R, Dierich A, Benoist C, Mathis D. Projection of an immunological self shadow within the thymus by the aire protein. Science. 2002;298:1395–401. doi: 10.1126/science.1075958. [DOI] [PubMed] [Google Scholar]
  • 24.Shaheen VM, Satoh M, Richards HB, Yoshida H, Shaw M, Jennette JC, Reeves WH. Immunopathogenesis of environmentally induced lupus in mice. Environ Health Perspect. 1999;107(Suppl 5):723–7. doi: 10.1289/ehp.99107s5723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ronnblom L, Pascual V. The innate immune system in SLE: type I interferons and dendritic cells. Lupus. 2008;17:394–9. doi: 10.1177/0961203308090020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Davies AJ. Immunological tolerance and the autoimmune response. Autoimmun Rev. 2008;7:538–43. doi: 10.1016/j.autrev.2008.04.007. [DOI] [PubMed] [Google Scholar]
  • 27.Aydar Y, Sukumar S, Szakal AK, Tew JG. The influence of immune complex-bearing follicular dendritic cells on the IgM response, Ig class switching, and production of high affinity IgG. J Immunol. 2005;174:5358–66. doi: 10.4049/jimmunol.174.9.5358. [DOI] [PubMed] [Google Scholar]
  • 28.Takai T. Fc receptors and their role in immune regulation and autoimmunity. J Clin Immunol. 2005;25:1–18. doi: 10.1007/s10875-005-0353-8. [DOI] [PubMed] [Google Scholar]
  • 29.Tan Sardjono C, Mottram PL, Hogarth PM. The role of FcgammaRIIa as an inflammatory mediator in rheumatoid arthritis and systemic lupus erythematosus. Immunol Cell Biol. 2003;81:374–81. doi: 10.1046/j.1440-1711.2003.01182.x. [DOI] [PubMed] [Google Scholar]
  • 30.Tan Sardjono C, Mottram PL, van de Velde NC, Powell MS, Power D, Slocombe RF, Wicks IP, Campbell IK, McKenzie SE, Brooks M, Stevenson AW, Hogarth PM. Development of spontaneous multisystem autoimmune disease and hypersensitivity to antibody-induced inflammation in Fcgamma receptor IIa-transgenic mice. Arthritis Rheum. 2005;52:3220–9. doi: 10.1002/art.21344. [DOI] [PubMed] [Google Scholar]
  • 31.Campbell IK, Rich MJ, Bischof RJ, Hamilton JA. The colony-stimulating factors and collagen-induced arthritis: exacerbation of disease by M-CSF and G-CSF and requirement for endogenous M-CSF. J Leukoc Biol. 2000;68:144–50. [PubMed] [Google Scholar]
  • 32.Boonyarattanakalin S, Martin SE, Sun Q, Peterson BR. A synthetic mimic of human Fc receptors: defined chemical modification of cell surfaces enables efficient endocytic uptake of human immunoglobulin-G. J Am Chem Soc. 2006;128:11463–70. doi: 10.1021/ja062377w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Belostocki K, Park MS, Redecha PB, Masuda E, Salmon JE, Pricop L. FcgammaRIIa is a target for modulation by TNFalpha in human neutrophils. Clin Immunol. 2005;117:78–86. doi: 10.1016/j.clim.2005.07.001. [DOI] [PubMed] [Google Scholar]
  • 34.Abrahamson DR, Rodewald R. Evidence for the sorting of endocytic vesicle contents during the receptor-mediated transport of IgG across the newborn rat intestine. J Cell Biol. 1981;91:270–80. doi: 10.1083/jcb.91.1.270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Brambell FW. The transmission of immunity from mother to young and the catabolism of immunoglobulins. Lancet. 1966;2:1087–93. doi: 10.1016/s0140-6736(66)92190-8. [DOI] [PubMed] [Google Scholar]
  • 36.Jones EA, Waldmann TA. The mechanism of intestinal uptake and transcellular transport of IgG in the neonatal rat. J Clin Invest. 1972;51:2916–27. doi: 10.1172/JCI107116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Roberts DM, Guenthert M, Rodewald R. Isolation and characterization of the Fc receptor from the fetal yolk sac of the rat. J Cell Biol. 1990;111:1867–76. doi: 10.1083/jcb.111.5.1867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Medesan C, Radu C, Kim JK, Ghetie V, Ward ES. Localization of the site of the IgG molecule that regulates maternofetal transmission in mice. Eur J Immunol. 1996;26:2533–6. doi: 10.1002/eji.1830261038. [DOI] [PubMed] [Google Scholar]
  • 39.Ghetie V, Hubbard JG, Kim JK, Tsen MF, Lee Y, Ward ES. Abnormally short serum half-lives of IgG in beta 2-microglobulin-deficient mice. Eur J Immunol. 1996;26:690–6. doi: 10.1002/eji.1830260327. [DOI] [PubMed] [Google Scholar]
  • 40.Junghans RP, Anderson CL. The protection receptor for IgG catabolism is the beta2-microglobulin-containing neonatal intestinal transport receptor. Proc Natl Acad Sci USA. 1996;93:5512–6. doi: 10.1073/pnas.93.11.5512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Christianson GJ, Brooks W, Vekasi S, Manolfi EA, Niles J, Roopenian SL, Roths JB, Rothlein R, Roopenian DC. Beta 2-microglobulin-deficient mice are protected from hypergamma-globulinemia and have defective antibody responses because of increased IgG catabolism. J Immunol. 1997;159:4781–92. [PubMed] [Google Scholar]
  • 42.Israel EJ, Wilsker DF, Hayes KC, Schoenfeld D, Simister NE. Increased clearance of IgG in mice that lack beta 2-microglobulin: possible protective role of FcRn. Immunology. 1996;89:573–8. doi: 10.1046/j.1365-2567.1996.d01-775.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Simister NE, Mostov KE. An Fc receptor structurally related to MHC class I antigens. Nature. 1989;337:184–7. doi: 10.1038/337184a0. [DOI] [PubMed] [Google Scholar]
  • 44.Burmeister WP, Gastinel LN, Simister NE, Blum ML, Bjorkman PJ. Crystal structure at 2. 2 A resolution of the MHC-related neonatal Fc receptor. Nature. 1994;372:336–43. doi: 10.1038/372336a0. [DOI] [PubMed] [Google Scholar]
  • 45.Simister NE, Story CM. Human placental Fc receptors and the transmission of antibodies from mother to fetus. J Reprod Immunol. 1997;37:1–23. doi: 10.1016/s0165-0378(97)00068-5. [DOI] [PubMed] [Google Scholar]
  • 46.Ahouse JJ, Hagerman CL, Mittal P, Gilbert DJ, Copeland NG, Jenkins NA, Simister NE. Mouse MHC class I-like Fc receptor encoded outside the MHC. J Immunol. 1993;151:6076–88. [PubMed] [Google Scholar]
  • 47.Story CM, Mikulska JE, Simister NE. A major histocompatibility complex class I-like Fc receptor cloned from human placenta: possible role in transfer of immunoglobulin G from mother to fetus. J Exp Med. 1994;180:2377–81. doi: 10.1084/jem.180.6.2377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kacskovics I, Wu Z, Simister NE, Frenyo LV, Hammarstrom L. Cloning and characterization of the bovine MHC class I-like Fc receptor. J Immunol. 2000;164:1889–97. doi: 10.4049/jimmunol.164.4.1889. [DOI] [PubMed] [Google Scholar]
  • 49.Butler JE, Sun J, Weber P, Ford SP, Rehakova Z, Sinkora J, Francis D, Lager K. Switch recombination in fetal porcine thymus is uncoupled from somatic mutation. Vet Immunol Immunopathol. 2002;87:307–19. doi: 10.1016/s0165-2427(02)00057-0. [DOI] [PubMed] [Google Scholar]
  • 50.Mayer B, Zolnai A, Frenyo LV, Jancsik V, Szentirmay Z, Hammarstrom L, Kacskovics I. Redistribution of the sheep neonatal Fc receptor in the mammary gland around the time of parturition in ewes and its localization in the small intestine of neonatal lambs. Immunology. 2002;107:288–96. doi: 10.1046/j.1365-2567.2002.01514.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Blumberg RS, Koss T, Story CM, Barisani D, Polischuk J, Lipin A, Pablo L, Green R, Simister NE. A major histocompatibility complex class I-related Fc receptor for IgG on rat hepatocytes. J Clin Invest. 1995;95:2397–402. doi: 10.1172/JCI117934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Borvak J, Richardson J, Medesan C, Antohe F, Radu C, Simionescu M, Ghetie V, Ward ES. Functional expression of the MHC class I-related receptor, FcRn, in endothelial cells of mice. Int Immunol. 1998;10:1289–98. doi: 10.1093/intimm/10.9.1289. [DOI] [PubMed] [Google Scholar]
  • 53.Cianga P, Medesan C, Richardson JA, Ghetie V, Ward ES. Identification and function of neonatal Fc receptor in mammary gland of lactating mice. Eur J Immunol. 1999;29:2515–23. doi: 10.1002/(SICI)1521-4141(199908)29:08<2515::AID-IMMU2515>3.0.CO;2-D. [DOI] [PubMed] [Google Scholar]
  • 54.Israel EJ, Taylor S, Wu Z, Mizoguchi E, Blumberg RS, Bhan A, Simister NE. Expression of the neonatal Fc receptor, FcRn, on human intestinal epithelial cells. Immunology. 1997;92:69–74. doi: 10.1046/j.1365-2567.1997.00326.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Dickinson BL, Badizadegan K, Wu Z, Ahouse JC, Zhu X, Simister NE, Blumberg RS, Lencer WI. Bidirectional FcRn-dependent IgG transport in a polarized human intestinal epithelial cell line. J Clin Invest. 1999;104:903–11. doi: 10.1172/JCI6968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Adamski FM, King AT, Demmer J. Expression of the Fc receptor in the mammary gland during lactation in the marsupial Trichosurus vulpecula (brushtail possum) Mol Immunol. 2000;37:435–44. doi: 10.1016/s0161-5890(00)00065-1. [DOI] [PubMed] [Google Scholar]
  • 57.Leach JL, Sedmak DD, Osborne JM, Rahill B, Lairmore MD, Anderson CL. Isolation from human placenta of the IgG transporter, FcRn, and localization to the syncytiotrophoblast: implications for maternal-fetal antibody transport. J Immunol. 1996;157:3317–22. [PubMed] [Google Scholar]
  • 58.Spiekermann GM, Finn PW, Ward ES, Dumont J, Dickinson BL, Blumberg RS, Lencer WI. Receptor-mediated immunoglobulin G transport across mucosal barriers in adult life: functional expression of FcRn in the mammalian lung. J Exp Med. 2002;196:303–10. doi: 10.1084/jem.20020400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Zhu X, Meng G, Dickinson BL, Li X, Mizoguchi E, Miao L, Wang Y, Robert C, Wu B, Smith PD, Lencer WI, Blumberg RS. MHC class I-related neonatal Fc receptor for IgG is functionally expressed in monocytes, intestinal macrophages, and dendritic cells. J Immunol. 2001;166:3266–76. doi: 10.4049/jimmunol.166.5.3266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Simister NE, Story CM, Chen HL, Hunt JS. An IgG-transporting Fc receptor expressed in the syncytiotrophoblast of human placenta. Eur J Immunol. 1996;26:1527–31. doi: 10.1002/eji.1830260718. [DOI] [PubMed] [Google Scholar]
  • 61.Firan M, Bawdon R, Radu C, Ober RJ, Eaken D, Antohe F, Ghetie V, Ward ES. The MHC class I-related receptor, FcRn, plays an essential role in the maternofetal transfer of gamma-globulin in humans. Int Immunol. 2001;13:993–1002. doi: 10.1093/intimm/13.8.993. [DOI] [PubMed] [Google Scholar]
  • 62.Dumont JA, Bitonti AJ, Clark D, Evans S, Pickford M, Newman SP. Delivery of an erythropoietin-Fc fusion protein by inhalation in humans through an immunoglobulin transport pathway. J Aerosol Med. 2005;18:294–303. doi: 10.1089/jam.2005.18.294. [DOI] [PubMed] [Google Scholar]
  • 63.Bitonti AJ, Dumont JA, Low SC, Peters RT, Kropp KE, Palombella VJ, Stattel JM, Lu Y, Tan CA, Song JJ, Garcia AM, Simister NE, Spiekermann GM, Lencer WI, Blumberg RS. Pulmonary delivery of an erythropoietin Fc fusion protein in non-human primates through an immunoglobulin transport pathway. Proc Natl Acad Sci USA. 2004;101:9763–8. doi: 10.1073/pnas.0403235101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Ghetie V, Ward ES. Transcytosis and catabolism of antibody. Immunol Res. 2002;25:97–113. doi: 10.1385/IR:25:2:097. [DOI] [PubMed] [Google Scholar]
  • 65.Anderson CL, Chaudhury C, Kim J, Bronson CL, Wani MA, Mohanty S. Perspective-- FcRn transports albumin: relevance to immunology and medicine. Trends Immunol. 2006;27:343–8. doi: 10.1016/j.it.2006.05.004. [DOI] [PubMed] [Google Scholar]
  • 66.Qiao SW, Kobayashi K, Johansen FE, Sollid LM, Andersen JT, Milford E, Roopenian DC, Lencer WI, Blumberg RS. Dependence of antibody-mediated presentation of antigen on FcRn. Proc Natl Acad Sci USA. 2008;105:9337–42. doi: 10.1073/pnas.0801717105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Raghavan M, Bonagura VR, Morrison SL, Bjorkman PJ. Analysis of the pH dependence of the neonatal Fc receptor/immunoglobulin G interaction using antibody and receptor variants. Biochemistry. 1995;34:14649–57. doi: 10.1021/bi00045a005. [DOI] [PubMed] [Google Scholar]
  • 68.Liu Z, Roopenian DC, Zhou X, Christianson GJ, Diaz LA, Sedmak DD, Anderson CL. Beta2-microglobulin-deficient mice are resistant to bullous pemphigoid. J Exp Med. 1997;186:777–83. doi: 10.1084/jem.186.5.777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Roopenian DC, Christianson GJ, Sproule TJ, Brown AC, Akilesh S, Jung N, Petkova S, Avanessian L, Choi EY, Shaffer DJ, Eden PA, Anderson CL. The MHC class I-like IgG receptor controls perinatal IgG transport, IgG homeostasis, and fate of IgG-Fc-coupled drugs. J Immunol. 2003;170:3528–33. doi: 10.4049/jimmunol.170.7.3528. [DOI] [PubMed] [Google Scholar]
  • 70.Claypool SM, Dickinson BL, Wagner JS, Johansen FE, Venu N, Borawski JA, Lencer WI, Blumberg RS. Bidirectional transepithelial IgG transport by a strongly polarized basolateral membrane Fcgamma-receptor. Mol Biol Cell. 2004;15:1746–59. doi: 10.1091/mbc.E03-11-0832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Ober RJ, Martinez C, Vaccaro C, Zhou J, Ward ES. Visualizing the site and dynamics of IgG salvage by the MHC class I-related receptor, FcRn. J Immunol. 2004;172:2021–9. doi: 10.4049/jimmunol.172.4.2021. [DOI] [PubMed] [Google Scholar]
  • 72.Claypool SM, Dickinson BL, Yoshida M, Lencer WI, Blumberg RS. Functional reconstitution of human FcRn in Madin-Darby canine kidney cells requires co-expressed human beta 2-microglobulin. J Biol Chem. 2002;277:28038–50. doi: 10.1074/jbc.M202367200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.McCarthy KM, Yoong Y, Simister NE. Bidirectional transcytosis of IgG by the rat neonatal Fc receptor expressed in a rat kidney cell line: a system to study protein transport across epithelia. J Cell Sci. 2000;113:1277–85. doi: 10.1242/jcs.113.7.1277. [DOI] [PubMed] [Google Scholar]
  • 74.Ellinger I, Schwab M, Stefanescu A, Hunziker W, Fuchs R. IgG transport across trophoblast-derived BeWo cells: a model system to study IgG transport in the placenta. Eur J Immunol. 1999;29:733–44. doi: 10.1002/(SICI)1521-4141(199903)29:03<733::AID-IMMU733>3.0.CO;2-C. [DOI] [PubMed] [Google Scholar]
  • 75.Yoshida M, Claypool SM, Wagner JS, Mizoguchi E, Mizoguchi A, Roopenian DC, Lencer WI, Blumberg RS. Human neonatal Fc receptor mediates transport of IgG into luminal secretions for delivery of antigens to mucosal dendritic cells. Immunity. 2004;20:769–83. doi: 10.1016/j.immuni.2004.05.007. [DOI] [PubMed] [Google Scholar]
  • 76.Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell. 2005;122:107–18. doi: 10.1016/j.cell.2005.05.007. [DOI] [PubMed] [Google Scholar]
  • 77.Rescigno M, Urbano M, Valzasina B, Francolini M, Rotta G, Bonasio R, Granucci F, Kraehenbuhl JP, Ricciardi-Castagnoli P. Dendritic cells express tight junction proteins and penetrate gut epithelial monolayers to sample bacteria. Nat Immunol. 2001;2:361–7. doi: 10.1038/86373. [DOI] [PubMed] [Google Scholar]
  • 78.Kraus TA, Brimnes J, Muong C, Liu JH, Moran TM, Tappenden KA, Boros P, Mayer L. Induction of mucosal tolerance in Peyer’s patch-deficient, ligated small bowel loops. J Clin Invest. 2005;115:2234–43. doi: 10.1172/JCI19102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Yoshida M, Kobayashi K, Kuo TT, Bry L, Glickman JN, Claypool SM, Kaser A, Nagaishi T, Higgins DE, Mizoguchi E, Wakatsuki Y, Roopenian DC, Mizoguchi A, Lencer WI, Blumberg RS. Neonatal Fc receptor for IgG regulates mucosal immune responses to luminal bacteria. J Clin Invest. 2006;116:2142–2151. doi: 10.1172/JCI27821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Bry L, Brenner MB. Critical role of T cell-dependent serum antibody, but not the gut-associated lymphoid tissue, for surviving acute mucosal infection with Citrobacter rodentium, an attaching and effacing pathogen. J Immunol. 2004;172:433–41. doi: 10.4049/jimmunol.172.1.433. [DOI] [PubMed] [Google Scholar]
  • 81.Maaser C, Housley MP, Iimura M, Smith JR, Vallance BA, Finlay BB, Schreiber JR, Varki NM, Kagnoff MF, Eckmann L. Clearance of Citrobacter rodentium requires B cells but not secretory immunoglobulin A (IgA) or IgM antibodies. Infect Immun. 2004;72:3315–24. doi: 10.1128/IAI.72.6.3315-3324.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Brambell FW, Hemmings WA, Morris IG. A Theoretical Model of Gamma-Globulin Catabolism. Nature. 1964;203:1352–4. doi: 10.1038/2031352a0. [DOI] [PubMed] [Google Scholar]
  • 83.Humphrey JH, Fahey JL. The Metabolism of Normal Plasma Proteins and Gamma-Myeloma Protein in Mice Bearing Plasma-Cell Tumors. J Clin Invest. 1961;40:1696–705. doi: 10.1172/JCI104392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Yu Z, Lennon VA. Mechanism of intravenous immune globulin therapy in antibody-mediated autoimmune diseases. N Engl J Med. 1999;340:227–8. doi: 10.1056/NEJM199901213400311. [DOI] [PubMed] [Google Scholar]
  • 85.Samuelsson A, Towers TL, Ravetch JV. Anti-inflammatory activity of IVIG mediated through the inhibitory Fc receptor. Science. 2001;291:484–6. doi: 10.1126/science.291.5503.484. [DOI] [PubMed] [Google Scholar]
  • 86.Li N, Zhao M, Hilario-Vargas J, Prisayanh P, Warren S, Diaz LA, Roopenian DC, Liu Z. Complete FcRn dependence for intravenous Ig therapy in autoimmune skin blistering diseases. J Clin Invest. 2005;115:3440–50. doi: 10.1172/JCI24394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Akilesh S, Petkova S, Sproule TJ, Shaffer DJ, Christianson GJ, Roopenian D. The MHC class I-like Fc receptor promotes humorally mediated autoimmune disease. J Clin Invest. 2004;113:1328–33. doi: 10.1172/JCI18838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Raghavan M, Chen MY, Gastinel LN, Bjorkman PJ. Investigation of the interaction between the class I MHC-related Fc receptor and its immunoglobulin G ligand. Immunity. 1994;1:303–15. doi: 10.1016/1074-7613(94)90082-5. [DOI] [PubMed] [Google Scholar]
  • 89.Getman KE, Balthasar JP. Pharmacokinetic effects of 4C9, an anti-FcRn antibody, in rats: implications for the use of FcRn inhibitors for the treatment of humoral autoimmune and alloimmune conditions. J Pharm Sci. 2005;94:718–29. doi: 10.1002/jps.20297. [DOI] [PubMed] [Google Scholar]
  • 90.Liu L, Garcia AM, Santoro H, Zhang Y, McDonnell K, Dumont J, Bitonti A. Amelioration of experimental autoimmune myasthenia gravis in rats by neonatal FcR blockade. J Immunol. 2007;178:5390–8. doi: 10.4049/jimmunol.178.8.5390. [DOI] [PubMed] [Google Scholar]
  • 91.Vaccaro C, Zhou J, Ober RJ, Ward ES. Engineering the Fc region of immunoglobulin G to modulate in vivo antibody levels. Nat Biotechnol. 2005;23:1283–8. doi: 10.1038/nbt1143. [DOI] [PubMed] [Google Scholar]
  • 92.Blumberg RS, Lencer WI. Antibodies in the breakdown lane. Nat Biotechnol. 2005;23:1232–4. doi: 10.1038/nbt1005-1232. [DOI] [PubMed] [Google Scholar]
  • 93.Mezo AR, McDonnell KA, Hehir CA, Low SC, Palombella VJ, Stattel JM, Kamphaus GD, Fraley C, Zhang Y, Dumont JA, Bitonti AJ. Reduction of IgG in nonhuman primates by a peptide antagonist of the neonatal Fc receptor FcRn. Proc Natl Acad Sci USA. 2008;105:2337–42. doi: 10.1073/pnas.0708960105. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES