201
|
Kuo TT, Baker K, Yoshida M, Qiao SW, Aveson VG, Lencer WI, Blumberg RS. Neonatal Fc receptor: from immunity to therapeutics. J Clin Immunol 2010; 30:777-89. [PMID: 20886282 PMCID: PMC2970823 DOI: 10.1007/s10875-010-9468-4] [Citation(s) in RCA: 175] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 09/14/2010] [Indexed: 11/30/2022]
Abstract
The neonatal Fc receptor (FcRn), also known as the Brambell receptor and encoded by Fcgrt, is a MHC class I like molecule that functions to protect IgG and albumin from catabolism, mediates transport of IgG across epithelial cells, and is involved in antigen presentation by professional antigen presenting cells. Its function is evident in early life in the transport of IgG from mother to fetus and neonate for passive immunity and later in the development of adaptive immunity and other functions throughout life. The unique ability of this receptor to prolong the half-life of IgG and albumin has guided engineering of novel therapeutics. Here, we aim to summarize the basic understanding of FcRn biology, its functions in various organs, and the therapeutic design of antibody- and albumin-based therapeutics in light of their interactions with FcRn.
Collapse
Affiliation(s)
- Timothy T Kuo
- Division of Gastroenterology, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA 02115, USA.
| | | | | | | | | | | | | |
Collapse
|
202
|
Meslier Y, André S, Teyssandier M, Kaveri SV, Lacroix-Desmazes S. Maternally transferred anti-factor VIII IgG reduce the anti-factor VIII humoral immune response in factor VIII-deficient mice. Immunology 2010; 131:549-55. [PMID: 20673239 DOI: 10.1111/j.1365-2567.2010.03327.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Replacement therapy with exogenous factor VIII (FVIII) to treat haemorrhages or used in prophylaxis induces inhibitory anti-FVIII immunoglobulin G (IgG) in some patients with haemophilia A. Therapeutic strategies to prevent the onset of the deleterious anti-FVIII immune response are still lacking. Maternal IgG is transferred to the offspring during fetal and neonatal life. While protecting the offspring from bacterial and viral infections, maternal IgG may alter the repertoires of T and B lymphocytes, and may impair vaccination in early infancy. Using haemophilic mice, we demonstrate that the transfer of maternal anti-FVIII IgG modulates the onset of anti-FVIII inhibitory IgG in early adulthood. The protective effect is reproduced upon reconstitution of naive mice with anti-FVIII IgG, suggesting that the reduced ability to mount an anti-FVIII immune response is the result of an interference between circulating anti-FVIII IgG and the administered FVIII rather than to a profound remodelling of lymphocyte repertoires occurring during the ontogeny of the immune system.
Collapse
Affiliation(s)
- Yann Meslier
- INSERM U872, Centre de recherche des Cordeliers, Paris, France.
| | | | | | | | | |
Collapse
|
203
|
Sesarman A, Vidarsson G, Sitaru C. The neonatal Fc receptor as therapeutic target in IgG-mediated autoimmune diseases. Cell Mol Life Sci 2010; 67:2533-50. [PMID: 20217455 PMCID: PMC11115620 DOI: 10.1007/s00018-010-0318-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2009] [Revised: 01/29/2010] [Accepted: 02/12/2010] [Indexed: 01/01/2023]
Abstract
Therapy approaches based on lowering levels of pathogenic autoantibodies represent rational, effective, and safe treatment modalities of autoimmune diseases. The neonatal Fc receptor (FcRn) is a major factor regulating the serum levels of IgG antibodies. While FcRn-mediated half-life extension is beneficial for IgG antibody responses against pathogens, it also prolongs the serum half-life of IgG autoantibodies and thus promotes tissue damage in autoimmune diseases. In the present review article, we examine current evidence on the relevance of FcRn in maintaining high autoantibody levels and discuss FcRn-targeted therapeutic approaches. Further investigation of the FcRn-IgG interaction will not only provide mechanistic insights into the receptor function, but should also greatly facilitate the design of therapeutics combining optimal pharmacokinetic properties with the appropriate antibody effector functions in autoimmune diseases.
Collapse
Affiliation(s)
- Alina Sesarman
- Department of Dermatology, University of Freiburg, Hauptstrasse 7, 79104 Freiburg, Germany
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Cassian Sitaru
- Department of Dermatology, University of Freiburg, Hauptstrasse 7, 79104 Freiburg, Germany
- Centre for Biological Signalling Studies (bioss), University of Freiburg, Freiburg, Germany
| |
Collapse
|
204
|
Masuda A, Yoshida M, Shiomi H, Morita Y, Kutsumi H, Inokuchi H, Mizuno S, Nakamura A, Takai T, Blumberg RS, Azuma T. Role of Fc Receptors as a therapeutic target. ACTA ACUST UNITED AC 2009; 8:80-6. [PMID: 19275696 DOI: 10.2174/187152809787582525] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
It has been forty years since the discovery of Fc Receptors and their function. Fc Receptors include the IgG receptors (FcgammaR), high-affinity IgE receptor (FcepsilonRI), IgA and IgA/IgM receptors, and neonatal Fc receptor for IgG (FcRn). In particular, the FcgammaRs 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 FcgammaRs 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.
Collapse
Affiliation(s)
- Atsuhiro Masuda
- Department of Gastroenterology, Kobe University School of Medicine, Kobe, Hyogo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
205
|
Farkas T, Dufour J, Jiang X, Sestak K. Detection of norovirus-, sapovirus- and rhesus enteric calicivirus-specific antibodies in captive juvenile macaques. J Gen Virol 2009; 91:734-8. [PMID: 19889933 DOI: 10.1099/vir.0.015263-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The objective of this study was to determine the prevalence of anti-norovirus (NoV), -sapovirus (SaV) and -Tulane virus (TV) antibodies in rhesus macaques of the Tulane National Primate Research Center and to evaluate the antigenic relationship between these viruses. A high prevalence of NoV-binding (51-61 %) and SaV-binding (50-56 %) antibodies and TV-neutralizing (69 %) antibodies were detected. Serum samples obtained during a human NoV outbreak and a multivalent anti-NoV hyperimmune serum were not able to neutralize TV infectivity. Conversely, low levels of cross-reactivity between the prototype TV and NoVs, but not between the TV and SaVs were detected by ELISA. These data indicate the preservation of some cross-reactive B-cell epitopes between the rhesus and human caliciviruses (CVs). The high prevalence of human and rhesus CV-specific serum antibodies suggests the frequent exposure of colony macaques to enteric CVs including the possibility of CV transmission between human and non-human primate hosts.
Collapse
Affiliation(s)
- Tibor Farkas
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | | | | | | |
Collapse
|
206
|
Kobayashi K, Qiao SW, Yoshida M, Baker K, Lencer WI, Blumberg RS. An FcRn-dependent role for anti-flagellin immunoglobulin G in pathogenesis of colitis in mice. Gastroenterology 2009; 137:1746-56.e1. [PMID: 19664634 PMCID: PMC2787451 DOI: 10.1053/j.gastro.2009.07.059] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 06/25/2009] [Accepted: 07/22/2009] [Indexed: 12/31/2022]
Abstract
BACKGROUND & AIMS The neonatal Fc receptor for immunoglobulin (Ig)G (FcRn) protects monomeric IgG from catabolism in parenchymal and hematopoietic cells during adult life. In dendritic cells, FcRn also promotes presentation of antigens in association with IgG. Because IgGs with anti-bacterial specificity are a hallmark of inflammatory bowel disease, we sought to determine their significance and relationship to FcRn expression in antigen-presenting cells, focusing on IgGs specific for flagellin. METHODS Levels of circulating anti-flagellin IgG were induced in wild-type and FcRn(-/-) mice, followed by induction of colitis with dextran sodium sulfate (DSS). Bone marrow chimera models were used to localize the site of FcRn action. RESULTS Wild-type mice that received anti-flagellin IgG exhibited more severe colitis following administration of DSS, compared with mice that received control IgG. Wild-type mice immunized with flagellin exhibited significantly more severe colitis in response to DSS administration than that observed in similarly treated FcRn(-/-) mice. In chimera studies, FcRn(-/-) mice given wild-type bone marrow and immunized with flagellin exhibited significantly more colitis than wild-type mice given FcRn(-/-) bone marrow and immunized with flagellin. Serum anti-flagellin IgG levels were similar in both sets of chimeric mice, consistent with the equal participation of hematopoietic and nonhematopoeitic cells in FcRn-mediated IgG protection. CONCLUSIONS Anti-bacterial IgG antibodies are involved in the pathogenesis of colitis; this pathway requires FcRn in antigen presenting cells, the major subset of hematopoietic cells that express FcRn.
Collapse
Affiliation(s)
- Kanna Kobayashi
- Gastroenterology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | |
Collapse
|
207
|
Effects of endocytosis inhibitors on internalization of human IgG by Caco-2 human intestinal epithelial cells. Life Sci 2009; 85:800-7. [PMID: 19879882 DOI: 10.1016/j.lfs.2009.10.012] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Revised: 09/17/2009] [Accepted: 10/20/2009] [Indexed: 11/23/2022]
Abstract
AIMS The purpose of this study was to characterize the internalization mechanism of human IgG into the epithelial cells of human small intestine, employing human intestinal epithelial cell line Caco-2 as an in vitro model system. MAIN METHODS Real-time PCR analysis and uptake studies of fluorescein isothiocyanate-labeled IgG (FITC-IgG) from human serum were performed using Caco-2 cells. KEY FINDINGS Real-time PCR analysis showed that mRNA level of the neonatal Fc receptor (FcRn) was increased during the differentiation process in Caco-2 cells. The binding of FITC-labeled human IgG to the membrane surface of Caco-2 cells increased with a decrease in pH of incubation buffer. The uptake of FITC-IgG was also stimulated at acidic pH and was time-dependent. The binding and uptake of FITC-IgG at pH 6.0 was partially, but significantly, decreased by human gamma-globulin in a concentration-dependent manner. A mixture of metabolic inhibitors (sodium azide and 2-deoxyglucose) significantly inhibited the uptake, but not the binding, of FITC-IgG. In addition, endosomal acidification inhibitors such as bafilomycin A(1) and chloroquine significantly increased the accumulation of FITC-IgG. Clathrin-dependent endocytosis inhibitors (phenylarsine oxide and chlorpromazine) and caveolin-dependent endocytosis inhibitors (nystatin and indomethacin) did not decrease the uptake of FITC-IgG at pH 6.0. In contrast, macropinocytosis inhibitors such as cytochalasin B and 5-(N-ethyl-N-isopropyl) amiloride significantly decreased the uptake of FITC-IgG at pH 6.0. SIGNIFICANCE The internalization of human IgG in human intestine might be, at least in part, due to FcRn-mediated endocytosis, which could occur by a process other than clathrin- and caveolin-dependent mechanisms.
Collapse
|
208
|
Toxin-mediated effects on the innate mucosal defenses: implications for enteric vaccines. Infect Immun 2009; 77:5206-15. [PMID: 19737904 DOI: 10.1128/iai.00712-09] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Recent studies have confirmed older observations that the enterotoxins enhance enteric bacterial colonization and pathogenicity. How and why this happens remains unknown at this time. It appears that toxins such as the heat-labile enterotoxin (LT) from Escherichia coli can help overcome the innate mucosal barrier as a key step in enteric pathogen survival. We review key observations relevant to the roles of LT and cholera toxin in protective immunity and the effects of these toxins on innate mucosal defenses. We suggest either that toxin-mediated fluid secretion mechanically disrupts the mucus layer or that toxins interfere with innate mucosal defenses by other means. Such a breach gives pathogens access to the enterocyte, leading to binding and pathogenicity by enterotoxigenic E. coli (ETEC) and other organisms. Given the common exposure to LT(+) ETEC by humans visiting or residing in regions of endemicity, barrier disruption should frequently render the gut vulnerable to ETEC and other enteric infections. Conversely, toxin immunity would be expected to block this process by protecting the innate mucosal barrier. Years ago, Peltola et al. (Lancet 338:1285-1289, 1991) observed unexpectedly broad protective effects against LT(+) ETEC and mixed infections when using a toxin-based enteric vaccine. If toxins truly exert barrier-disruptive effects as a key step in pathogenesis, then a return to classic toxin-based vaccine strategies for enteric disease is warranted and can be expected to have unexpectedly broad protective effects.
Collapse
|
209
|
Palmeira P, Costa-Carvalho BT, Arslanian C, Pontes GN, Nagao AT, Carneiro-Sampaio MMS. Transfer of antibodies across the placenta and in breast milk from mothers on intravenous immunoglobulin. Pediatr Allergy Immunol 2009; 20:528-35. [PMID: 19220771 DOI: 10.1111/j.1399-3038.2008.00828.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We studied the levels of immunoglobulins in colostrum, milk and sera from two common variable immunodeficiency (CVID) mothers (M1 and M2), and in sera from their newborn infants. During pregnancy they continued intravenous immunoglobulin therapy (IVIG). Antibody levels from maternal and cord blood collected at delivery and colostrum and milk, collected on the 3rd and 7th post-partum days, respectively, were analyzed. Although cord/maternal blood ratios of total immunoglobulins and subclasses, as well as specific antibodies differed between M1 and M2, both showed good placental transfer of anti-protein and anti-polysaccharide antibodies, despite lower cord/maternal blood ratios in M2. Anti-Streptococcus pneumoniae antibody avidity indexes were similar between paired maternal and cord serum. Both mothers' colostrum and milk samples showed only traces of IgA, and IgM and IgG levels in colostrum were within normal range in M1, whereas M2 presented elevated IgG and low IgM levels, when compared with healthy mothers. The study of colostrum and milk activity showed that they strongly inhibited enteropathogenic Escherichia coli adhesion in vitro. CVID patients must be informed about the relevance of regular IVIG administration during pregnancy, not only for their own health but also for their immune immature offspring. Breast-feeding should be encouraged as colostra from these CVID patients strongly inhibited E. coli adhesion to human epithelial cells thus providing immunological protection plus nutritional and psychological benefits for the infant.
Collapse
Affiliation(s)
- Patricia Palmeira
- Department of Pediatrics, Faculdade de Medicina, Universidade Federal de São Paulo, CEP: 05403-000, São Paulo, SP, Brazil.
| | | | | | | | | | | |
Collapse
|
210
|
Garg A, Balthasar JP. Investigation of the influence of FcRn on the distribution of IgG to the brain. AAPS JOURNAL 2009; 11:553-7. [PMID: 19636712 DOI: 10.1208/s12248-009-9129-9] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2008] [Accepted: 06/23/2009] [Indexed: 12/11/2022]
Abstract
It has been suggested that the neonatal Fc receptor (FcRn) is a primary determinant of the distribution of IgG to the brain. In the present report, (125)I-labeled 7E3, a monoclonal IgG1 antibody, was injected intravenously to groups of FcRn-deficient mice and C57BL/6J control mice. Sub-groups of three mice were sacrificed at several time points. Blood and brain tissue were harvested and radioactivity was assessed. Antibody concentrations in brain were corrected for residual blood using (51)Cr-labeled red blood cells. Data were analyzed via WinNonlin, and areas under plasma and tissue concentration vs. time curves (AUCs) were assessed via the Bailer method. The apparent plasma elimination half-life and clearance of 7E3 were 13.61 +/- 0.61 days and 6.5 +/- 0.10 ml/day/kg in control mice and 0.70 +/- 0.05 days and 63.5 +/- 2.7 ml/day/kg in the knockout mice. Plasma and brain AUCs (0-10 days) were found to be 3,338.7 +/- 50.4 and 7.46 +/- 0.5 nM day in control animals and 781.2 +/- 16.6 and 1.65 +/- 0.1 nM day in FcRn-deficient animals. There was no significant difference between brain-to-plasma AUC ratios in control and FcRn-deficient mice (0.0022 +/- 0.00015 vs. 0.0021 +/- 0.00011, p = 0.3347). Two-way analysis of variance showed no significant differences, at any time point, between brain-to-plasma concentration ratios determined from control and knockout animals. The results suggest that FcRn does not contribute significantly to the "blood-brain barrier" for IgG in mice, and the data suggest that FcRn is not responsible for the low exposure of IgG in the brain relative to plasma.
Collapse
Affiliation(s)
- Amit Garg
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | | |
Collapse
|
211
|
Baker K, Qiao SW, Kuo T, Kobayashi K, Yoshida M, Lencer WI, Blumberg RS. Immune and non-immune functions of the (not so) neonatal Fc receptor, FcRn. Semin Immunopathol 2009; 31:223-36. [PMID: 19495758 PMCID: PMC3898171 DOI: 10.1007/s00281-009-0160-9] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2009] [Accepted: 05/14/2009] [Indexed: 02/06/2023]
Abstract
Careful regulation of the body's immunoglobulin-G (IgG) and albumin concentrations is necessitated by the importance of their respective functions. As such, the neonatal Fc receptor (FcRn) which, as a single receptor, is capable of regulating both of these molecules, has become an important focus of investigation. In addition to these essential protection functions, FcRn possesses a host of other functions that are equally as critical. During the very first stages of life, FcRn mediates the passive transfer of IgG from mother to offspring both before and after birth. In the adult, FcRn regulates the persistence of both IgG and albumin in the serum as well as the movement of IgG, and any bound cargo, between different compartments of the body. This shuttling allows for the movement not only of monomeric ligand but also of antigen/antibody complexes from one cell type to another in such a way as to facilitate the efficient initiation of immune responses towards opsonized pathogens. As such, FcRn continues to play the role of an immunological sensor throughout adult life, particularly in regions such as the gut which are exposed to a large number of infectious antigens. Increasing appreciation for the contributions of FcRn to both homeostatic and pathological states is generating an intense interest in the potential for therapeutic modulation of FcRn binding. A greater understanding of FcRn's pleiotropic roles is thus imperative for a variety of therapeutic purposes.
Collapse
Affiliation(s)
- Kristi Baker
- Division of Gastroenterology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA, Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Shuo-Wang Qiao
- Rikshospitalet University Hospital, 0027 Oslo, Norway, University of Oslo, 0027 Oslo, Norway
| | - Timothy Kuo
- Division of Gastroenterology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA, Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Kanna Kobayashi
- Division of Gastroenterology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA, Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Masaru Yoshida
- Department of Gastroenterology & The Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medecine, Hyogo, Japan
| | - Wayne I. Lencer
- Harvard Digestive Diseases Center, Boston, MA 02115, USA, GI Cell Biology, Division of Pediatric Gastroenterology and Nutrition, Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Richard S. Blumberg
- Division of Gastroenterology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA, Department of Medicine, Harvard Medical School, Boston, MA 02115, USA, Harvard Digestive Diseases Center, Boston, MA 02115, USA
| |
Collapse
|
212
|
Yeung YA, Leabman MK, Marvin JS, Qiu J, Adams CW, Lien S, Starovasnik MA, Lowman HB. Engineering Human IgG1 Affinity to Human Neonatal Fc Receptor: Impact of Affinity Improvement on Pharmacokinetics in Primates. THE JOURNAL OF IMMUNOLOGY 2009; 182:7663-71. [DOI: 10.4049/jimmunol.0804182] [Citation(s) in RCA: 192] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
213
|
Tzaban S, Massol RH, Yen E, Hamman W, Frank SR, Lapierre LA, Hansen SH, Goldenring JR, Blumberg RS, Lencer WI. The recycling and transcytotic pathways for IgG transport by FcRn are distinct and display an inherent polarity. J Cell Biol 2009; 185:673-84. [PMID: 19451275 PMCID: PMC2711563 DOI: 10.1083/jcb.200809122] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2008] [Accepted: 04/15/2009] [Indexed: 11/30/2022] Open
Abstract
The Fc receptor FcRn traffics immunoglobulin G (IgG) in both directions across polarized epithelial cells that line mucosal surfaces, contributing to host defense. We show that FcRn traffics IgG from either apical or basolateral membranes into the recycling endosome (RE), after which the actin motor myosin Vb and the GTPase Rab25 regulate a sorting step that specifies transcytosis without affecting recycling. Another regulatory component of the RE, Rab11a, is dispensable for transcytosis, but regulates recycling to the basolateral membrane only. None of these proteins affect FcRn trafficking away from lysosomes. Thus, FcRn transcytotic and recycling sorting steps are distinct. These results are consistent with a single structurally and functionally heterogeneous RE compartment that traffics FcRn to both cell surfaces while discriminating between recycling and transcytosis pathways polarized in their direction of transport.
Collapse
Affiliation(s)
- Salit Tzaban
- Children's Hospital, Gastroenterology Division; Brigham and Women's Hospital, Gastroenterology Division; and the Harvard Digestive Diseases Center, Harvard Medical School, Boston, MA 02115
| | - Ramiro H. Massol
- Children's Hospital, Gastroenterology Division; Brigham and Women's Hospital, Gastroenterology Division; and the Harvard Digestive Diseases Center, Harvard Medical School, Boston, MA 02115
| | - Elizabeth Yen
- Children's Hospital, Gastroenterology Division; Brigham and Women's Hospital, Gastroenterology Division; and the Harvard Digestive Diseases Center, Harvard Medical School, Boston, MA 02115
| | - Wendy Hamman
- Children's Hospital, Gastroenterology Division; Brigham and Women's Hospital, Gastroenterology Division; and the Harvard Digestive Diseases Center, Harvard Medical School, Boston, MA 02115
| | - Scott R. Frank
- Children's Hospital, Gastroenterology Division; Brigham and Women's Hospital, Gastroenterology Division; and the Harvard Digestive Diseases Center, Harvard Medical School, Boston, MA 02115
| | - Lynne A. Lapierre
- Deptartment of Surgery, Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37232
- Nashville Veterans Affairs Medical Center, Nashville, TN 37232
| | - Steen H. Hansen
- Children's Hospital, Gastroenterology Division; Brigham and Women's Hospital, Gastroenterology Division; and the Harvard Digestive Diseases Center, Harvard Medical School, Boston, MA 02115
| | - James R. Goldenring
- Deptartment of Surgery, Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37232
- Nashville Veterans Affairs Medical Center, Nashville, TN 37232
| | - Richard S. Blumberg
- Children's Hospital, Gastroenterology Division; Brigham and Women's Hospital, Gastroenterology Division; and the Harvard Digestive Diseases Center, Harvard Medical School, Boston, MA 02115
| | - Wayne I. Lencer
- Children's Hospital, Gastroenterology Division; Brigham and Women's Hospital, Gastroenterology Division; and the Harvard Digestive Diseases Center, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
214
|
Gan Z, Ram S, Vaccaro C, Ober RJ, Ward ES. Analyses of the recycling receptor, FcRn, in live cells reveal novel pathways for lysosomal delivery. Traffic 2009; 10:600-14. [PMID: 19192244 PMCID: PMC2813311 DOI: 10.1111/j.1600-0854.2009.00887.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Lysosomes play a central role in the degradation of proteins and other macromolecules. The mechanisms by which receptors are transferred to lysosomes for constitutive degradation are poorly understood. We have analyzed the processes that lead to the lysosomal delivery of the Fc receptor, FcRn. These studies provide support for a novel pathway for receptor delivery. Specifically, unlike other receptors that enter intraluminal vesicles in late endosomes, FcRn is transferred from the limiting membrane of such endosomes to lysosomes, and is rapidly internalized into the lysosomal lumen. By contrast, LAMP-1 persists on the limiting membrane. Receptor transfer is mediated by tubular extensions from late endosomes to lysosomes, or by interactions of the two participating organelles in kiss-and-linger-like processes, whereas full fusion is rarely observed. The persistence of FcRn on the late endosomal limiting membrane, together with selective transfer to lysosomes, allows this receptor to undergo recycling or degradation. Consequently, late endosomes have functional plasticity, consistent with the presence of the Rab5 GTPase in discrete domains on these compartments.
Collapse
Affiliation(s)
- Zhuo Gan
- Biomedical Engineering Graduate Program, University of Texas Southwestern Medical Center at Dallas, Dallas TX 75390, USA
- Department of Immunology, University of Texas Southwestern Medical Center at Dallas, Dallas TX 75390, USA
| | - Sripad Ram
- Department of Immunology, University of Texas Southwestern Medical Center at Dallas, Dallas TX 75390, USA
| | - Carlos Vaccaro
- Department of Immunology, University of Texas Southwestern Medical Center at Dallas, Dallas TX 75390, USA
| | - Raimund J. Ober
- Department of Immunology, University of Texas Southwestern Medical Center at Dallas, Dallas TX 75390, USA
- Department of Electrical Engineering, University of Texas at Dallas, Richardson, TX 75080, USA
| | - E. Sally Ward
- Department of Immunology, University of Texas Southwestern Medical Center at Dallas, Dallas TX 75390, USA
| |
Collapse
|
215
|
Montoyo HP, Vaccaro C, Hafner M, Ober RJ, Mueller W, Ward ES. Conditional deletion of the MHC class I-related receptor FcRn reveals the sites of IgG homeostasis in mice. Proc Natl Acad Sci U S A 2009; 106:2788-93. [PMID: 19188594 PMCID: PMC2650344 DOI: 10.1073/pnas.0810796106] [Citation(s) in RCA: 168] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Indexed: 02/06/2023] Open
Abstract
The MHC class I-related receptor FcRn regulates the levels and persistence of IgG in vivo. This receptor salvages IgG from lysosomal degradation within cells, and the binding properties of an IgG for FcRn correlate with in vivo half-life. FcRn is expressed at multiple different sites throughout adult life. However, the cell types and sites at which FcRn maintains IgG homeostasis are not well defined. Toward understanding the sites of FcRn function, we have generated a mouse strain in which this Fc receptor can be conditionally deleted. In combination with mice that express Cre recombinase under the control of the Tie2 promoter (Tie2-Cre), the effect of site-specific deletion of floxed FcRn in endothelial and hematopoietic cells on IgG persistence was analyzed. The pharmacokinetics and steady-state levels of IgG in Tie2-Cre mice that are homozygous for the floxed FcRn allele reveal a complete loss of FcRn function in regulating the half-lives of wild-type IgG. The primary sites for the maintenance of endogenous IgGs in mice are therefore endothelial and hematopoietic cells.
Collapse
Affiliation(s)
- Héctor Pérez Montoyo
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390-9093
| | - Carlos Vaccaro
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390-9093
| | - Martin Hafner
- Helmholtz Centre for Infection Research, Braunschweig 38124, Germany
| | - Raimund J. Ober
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390-9093
- Department of Electrical Engineering, University of Texas at Dallas, Richardson, TX 75080; and
| | - Werner Mueller
- Helmholtz Centre for Infection Research, Braunschweig 38124, Germany
- Faculty of Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom
| | - E. Sally Ward
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390-9093
| |
Collapse
|
216
|
Ward ES, Ober RJ. Chapter 4: Multitasking by exploitation of intracellular transport functions the many faces of FcRn. Adv Immunol 2009; 103:77-115. [PMID: 19755184 DOI: 10.1016/s0065-2776(09)03004-1] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The MHC Class I-related receptor, FcRn, transports antibodies of the immunoglobulin G (IgG) class within and across a diverse array of different cell types. Through this transport, FcRn serves multiple roles throughout adult life that extend well beyond its earlier defined function of transcytosing IgGs from mother to offspring. These roles include the maintenance of IgG levels and the delivery of antigen in the form of immune complexes to degradative compartments within cells. Recent studies have led to significant advances in knowledge of the intracellular trafficking of FcRn and (engineered) IgGs at both the molecular and cellular levels. The engineering of FcRn-IgG (or Fc) interactions to generate antibodies of increased longevity represents an area of active interest, particularly in the light of the expanding use of antibodies in therapy. The strict pH dependence of FcRn-IgG interactions, with binding at pH 6 that becomes essentially undetectable as near neutral pH is approached, is essential for efficient transport. The requirement for retention of low affinity at near neutral pH increases the complexity of engineering antibodies for increased half-life. Conversely, engineered IgGs that have gained significant binding for FcRn at this pH can be potent inhibitors of FcRn that lower endogenous IgG levels and have multiple potential uses as therapeutics. In addition, molecular studies of FcRn-IgG interactions indicate that mice have limitations as preclinical models for FcRn function, primarily due to cross-species differences in FcRn-binding specificity.
Collapse
Affiliation(s)
- E Sally Ward
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | |
Collapse
|
217
|
Andersen JT, Sandlie I. The Versatile MHC Class I-related FcRn Protects IgG and Albumin from Degradation: Implications for Development of New Diagnostics and Therapeutics. Drug Metab Pharmacokinet 2009; 24:318-32. [DOI: 10.2133/dmpk.24.318] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
218
|
Tillinger W, Jilch R, Jilma B, Brunner H, Koeller U, Lichtenberger C, Waldhör T, Reinisch W. Expression of the high-affinity IgG receptor FcRI (CD64) in patients with inflammatory bowel disease: a new biomarker for gastroenterologic diagnostics. Am J Gastroenterol 2009; 104:102-9. [PMID: 19098857 DOI: 10.1038/ajg.2008.6] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES We sought to determine the quantitative expression of the high-affinity Fc receptor (CD64) on polymorphonuclear neutrophils (PMNs) in inflammatory and functional conditions of the intestine and investigated its correlation with clinical and biological parameters of inflammation. METHODS The quantitative expression of CD64 was determined by fluorescence-activated cell sorting analysis in patients with active or inactive inflammatory bowel disease (IBD, n=76), infectious enterocolitis, lactose and/or fructose intolerance, and healthy subjects. RESULTS The quantitative expression of CD64 in patients with active IBD (3,398+/-3,589 molecules per PMN, n=27) was significantly higher than in healthy subjects (607+/-265, n=28, P<0.001) or in patients with lactose/fructose intolerance (531+/-150, n=32, P<0.001). The expression of CD64 correlated significantly with C-reactive protein (CRP, 0.65, P<0.0001), Crohn's disease activity index (CDAI, 0.53, P<0.0001), and colitis activity index (CAI, 0.63, P<0.0001) in patients with IBD. With a cutoff point of 800, CD64 had a sensitivity of 88% and a specificity of 93% in discriminating between lactose/fructose intolerance and active IBD. The quantitative expression of CD64 in patients with infectious enterocolitis (19,190+/-8,920, n=22) was significantly higher than in patients with active IBD (P<0.001). With a cutoff point of 10,000, CD64 showed a sensitivity of 96% and a specificity of 97% in discriminating between infectious enterocolitis and active IBD. CONCLUSIONS CD64 could serve as a valuable tool to discriminate between IBD, infectious enterocolitis, and functional intestinal disorders.
Collapse
Affiliation(s)
- Wolfgang Tillinger
- 1st Medical Department, Hietzing Hospital, Wolkersbergenstrasse 1, Vienna, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
219
|
Mi W, Wanjie S, Lo ST, Gan Z, Pickl-Herk B, Ober RJ, Ward ES. Targeting the neonatal fc receptor for antigen delivery using engineered fc fragments. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:7550-61. [PMID: 19017944 PMCID: PMC2738423 DOI: 10.4049/jimmunol.181.11.7550] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The development of approaches for Ag delivery to the appropriate subcellular compartments of APCs and the optimization of Ag persistence are both of central relevance for the induction of protective immunity or tolerance. The expression of the neonatal Fc receptor, FcRn, in APCs and its localization to the endosomal system suggest that it might serve as a target for Ag delivery using engineered Fc fragment-epitope fusions. The impact of FcRn binding characteristics of an Fc fragment on in vivo persistence allows this property to also be modulated. We have therefore generated recombinant Fc (mouse IgG1-derived) fusions containing the N-terminal epitope of myelin basic protein that is associated with experimental autoimmune encephalomyelitis in H-2(u) mice. The Fc fragments have distinct binding properties for FcRn that result in differences in intracellular trafficking and in vivo half-lives, allowing the impact of these characteristics on CD4(+) T cell responses to be evaluated. To dissect the relative roles of FcRn and the "classical" FcgammaRs in Ag delivery, analogous aglycosylated Fc-MBP fusions have been generated. We show that engineered Fc fragments with increased affinities for FcRn at pH 6.0-7.4 are more effective in delivering Ag to FcRn-expressing APCs in vitro relative to their lower affinity counterparts. However, higher affinity of the FcRn-Fc interaction at near neutral pH results in decreased in vivo persistence. The trade-off between improved FcRn targeting efficiency and lower half-life becomes apparent during analyses of T cell proliferative responses in mice, particularly when Fc-MBP fusions with both FcRn and FcgammaR binding activity are used.
Collapse
Affiliation(s)
- Wentao Mi
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9093
| | - Sylvia Wanjie
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9093
| | - Su-Tang Lo
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9093
| | - Zhuo Gan
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9093
| | - Beatrix Pickl-Herk
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9093
| | - Raimund J. Ober
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9093
- Department of Electrical Engineering, University of Texas at Dallas, Richardson, TX 75080
| | - E. Sally Ward
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9093
| |
Collapse
|
220
|
Tayer-Shifman OE, Ilan Y. Immune modulation by antibodies and antibody receptors. Expert Opin Ther Pat 2008. [DOI: 10.1517/13543770802547196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
221
|
Cervenak J, Kacskovics I. The neonatal Fc receptor plays a crucial role in the metabolism of IgG in livestock animals. Vet Immunol Immunopathol 2008; 128:171-7. [PMID: 19027179 DOI: 10.1016/j.vetimm.2008.10.300] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The role of the FcRn is fundamental in IgG metabolism. It is involved in transporting maternal immunity and protects IgG from fast degradation throughout life. While the acquisition of the humoral immunity through the transfer of IgG from mother to offspring shows species-specific differences, the mechanism how FcRn protects IgG from degradation is highly similar in all species analyzed so far. This review summarizes the current understanding of the FcRn-mediated IgG metabolism in livestock animals (cattle, sheep and pig) and point out those aspects that remain to be exposed for better understanding the function of this system in these species and also to take advantages of it for economical purposes.
Collapse
Affiliation(s)
- Judit Cervenak
- Department of Immunology, Institute of Biology, Faculty of Science, Eötvös Loránd University, Pázmány Péter s. 1/C, Budapest H-1117, Hungary
| | | |
Collapse
|
222
|
Goebl NA, Babbey CM, Datta-Mannan A, Witcher DR, Wroblewski VJ, Dunn KW. Neonatal Fc receptor mediates internalization of Fc in transfected human endothelial cells. Mol Biol Cell 2008; 19:5490-505. [PMID: 18843053 DOI: 10.1091/mbc.e07-02-0101] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The neonatal Fc receptor, FcRn mediates an endocytic salvage pathway that prevents degradation of IgG, thus contributing to the homeostasis of circulating IgG. Based on the low affinity of IgG for FcRn at neutral pH, internalization of IgG by endothelial cells is generally believed to occur via fluid-phase endocytosis. To investigate the role of FcRn in IgG internalization, we used quantitative confocal microscopy to characterize internalization of fluorescent Fc molecules by HULEC-5A lung microvascular endothelia transfected with GFP fusion proteins of human or mouse FcRn. In these studies, cells transfected with FcRn accumulated significantly more intracellular Fc than untransfected cells. Internalization of FcRn-binding forms of Fc was proportional to FcRn expression level, was enriched relative to dextran internalization in proportion to FcRn expression level, and was blocked by incubation with excess unlabeled Fc. Because we were unable to detect either surface expression of FcRn or surface binding of Fc, these results suggest that FcRn-dependent internalization of Fc may occur through sequestration of Fc by FcRn in early endosomes. These studies indicate that FcRn-dependent internalization of IgG may be important not only in cells taking up IgG from an extracellular acidic space, but also in endothelial cells participating in homeostatic regulation of circulating IgG levels.
Collapse
Affiliation(s)
- Nancy A Goebl
- Department of Drug Disposition Development/Commercialization, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | | | | | | | | | | |
Collapse
|
223
|
Ye L, Liu X, Rout SN, Li Z, Yan Y, Lu L, Kamala T, Nanda NK, Song W, Samal SK, Zhu X. The MHC class II-associated invariant chain interacts with the neonatal Fc gamma receptor and modulates its trafficking to endosomal/lysosomal compartments. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:2572-85. [PMID: 18684948 PMCID: PMC2667114 DOI: 10.4049/jimmunol.181.4.2572] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The neonatal Fc receptor for IgG (FcRn) transfers maternal IgG to the offspring and protects IgG from degradation. The FcRn resides in an acidic intracellular compartment, allowing it to bind IgG. In this study, we found the association of FcRn and invariant chain (Ii). The interaction was initiated within the endoplasmic reticulum by Ii binding to either the FcRn H chain alone or FcRn H chain-beta(2)-microglobulin complex and appeared to be maintained throughout the endocytic pathway. The CLIP in Ii was not required for FcRn-Ii association. The interaction was also detected in IFN-gamma-treated THP-1, epithelial and endothelial cells, and immature mouse DCs. A truncated FcRn without the cytoplasmic tail was unable to traffic to early endosomes; however, its location in early endosomes was restored by Ii expression. FcRn was also detected in the late endosome/lysosome only in the presence of Ii or on exposure to IFN-gamma. In immature human or mouse DCs, FcRn was barely detected in the late endosome/lysosome in the absence of Ii. Furthermore, the cytoplasmic tail of Ii conferred tailless FcRn to route to both the early endosome and late endosome/lysosome in a hybrid molecule. Because the FcRn is expressed in macrophages and DCs or epithelial and endothelial cells where Ii is induced under inflammation and infection, these results reveal the complexity of FcRn trafficking in which Ii is capable of expanding the boundary of FcRn trafficking. Taken together, the intracellular trafficking of FcRn is regulated by its intrinsic sorting information and/or an interaction with Ii chain.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Differentiation, B-Lymphocyte/genetics
- Antigens, Differentiation, B-Lymphocyte/metabolism
- Antigens, Differentiation, B-Lymphocyte/physiology
- CHO Cells
- Caco-2 Cells
- Cell Line, Tumor
- Cricetinae
- Cricetulus
- Endosomes/immunology
- Endosomes/metabolism
- Endosomes/pathology
- HT29 Cells
- HeLa Cells
- Histocompatibility Antigens Class I/metabolism
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/metabolism
- Histocompatibility Antigens Class II/physiology
- Humans
- Inflammation Mediators/physiology
- Lysosomes/immunology
- Lysosomes/metabolism
- Lysosomes/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Molecular Sequence Data
- Protein Transport/genetics
- Protein Transport/immunology
- Receptors, Fc/metabolism
Collapse
Affiliation(s)
- Lilin Ye
- Laboratory of Immunology, University of Maryland, College Park, MD 20742
- Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742
| | - Xindong Liu
- Laboratory of Immunology, University of Maryland, College Park, MD 20742
- Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742
| | - Subrat N. Rout
- Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742
| | - Zili Li
- Laboratory of Immunology, University of Maryland, College Park, MD 20742
- Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742
| | - Yongqi Yan
- Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742
| | - Li Lu
- Laboratory of Immunology, University of Maryland, College Park, MD 20742
| | - Tirumalai Kamala
- Laboratory of Cellular and Molecular Immunology, Bethesda, MD 20892
- T-Cell Tolerance and Memory Section, National Institutes of Health, Bethesda, MD 20892
| | - Navreet K. Nanda
- Laboratory of Cellular and Molecular Immunology, Bethesda, MD 20892
| | - Wenxia Song
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
- Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742
| | - Siba K. Samal
- Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742
| | - Xiaoping Zhu
- Laboratory of Immunology, University of Maryland, College Park, MD 20742
- Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742
| |
Collapse
|
224
|
Ligand binding and antigenic properties of a human neonatal Fc receptor with mutation of two unpaired cysteine residues. FEBS J 2008; 275:4097-110. [PMID: 18637944 DOI: 10.1111/j.1742-4658.2008.06551.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The neonatal Fc receptor (FcRn) is a major histocompatibility complex class I-related molecule that regulates the half-life of IgG and albumin. In addition, FcRn directs the transport of IgG across both mucosal epithelium and placenta and also enhances phagocytosis in neutrophils. This new knowledge gives incentives for the design of IgG and albumin-based diagnostics and therapeutics. To study FcRn in vitro and to select and characterize FcRn binders, large quantities of soluble human FcRn are needed. In this report, we explored the impact of two free cysteine residues (C48 and C251) of the FcRn heavy chain on the overall structure and function of soluble human FcRn and described an improved bacterial production strategy based on removal of these residues, yielding approximately 70 mg.L(-1) of fermentation of refolded soluble human FcRn. The structural and functional integrity was proved by CD, surface plasmon resonance and MALDI-TOF peptide mapping analyses. The strategy may generally be translated to the large-scale production of other major histocompatibility complex class I-related molecules with nonfunctional unpaired cysteine residues. Furthermore, the anti-FcRn response in goats immunized with the FcRn heavy chain alone was analyzed following affinity purification on heavy chain-coupled Sepharose. Importantly, purified antibodies blocked the binding of both ligands to soluble human FcRn and were thus directed to both binding sites. This implies that the FcRn heavy chain, without prior assembly with human beta2-microglobulin, contains the relevant epitopes found in soluble human FcRn, and is therefore sufficient to obtain binders to either ligand-binding site. This finding will greatly facilitate the selection and characterization of such binders.
Collapse
|
225
|
Liu X, Ye L, Bai Y, Mojidi H, Simister NE, Zhu X. Activation of the JAK/STAT-1 signaling pathway by IFN-gamma can down-regulate functional expression of the MHC class I-related neonatal Fc receptor for IgG. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:449-63. [PMID: 18566411 PMCID: PMC2667120 DOI: 10.4049/jimmunol.181.1.449] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Expression of many MHC genes is enhanced at the transcriptional or posttranscriptional level following exposure to the cytokine IFN-gamma. However, in this study we found that IFN-gamma down-regulated the constitutive expression of the neonatal Fc receptor (FcRn), an MHC class I-related molecule that functions to transport maternal IgG and protect IgG and albumin from degradation. Epithelial cell, macrophage-like THP-1 cell, and freshly isolated human PBMC exposure to IFN-gamma resulted in a significant decrease of FcRn expression as assessed by real-time RT-PCR and Western blotting. The down-regulation of FcRn was not caused by apoptosis or the instability of FcRn mRNA. Chromatin immunoprecipitation and gel mobility shift assays showed that STAT-1 bound to an IFN-gamma activation site in the human FcRn promoter region. Luciferase expression from an FcRn promoter-luciferase reporter gene construct was not altered in JAK1- and STAT-1-deficient cells following exposure to IFN-gamma, whereas expression of JAK1 or STAT-1 protein restored the IFN-gamma inhibitory effect on luciferase activity. The repressive effect of IFN-gamma on the FcRn promoter was selectively reversed or blocked by mutations of the core nucleotides in the IFN-gamma activation site sequence and by overexpression of the STAT-1 inhibitor PIAS1 or the dominant negative phospho-STAT-1 mutations at Tyr-701 and/or Ser-727 residues. Furthermore, STAT-1 might down-regulate FcRn transcription through sequestering the transcriptional coactivator CREB binding protein/p300. Functionally, IFN-gamma stimulation dampened bidirectional transport of IgG across a polarized Calu-3 lung epithelial monolayer. Taken together, our results indicate that the JAK/STAT-1 signaling pathway was necessary and sufficient to mediate the down-regulation of FcRn gene expression by IFN-gamma.
Collapse
Affiliation(s)
- Xindong Liu
- Laboratory of Immunology, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742
| | - Lilin Ye
- Laboratory of Immunology, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742
| | - Yu Bai
- Laboratory of Immunology, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742
| | - Habi Mojidi
- Laboratory of Immunology, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742
- Maryland Pathogen Research Institute, Graduate Program in Molecular and Cell Biology, University of Maryland, College Park, MD 20742
| | - Neil E. Simister
- Rosenstiel Center for Basic Biomedical Sciences and Biology Department, Brandeis University, Waltham, MA 02254
| | - Xiaoping Zhu
- Laboratory of Immunology, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742
- Maryland Pathogen Research Institute, Graduate Program in Molecular and Cell Biology, University of Maryland, College Park, MD 20742
| |
Collapse
|
226
|
Andersen JT, Justesen S, Berntzen G, Michaelsen TE, Lauvrak V, Fleckenstein B, Buus S, Sandlie I. A strategy for bacterial production of a soluble functional human neonatal Fc receptor. J Immunol Methods 2008; 331:39-49. [DOI: 10.1016/j.jim.2007.11.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2007] [Revised: 09/19/2007] [Accepted: 11/13/2007] [Indexed: 12/31/2022]
|
227
|
Structural basis for potent cross-neutralizing human monoclonal antibody protection against lethal human and zoonotic severe acute respiratory syndrome coronavirus challenge. J Virol 2008; 82:3220-35. [PMID: 18199635 DOI: 10.1128/jvi.02377-07] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV) emerged in 2002, and detailed phylogenetic and epidemiological analyses have suggested that it originated from animals. The spike (S) glycoprotein has been identified as a major component of protective immunity, and 23 different amino acid changes were noted during the expanding epidemic. Using a panel of SARS-CoV recombinants bearing the S glycoproteins from isolates representing the zoonotic and human early, middle, and late phases of the epidemic, we identified 23 monoclonal antibodies (MAbs) with neutralizing activity against one or multiple SARS-CoV spike variants and determined the presence of at least six distinct neutralizing profiles in the SARS-CoV S glycoprotein. Four of these MAbs showed cross-neutralizing activity against all human and zoonotic S variants in vitro, and at least three of these were mapped in distinct epitopes using escape mutants, structure analyses, and competition assays. These three MAbs (S109.8, S227.14, and S230.15) were tested for use in passive vaccination studies using lethal SARS-CoV challenge models for young and senescent mice with four different homologous and heterologous SARS-CoV S variants. Both S227.14 and S230.15 completely protected young and old mice from weight loss and virus replication in the lungs for all viruses tested, while S109.8 completely protected mice from weight loss and clinical signs in the presence of viral titers. We conclude that a single human MAb can confer broad protection against lethal challenge with multiple zoonotic and human SARS-CoV isolates, and we identify a robust cocktail formulation that targets distinct epitopes and minimizes the likely generation of escape mutants.
Collapse
|
228
|
Podocytes use FcRn to clear IgG from the glomerular basement membrane. Proc Natl Acad Sci U S A 2008; 105:967-72. [PMID: 18198272 DOI: 10.1073/pnas.0711515105] [Citation(s) in RCA: 190] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The glomerular filtration barrier prevents large serum proteins from being lost into the urine. It is not known, however, why the filter does not routinely clog with large proteins that enter the glomerular basement membrane (GBM). Here, we provide evidence that an active transport mechanism exists to remove immunoglobulins that accumulate at the filtration barrier. We found that FcRn, an IgG and albumin transport receptor, is expressed in podocytes and functions to internalize IgG from the GBM. Mice lacking FcRn accumulated IgG in the GBM as they aged, and tracer studies showed delayed clearance of IgG from the kidneys of FcRn-deficient mice. Supporting a role for this pathway in disease, saturating the clearance mechanism potentiated the pathogenicity of nephrotoxic sera. These studies support the idea that podocytes play an active role in removing proteins from the GBM and suggest that genetic or acquired impairment of the clearance machinery is likely to be a common mechanism promoting glomerular diseases.
Collapse
|
229
|
Pandolfino JE, Ghosh SK, Rice J, Clarke JO, Kwiatek MA, Kahrilas PJ. Classifying esophageal motility by pressure topography characteristics: a study of 400 patients and 75 controls. Am J Gastroenterol 2008; 103:27-37. [PMID: 17900331 DOI: 10.1111/j.1572-0241.2007.01532.x] [Citation(s) in RCA: 291] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
AIM This study aimed to devise a scheme for the systematic analysis of esophageal high-resolution manometry (HRM) studies displayed using topographic plotting. METHODS A total of 400 patients and 75 control subjects were studied with a 36-channel HRM assembly. Studies were analyzed in a stepwise fashion for: (a) the adequacy of deglutitive esophagogastric junction (EGJ) relaxation, (b) the presence and propagation characteristics of distal esophageal persitalsis, and (c) an integral of the magnitude and span of the distal esophageal contraction. RESULTS Two strengths of pressure topography plots compared to conventional manometric recordings were: (a) the ability to delineate the spatial limits, vigor, and integrity of individual contractile segments along the esophagus, and (b) the ability to distinguish between loci of compartmentalized intraesophageal pressurization and rapidly propagated contractions. Making these distinctions objectified the identification of distal esophageal spasm (DES), vigorous achalasia, functional obstruction, and nutcracker esophagus subtypes. Applying these distinctions made the diagnosis of spastic disorders quite rare: (a) DES in 1.5% patients, (b) vigorous achalasia in 1.5%, and (c) a newly defined entity, spastic nutcracker, in 1.5%. CONCLUSIONS We developed a systematic approach to analyzing esophageal motility using HRM and pressure topography plots. The resultant scheme is consistent with conventional classifications with the caveats that: (a) hypercontractile conditions are more specifically defined, (b) distinctions are made between rapidly propagated contractions and compartmentalized esophageal pressurization, and (c) there is no "nonspecific esophageal motor disorder" classification. We expect that pressure topography analysis, with its well-defined functional implications, will prove valuable in the clinical management of esophageal motility disorders.
Collapse
Affiliation(s)
- John E Pandolfino
- Department of Medicine, The Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | | | | | | | | | | |
Collapse
|
230
|
Dickinson BL, Claypool SM, D'Angelo JA, Aiken ML, Venu N, Yen EH, Wagner JS, Borawski JA, Pierce AT, Hershberg R, Blumberg RS, Lencer WI. Ca2+-dependent calmodulin binding to FcRn affects immunoglobulin G transport in the transcytotic pathway. Mol Biol Cell 2008; 19:414-23. [PMID: 18003977 PMCID: PMC2174181 DOI: 10.1091/mbc.e07-07-0658] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2007] [Revised: 10/09/2007] [Accepted: 11/02/2007] [Indexed: 11/11/2022] Open
Abstract
The Fcgamma receptor FcRn transports immunoglobulin G (IgG) so as to avoid lysosomal degradation and to carry it bidirectionally across epithelial barriers to affect mucosal immunity. Here, we identify a calmodulin-binding site within the FcRn cytoplasmic tail that affects FcRn trafficking. Calmodulin binding to the FcRn tail is direct, calcium-dependent, reversible, and specific to residues comprising a putative short amphipathic alpha-helix immediately adjacent to the membrane. FcRn mutants with single residue substitutions in this motif, or FcRn mutants lacking the cytoplasmic tail completely, exhibit a shorter half-life and attenuated transcytosis. Chemical inhibitors of calmodulin phenocopy the mutant FcRn defect in transcytosis. These results suggest a novel mechanism for regulation of IgG transport by calmodulin-dependent sorting of FcRn and its cargo away from a degradative pathway and into a bidirectional transcytotic route.
Collapse
Affiliation(s)
- Bonny L. Dickinson
- *The Research Institute for Children, Children's Hospital, Department of Pediatrics, New Orleans, LA 70118
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Science Center, New Orleans, LA 70112
| | - Steven M. Claypool
- Division of Gastroenterology, Brigham and Women's Hospital and the Department of Medicine, Harvard Medical School, Boston, MA 02115
| | - June A. D'Angelo
- *The Research Institute for Children, Children's Hospital, Department of Pediatrics, New Orleans, LA 70118
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Science Center, New Orleans, LA 70112
| | - Martha L. Aiken
- *The Research Institute for Children, Children's Hospital, Department of Pediatrics, New Orleans, LA 70118
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Science Center, New Orleans, LA 70112
| | - Nanda Venu
- Gastrointestinal Cell Biology, Division of Pediatric Gastroenterology and Nutrition, Children's Hospital and the Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Elizabeth H. Yen
- Gastrointestinal Cell Biology, Division of Pediatric Gastroenterology and Nutrition, Children's Hospital and the Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Jessica S. Wagner
- Gastrointestinal Cell Biology, Division of Pediatric Gastroenterology and Nutrition, Children's Hospital and the Department of Pediatrics, Harvard Medical School, Boston, MA 02115
- Harvard Digestive Diseases Center, Boston, MA 02115
| | - Jason A. Borawski
- Gastrointestinal Cell Biology, Division of Pediatric Gastroenterology and Nutrition, Children's Hospital and the Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Amy T. Pierce
- *The Research Institute for Children, Children's Hospital, Department of Pediatrics, New Orleans, LA 70118
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Science Center, New Orleans, LA 70112
| | - Robert Hershberg
- Department of Medicine and Medical Genetics, University of Washington School of Medicine, Seattle, WA 98112; and
| | - Richard S. Blumberg
- Division of Gastroenterology, Brigham and Women's Hospital and the Department of Medicine, Harvard Medical School, Boston, MA 02115
- Harvard Digestive Diseases Center, Boston, MA 02115
| | - Wayne I. Lencer
- Gastrointestinal Cell Biology, Division of Pediatric Gastroenterology and Nutrition, Children's Hospital and the Department of Pediatrics, Harvard Medical School, Boston, MA 02115
- Harvard Digestive Diseases Center, Boston, MA 02115
| |
Collapse
|
231
|
Liu X, Ye L, Christianson GJ, Yang JQ, Roopenian DC, Zhu X. NF-kappaB signaling regulates functional expression of the MHC class I-related neonatal Fc receptor for IgG via intronic binding sequences. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 179:2999-3011. [PMID: 17709515 PMCID: PMC2667116 DOI: 10.4049/jimmunol.179.5.2999] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The neonatal Fc receptor for IgG (FcRn) functions to transport maternal IgG to a fetus or newborn and to protect IgG from degradation. Although FcRn is expressed in a variety of tissues and cell types, the extent to which FcRn expression is regulated by immunological and inflammatory events remains unknown. Stimulation of intestinal epithelial cell lines, macrophage-like THP-1, and freshly isolated human monocytes with the cytokine TNF-alpha rapidly up-regulated FcRn gene expression. In addition, the TLR ligands LPS and CpG oligodeoxynucleotide enhanced the level of FcRn expression in THP-1 and monocytes. Treatment of TNF-stimulated THP-1 cells with the NF-kappaB-specific inhibitor or overexpression of a dominant negative mutant inhibitory NF-kappaB (IkappaBalpha; S32A/S36A) resulted in down-regulation of FcRn expression. By using chromatin immunoprecipitation we identified three NF-kappaB binding sequences within introns 2 and 4 of the human FcRn gene. An EMSA confirmed the p50/p50 and/or p65/p50 complex (s) bound to intron 2- or 4-derived oligonucleotides containing putative NF-kappaB binding sequences, respectively. The intronic NF-kappaB sequences in combination with the promoter or alone regulated the expression of a luciferase reporter gene in response to TNF-alpha stimulation or overexpression of NF-kappaB p65 and p50. DNA looping interactions potentially occurred after the stimulation between intronic NF-kappaB sequences and the FcRn promoter as shown by a chromosome conformation capture assay. Finally, TNF-alpha stimulations enhanced IgG transport across an intestinal Caco-2 epithelial monolayer. Together, these data provide the first evidence that NF-kappaB signaling via intronic sequences regulates FcRn expression and function.
Collapse
Affiliation(s)
- Xindong Liu
- Laboratory of Immunology, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742
| | - Lilin Ye
- Laboratory of Immunology, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742
| | | | - Jun-Qi Yang
- Department of Genome Science, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | | | - Xiaoping Zhu
- Laboratory of Immunology, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742
| |
Collapse
|
232
|
Abstract
The neonatal Fc receptor for IgG (FcRn) has been well characterized in the transfer of passive humoral immunity from a mother to her fetus. In addition, throughout life, FcRn protects IgG from degradation, thereby explaining the long half-life of this class of antibody in the serum. In recent years, it has become clear that FcRn is expressed in various sites in adults, where its potential function is now beginning to emerge. In addition, recent studies have examined the interaction between FcRn and the Fc portion of IgG with the aim of either improving the serum half-life of therapeutic monoclonal antibodies or reducing the half-life of pathogenic antibodies. This Review summarizes these two areas of FcRn biology.
Collapse
Affiliation(s)
- Derry C Roopenian
- The Jackson Laboratory, 600 Main Street, Bar Harbor, Maine 04609, USA.
| | | |
Collapse
|
233
|
Slizhikova DK, Zinovyeva MV, Kuzmin DV, Snezhkov EV, Shakhparonov MI, Dmitriev RI, Antipova NV, Zavalova LL, Sverdlov ED. Decreased expression of the human immunoglobulin J-chain gene in squamous cell cancer and adenocarcinoma of the lungs. Mol Biol 2007. [DOI: 10.1134/s0026893307040115] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
234
|
Garg A, Balthasar JP. Physiologically-based pharmacokinetic (PBPK) model to predict IgG tissue kinetics in wild-type and FcRn-knockout mice. J Pharmacokinet Pharmacodyn 2007; 34:687-709. [PMID: 17636457 DOI: 10.1007/s10928-007-9065-1] [Citation(s) in RCA: 258] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2007] [Accepted: 05/25/2007] [Indexed: 12/30/2022]
Abstract
Although it is known that FcRn, the neonatal Fc-receptor, functions to protect immune gamma globulin (IgG) from elimination, the influence of FcRn on the tissue distribution of IgG has not been quantified. In the present work, a physiologically-based pharmacokinetic (PBPK) model has been developed to characterize and predict IgG disposition in plasma and in tissues. The model includes nine major compartments, connected in an anatomical manner, to represent tissues known to play a significant role in IgG disposition. Each tissue compartment was subdivided into vascular, endosomal and interstitial spaces. IgG transport between the blood and interstitial compartments may proceed by convection through paracellular pores in the vascular endothelium, or via FcRn-mediated transcytosis across vascular endosomal cells. The model was utilized to characterize plasma concentration-time data for 7E3, a monoclonal antiplatelet IgG1 antibody, in control and FcRn-knockout (KO) mice. These data showed that high dose intravenous immunoglobulin (IVIG), 1g/kg, increased 7E3 clearance in control mice from 5.2 +/- 0.3 to 14.4 +/- 1.4 ml/d/kg; however, IVIG failed to increase the clearance of 7E3 in KO mice (72.5 +/- 4.0 vs. 61.0 +/- 3.6 ml/d/kg). Based on model fitting to the 7E3 plasma concentration data, simulations were conducted to predict tissue concentrations of IgG in control and in KO mice, and the predictions were then tested by assessing 7E3 tissue distribution in KO mice and control mice. 7E3 was radiolabeled with Iodine-125 using chloramine T method, and (125)I-7E3 IgG was administered at a dose of 8 mg/kg to control and KO mice. At various time points, sub-groups of 3 mice were sacrificed, blood and tissue samples were collected, and radioactivity assessed by gamma counting. PBPK model performance was assessed by comparing model predictions with the observed data. The model accurately predicted 7E3 tissue concentrations, with mean predicted vs. observed AUC ratios of 1.04 +/- 0.2 and 0.86 +/- 0.3 in control and FcRn-KO mice. The PBPK model, which incorporates the influence of FcRn on IgG clearance and disposition, was found to provide accurate predictions of IgG tissue kinetics in control and FcRn-knockout mice.
Collapse
Affiliation(s)
- Amit Garg
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, Buffalo, NY 14260, USA
| | | |
Collapse
|
235
|
Burmakin MV, Seliverstova EV, Natochin YV. Dynamics of absorption in gut and reabsorption in kidney of yellow fluorescent protein in rat postnatal ontogenesis. J EVOL BIOCHEM PHYS+ 2007. [DOI: 10.1134/s0022093007020111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
236
|
Prabhat P, Gan Z, Chao J, Ram S, Vaccaro C, Gibbons S, Ober RJ, Ward ES. Elucidation of intracellular recycling pathways leading to exocytosis of the Fc receptor, FcRn, by using multifocal plane microscopy. Proc Natl Acad Sci U S A 2007; 104:5889-94. [PMID: 17384151 PMCID: PMC1851587 DOI: 10.1073/pnas.0700337104] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The intracellular events on the recycling pathway that lead from sorting endosomes to exocytosis at the plasma membrane are central to cellular function. However, despite intensive study, these processes are poorly characterized in spatial and dynamic terms. The primary reason for this is that, to date, it has not been possible to visualize rapidly moving intracellular compartments in three dimensions in cells. Here, we use a recently developed imaging setup in which multiple planes can be simultaneously imaged within the cell in conjunction with visualization of the plasma membrane plane by using total internal reflection fluorescence microscopy. This has allowed us to track and characterize intracellular events on the recycling pathway that lead to exocytosis of the MHC Class I-related receptor, FcRn. We observe both direct delivery of tubular and vesicular transport containers (TCs) from sorting endosomes to exocytic sites at the plasma membrane, and indirect pathways in which TCs that are not in proximity to sorting endosomes undergo exocytosis. TCs can also interact with different sorting endosomes before exocytosis. Our data provide insight into the intracellular events that precede exocytic fusion.
Collapse
Affiliation(s)
- Prashant Prabhat
- *Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390; and
- Department of Electrical Engineering, University of Texas at Dallas, Richardson, TX 75080
| | - Zhuo Gan
- *Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390; and
| | - Jerry Chao
- *Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390; and
- Department of Electrical Engineering, University of Texas at Dallas, Richardson, TX 75080
| | - Sripad Ram
- *Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390; and
| | - Carlos Vaccaro
- *Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390; and
| | - Steven Gibbons
- *Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390; and
| | - Raimund J. Ober
- *Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390; and
- Department of Electrical Engineering, University of Texas at Dallas, Richardson, TX 75080
- To whom correspondence may be addressed. E-mail: or
| | - E. Sally Ward
- *Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390; and
- To whom correspondence may be addressed. E-mail: or
| |
Collapse
|
237
|
Seliverstova EV, Burmakin MV, Natochin YV. Renal clearance of absorbed intact GFP in the frog and rat intestine. Comp Biochem Physiol A Mol Integr Physiol 2007; 147:1067-73. [PMID: 17442603 DOI: 10.1016/j.cbpa.2007.03.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2007] [Revised: 03/12/2007] [Accepted: 03/13/2007] [Indexed: 11/15/2022]
Abstract
Intestine absorption of intact green fluorescent protein (GFP) and its following accumulation in the renal proximal tubule cells after its intragastric administration have been established by confocal microscopy in the rat and frog. Reabsorbed GFP was revealed in the endosomes and lysosomes of the proximal tubule cells by the methods of GFP photooxidation and immunofluorescent microscopy. The GFP intestine absorption rate and GFP accumulation in the kidney were significantly higher in the frog than in the rat. No specific fluorescence was revealed in the liver and colon cells after the GFP intragastric administration. The data obtained indicate the ability of the small intestine in the frog and rat to absorb intact proteins and an important role of the kidney in exogenous protein metabolism.
Collapse
Affiliation(s)
- E V Seliverstova
- Laboratory of Renal Physiology, Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | | | | |
Collapse
|
238
|
Qiao SW, Lencer WI, Blumberg RS. How the controller is controlled - neonatal Fc receptor expression and immunoglobulin G homeostasis. Immunology 2007; 120:145-7. [PMID: 17140402 PMCID: PMC2265855 DOI: 10.1111/j.1365-2567.2006.02507.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2006] [Revised: 09/27/2006] [Accepted: 09/27/2006] [Indexed: 11/29/2022] Open
Affiliation(s)
- Shuo-Wang Qiao
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | | | | |
Collapse
|
239
|
Yuji M, Fujimoto M, Miyata H, Inamoto T, Qi WM, Yamamoto K, Warita K, Yokoyama T, Hoshi N, Kitagawa H. Persorption Mechanisms of Luminal Antigenic Particulates via Apoptotic Epithelial Cells of Intestinal Villi into Systemic Blood Circulation in Orally Immunized Rats. J Vet Med Sci 2007; 69:339-46. [PMID: 17485920 DOI: 10.1292/jvms.69.339] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The possibility of persorption of prefixed bovine serum albumin-coated sheep erythrocytes (BSA-SEs) from mucous epithelial cells and its mechanisms were investigated in rats orally immunized by BSA for 14 consecutive days. On the day after the final oral immunization, the rats were duodenally perfused by BSA-SEs or non-coated SEs. BSA-SEs were also duodenally perfused in non-immunized rats. Thirty min after perfusion, BSA-SEs were significantly more engulfed by late-apoptotic-stage villous columnar epithelial cells in the orally immunized rats than those in other experiments. The specific antibody (SpAb) was detected on the surfaces of BSA-SEs in rats with oral immunization. In Peyer's patches of all animals, no SEs reached the follicle-associated epithelium, because of the close attachment of follicle-associated intestinal villi and the thick mucous layer. BSA-SEs were more frequently persorbed into portal blood in the orally immunized rats than in other rats. Small numbers of BSA-SEs or SEs were detected in the systemic blood of all animals. BSA-SEs were also histologically found in the blood vessels of the liver, but not in mesenteric lymph nodes. These findings suggest that sensitized antigenic particulates are taken up by late-apoptotic-stage villous columnar epithelial cells in the small intestine and are finally persorbed into the systemic blood circulation. The uptake of antigenic particulates might be mediated by its luminal SpAb.
Collapse
Affiliation(s)
- Midori Yuji
- Department of Bioresource and Agrobiosciences, Graduate School of Science and Technology, Kobe University, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
240
|
Vaccaro C, Bawdon R, Wanjie S, Ober RJ, Ward ES. Divergent activities of an engineered antibody in murine and human systems have implications for therapeutic antibodies. Proc Natl Acad Sci U S A 2006; 103:18709-14. [PMID: 17116867 PMCID: PMC1693727 DOI: 10.1073/pnas.0606304103] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The MHC class I-related receptor, neonatal Fc receptor (FcRn), plays a central role in regulating the transport and in vivo persistence of immunoglobulin G (IgG). IgG-FcRn interactions can be targeted for engineering to modulate the in vivo longevity and transport of an antibody, and this has implications for the successful application of therapeutic IgGs. Although mice are widely used to preclinically test antibodies, human and mouse FcRn have significant differences in binding specificity. Here we show that an engineered human IgG1 has disparate properties in murine and human systems. The mutant shows improved transport relative to wild-type human IgG1 in assays of human FcRn function but has short in vivo persistence and competitively inhibits FcRn activity in mice. These studies indicate potential limitations of using mice as preclinical models for the analysis of engineered antibodies. Alternative assays are proposed that serve as indicators of the properties of IgGs in humans.
Collapse
Affiliation(s)
- Carlos Vaccaro
- *Center for Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390-9093
| | - Roger Bawdon
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9032; and
| | - Sylvia Wanjie
- *Center for Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390-9093
| | - Raimund J. Ober
- *Center for Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390-9093
- Department of Electrical Engineering, University of Texas at Dallas, Richardson, TX 75083
| | - E. Sally Ward
- *Center for Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390-9093
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
241
|
Melkebeek V, Sonck E, Verdonck F, Goddeeris BM, Cox E. Optimized FaeG expression and a thermolabile enterotoxin DNA adjuvant enhance priming of an intestinal immune response by an FaeG DNA vaccine in pigs. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2006; 14:28-35. [PMID: 17108289 PMCID: PMC1797719 DOI: 10.1128/cvi.00268-06] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
One of the problems hindering the development of DNA vaccines is the relatively low immunogenicity often seen in humans and large animals compared to that in mice. In the present study, we tried to enhance the immunogenicity of a pcDNA1/faeG19 DNA vaccine in pigs by optimizing the FaeG expression plasmid and by coadministration of the plasmid vectors encoding the A and B subunits of the Escherichia coli thermolabile enterotoxin (LT). The insertion of a Kozak sequence and optimization of vector (cellular localization and expression) and both vector and codon usage were all shown to enhance in vitro FaeG expression compared to that of pcDNA1/faeG19. Subsequently, pcDNA1/faeG19 and the vector-optimized and the vector-codon-optimized construct were tested for their immunogenicity in pigs. In line with the in vitro results, antibody responses were better induced with increasing expression. The LT vectors additionally enhanced the antibody response, although not significantly, and were necessary to induce an F4-specific cellular response. These vectors were also added because LT has been described to direct the systemic response towards a mucosal immunoglobulin A (IgA) response in mice. Here, however, the intradermal FaeG DNA prime-oral F4 boost immunization resulted in a mainly systemic IgG response, with only a marginal but significant reduction in F4+ E. coli fecal excretion when the piglets were primed with pWRGFaeGopt and pWRGFaeGopt with the LT vectors.
Collapse
Affiliation(s)
- V Melkebeek
- Laboratory of Veterinary Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium.
| | | | | | | | | |
Collapse
|
242
|
Berin MC, Li H, Sperber K. Antibody-mediated antigen sampling across intestinal epithelial barriers. Ann N Y Acad Sci 2006; 1072:253-61. [PMID: 17057205 DOI: 10.1196/annals.1326.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The epithelium of the gastrointestinal tract is the interface between luminal contents and the mucosal immune system. It must function as a selective barrier to limit penetration of antigens yet keep the mucosal immune system "informed" for the purpose of generating oral tolerance responses to food antigens or commensal organisms and host defense responses against pathogens. Alterations in epithelial barrier function have been proposed to play a significant role in gastrointestinal disease. In this review, we will discuss mechanisms of regulation of epithelial barrier function, and we will focus on the emerging understanding of how secreted immunoglobulins play a role in antigen-specific antigen sampling across the gastrointestinal epithelium.
Collapse
Affiliation(s)
- M Cecilia Berin
- Department of Pediatrics, Jaffe Food Allergy Institute, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA.
| | | | | |
Collapse
|
243
|
Bitonti AJ, Dumont JA. Pulmonary administration of therapeutic proteins using an immunoglobulin transport pathway. Adv Drug Deliv Rev 2006; 58:1106-18. [PMID: 16997417 DOI: 10.1016/j.addr.2006.07.015] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2005] [Accepted: 07/25/2006] [Indexed: 10/24/2022]
Abstract
We have applied a "physiologic" approach to the pulmonary delivery of therapeutic proteins, utilizing an immunoglobulin (antibody) transport pathway recently shown to be present predominantly in the conducting airways of the human respiratory tract. Therapeutic proteins are fused to the Fc-domain of an IgG1, allowing them to bind with high affinity to the antibody transport receptor, FcRn. Liquid aerosols are administered into the lung using normal breathing maneuvers and efficient delivery of several different Fc-fusion proteins has been achieved with retention of biological activity and an increase in circulating half-life. A new paradigm for the pulmonary delivery of therapeutic proteins and a fundamental advance in the construction of Fc-fusion proteins for this purpose will be described.
Collapse
Affiliation(s)
- Alan J Bitonti
- Syntonix Pharmaceuticals, 9 Fourth Avenue, Waltham, MA 02451, USA.
| | | |
Collapse
|
244
|
Yoshida M, Masuda A, Kuo TT, Kobayashi K, Claypool SM, Takagawa T, Kutsumi H, Azuma T, Lencer WI, Blumberg RS. IgG transport across mucosal barriers by neonatal Fc receptor for IgG and mucosal immunity. ACTA ACUST UNITED AC 2006; 28:397-403. [PMID: 17051393 DOI: 10.1007/s00281-006-0054-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2006] [Accepted: 10/03/2006] [Indexed: 12/21/2022]
Abstract
Mucosal secretions of the human gastrointestinal, respiratory, and genital tracts contain significant quantities of IgG. The neonatal Fc receptor for IgG (FcRn) plays a major role in regulating host IgG levels and transporting IgG and associated antigens across polarized epithelial barriers. The FcRn can then recycle the IgG/antigen complex back across the intestinal barrier into the lamina propria for processing by dendritic cells and presentation to CD4(+) T cells in regional organized lymphoid structures. FcRn, through its ability to secrete and absorb IgG, thus integrates luminal antigen encounters with systemic immune compartments and, as such, provides essential host defense and immunoregulatory functions at the mucosal surfaces.
Collapse
Affiliation(s)
- Masaru Yoshida
- Frontier Medical Science in Gastroenterology, ICMRT, Kobe University School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
245
|
Harris NL, Spoerri I, Schopfer JF, Nembrini C, Merky P, Massacand J, Urban JF, Lamarre A, Burki K, Odermatt B, Zinkernagel RM, Macpherson AJ. Mechanisms of Neonatal Mucosal Antibody Protection. THE JOURNAL OF IMMUNOLOGY 2006; 177:6256-62. [PMID: 17056555 DOI: 10.4049/jimmunol.177.9.6256] [Citation(s) in RCA: 169] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Following an abrupt transition at birth from the sterile uterus to an environment with abundant commensal and pathogenic microbes, neonatal mammals are protected by maternal Abs at mucosal surfaces. We show in mice that different Ab isotypes work in distinct ways to protect the neonatal mucosal surface. Secretory IgA acts to limit penetration of commensal intestinal bacteria through the neonatal intestinal epithelium: an apparently primitive process that does not require diversification of the primary natural Ab repertoire. In contrast, neonatal protection against the exclusively luminal parasite Heligmosomoides polygyrus required IgG from primed females. This immune IgG could either be delivered directly in milk or retrotransported via neonatal Fc receptor from the neonatal serum into the intestinal lumen to exert its protective effect.
Collapse
Affiliation(s)
- Nicola L Harris
- Institute of Experimental Immunology, Universitätsspital, Zürich, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
246
|
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. [PMID: 16841095 PMCID: PMC1501111 DOI: 10.1172/jci27821] [Citation(s) in RCA: 188] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2006] [Accepted: 05/16/2006] [Indexed: 01/22/2023] Open
Abstract
The neonatal Fc receptor for IgG (FcRn) plays a major role in regulating host IgG levels and transporting IgG and associated antigens across polarized epithelial barriers. Selective expression of FcRn in the epithelium is shown here to be associated with secretion of IgG into the lumen that allows for defense against an epithelium-associated pathogen (Citrobacter rodentium). This pathway of host resistance to a bacterial pathogen as mediated by FcRn involves retrieval of bacterial antigens from the lumen and initiation of adaptive immune responses in regional lymphoid structures. Epithelial-associated FcRn, through its ability to secrete and absorb IgG, may thus integrate luminal antigen encounters with systemic immune compartments and as such provide essential host defense and immunoregulatory functions at the mucosal surfaces.
Collapse
Affiliation(s)
- Masaru Yoshida
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Kanna Kobayashi
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Timothy T. Kuo
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Lynn Bry
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Jonathan N. Glickman
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Steven M. Claypool
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Arthur Kaser
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Takashi Nagaishi
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Darren E. Higgins
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Emiko Mizoguchi
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Yoshio Wakatsuki
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Derry C. Roopenian
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Atsushi Mizoguchi
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Wayne I. Lencer
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Richard S. Blumberg
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| |
Collapse
|
247
|
Tesar DB, Tiangco NE, Bjorkman PJ. Ligand valency affects transcytosis, recycling and intracellular trafficking mediated by the neonatal Fc receptor. Traffic 2006; 7:1127-42. [PMID: 17004319 PMCID: PMC1618808 DOI: 10.1111/j.1600-0854.2006.00457.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2005] [Accepted: 05/25/2006] [Indexed: 02/03/2023]
Abstract
The neonatal Fc receptor (FcRn) transports IgG across epithelial cell barriers to provide maternal antibodies to offspring and serves as a protection receptor by rescuing endocytosed IgG and albumin from lysosomal degradation. Here we describe the generation of polarized Madin-Darby canine kidney (MDCK) cells expressing rat FcRn (rFcRn) to investigate the potential requirement for ligand bivalency in FcRn-mediated transport. The rFcRn-MDCK cells bind, internalize and bidirectionally transcytose the bivalent ligands IgG and Fc across polarized cell monolayers. However, they cannot be used to study FcRn-mediated transport of the monovalent ligand albumin, as we observe no specific binding, internalization or transcytosis of rat albumin. To address whether ligand bivalency is required for transport, the ability of rFcRn to transcytose and recycle wild-type Fc homodimers (wtFc; two FcRn-binding sites) and a heterodimeric Fc (hdFc; one FcRn-binding site) was compared. We show that ligand bivalency is not required for transcytosis or recycling, but that wtFc is transported more efficiently than hdFc, particularly at lower concentrations. We also demonstrate that hdFc and wtFc have different intracellular fates, with more hdFc than wtFc being trafficked to lysosomes and degraded, suggesting a role for avidity effects in FcRn-mediated IgG transport.
Collapse
Affiliation(s)
- Devin B Tesar
- Division of Biology and California Institute of TechnologyPasadena, CA 91125, USA
| | - Noreen E Tiangco
- Division of Biology and California Institute of TechnologyPasadena, CA 91125, USA
- Howard Hughes Medical Institute, California Institute of TechnologyPasadena, CA 91125, USA
| | - Pamela J Bjorkman
- Division of Biology and California Institute of TechnologyPasadena, CA 91125, USA
- Howard Hughes Medical Institute, California Institute of TechnologyPasadena, CA 91125, USA
| |
Collapse
|
248
|
Hansen GH, Niels-Christiansen LL, Immerdal L, Danielsen EM. Antibodies in the small intestine: mucosal synthesis and deposition of anti-glycosyl IgA, IgM, and IgG in the enterocyte brush border. Am J Physiol Gastrointest Liver Physiol 2006; 291:G82-90. [PMID: 16565420 DOI: 10.1152/ajpgi.00021.2006] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Synthesis and deposition of immunoglobulins in the brush border was studied in organ-cultured pig small intestinal mucosal explants. Surprisingly, comparable amounts of IgM and IgA were synthesized during a 6-h pulse, and also newly made IgG was detected in media and explants, including the microvillar fraction. For IgA and IgM, this subcellular distribution is consistent with basolateral-to-apical transcytosis, mediated by the polymeric immunoglobulin receptor. IgG is a ligand for the Fc receptor FcRn, and beta2-microglobulin, the light chain of FcRn, coclustered in immunogold double labeling with IgG in subapical endosomes and in the basolateral membrane of enterocytes. In addition, beta2-microglobulin was copurified with IgG on protein G-Sepharose. Apical endocytosis of IgG, as judged by internalization of fluorescent protein G, was not detectable except in a few isolated cells. This suggests that IgG in the adult small intestine is transported across the enterocyte mainly in the basolateral to apical direction. Significant fractions of all immunoglobulins bound to lactoseagarose, indicating that "anti-glycosyl" antibodies, raised against commensal gut bacteria, are synthesized locally in the small intestine. By partial deposition in the brush border, these antibodies therefore may have a protective function by preventing lectin-like pathogens from gaining access to the brush border surface.
Collapse
Affiliation(s)
- Gert H Hansen
- Department of Medical Biochemistry and Genetics, The Panum Institute, University of Copenhagen, Copenhagen N, Denmark
| | | | | | | |
Collapse
|
249
|
Dall'Acqua WF, Kiener PA, Wu H. Properties of human IgG1s engineered for enhanced binding to the neonatal Fc receptor (FcRn). J Biol Chem 2006; 281:23514-24. [PMID: 16793771 DOI: 10.1074/jbc.m604292200] [Citation(s) in RCA: 467] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We describe here the functional implications of an increase in IgG binding to the neonatal Fc receptor. We have defined in a systematic fashion the relationship between enhanced FcRn binding of a humanized anti-respiratory syncytial virus (RSV) monoclonal antibody (MEDI-524) and the corresponding biological consequences in cynomolgus monkeys. The triple mutation M252Y/S254T/T256E (YTE) was introduced into the Fc portion of MEDI-524. Whereas these substitutions did not affect the ability of MEDI-524 to bind to its cognate antigen and inhibit RSV replication, they resulted in a 10-fold increase in its binding to both cynomolgus monkey and human FcRn at pH 6.0. MEDI-524-YTE was efficiently released from FcRn at pH 7.4 in both cases. We show that MEDI-524-YTE consistently exhibited a nearly 4-fold increase in serum half-life in cynomolgus monkeys when compared with MEDI-524. This constituted the largest half-life improvement described to date for an IgG in a primate. For the first time, we demonstrate that these sustained serum levels resulted in an up to 4-fold increase in lung bioavailability. Importantly, we also establish that our non-human primate model is relevant to human. Finally, we report that the YTE triple substitution provided a means to modulate the antibody-dependent cell-mediated cytotoxicity (ADCC) activity of a humanized IgG1 directed against the human integrin alpha(v)beta3. Therefore, the YTE substitutions allow the simultaneous modulation of serum half-life, tissue distribution and activity of a given human IgG1.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/blood
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal, Humanized
- Antibody Diversity
- Antibody-Dependent Cell Cytotoxicity
- Binding Sites, Antibody
- Half-Life
- Histocompatibility Antigens Class I/blood
- Histocompatibility Antigens Class I/metabolism
- Humans
- Immunoglobulin G/administration & dosage
- Immunoglobulin G/blood
- Immunoglobulin G/chemistry
- Immunoglobulin G/metabolism
- Macaca fascicularis
- Organ Specificity
- Protein Binding
- Protein Engineering/methods
- Receptors, Fc/blood
- Receptors, Fc/metabolism
- Receptors, IgG/metabolism
Collapse
Affiliation(s)
- William F Dall'Acqua
- Department of Antibody Discovery and Protein Engineering, MedImmune, Inc., Gaithersburg, Maryland 20878, USA. dall'
| | | | | |
Collapse
|
250
|
Maidji E, McDonagh S, Genbacev O, Tabata T, Pereira L. Maternal antibodies enhance or prevent cytomegalovirus infection in the placenta by neonatal Fc receptor-mediated transcytosis. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 168:1210-26. [PMID: 16565496 PMCID: PMC1606573 DOI: 10.2353/ajpath.2006.050482] [Citation(s) in RCA: 184] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
How human cytomegalovirus (CMV) reaches the fetus across the placenta is unknown. The major viral cause of congenital disease, CMV infects the uterine-placental interface with varied outcomes depending on the strength of maternal humoral immunity and gestational age. Covering the surface of villi that float in blood, syncytiotrophoblasts express the neonatal Fc receptor (FcRn) that transports IgG for passive immunity. Immunohistochemical analysis of early-gestation biopsy specimens showed an unusual pattern of CMV replication proteins in underlying villus cytotrophoblasts, whereas syncytiotrophoblasts were spared. Found in placentas with low to moderate CMV-neutralizing antibody titers, this pattern suggested virion transcytosis across the surface. In contrast, syncytiotrophoblasts from placentas with high neutralizing titers contained viral DNA and caveolin-1-positive vesicles in which IgG and CMV glycoprotein B co-localized. In villus explants, IgG-virion transcytosis and macrophage uptake were blocked with trypsin-treatment and soluble protein A. Quantitative analysis in polarized epithelial cells showed that FcRn-mediated transcytosis was blocked by the Fc fragment of IgG, but not F(ab')(2). Our results suggest that CMV virions could disseminate to the placenta by co-opting the receptor-mediated transport pathway for IgG. These findings could explain the efficacy of hyperimmune IgG for treatment of primary CMV infection during gestation and support vaccination.
Collapse
Affiliation(s)
- Ekaterina Maidji
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, 513 Parnassus, San Francisco, San Francisco, CA 94143-0512, USA
| | | | | | | | | |
Collapse
|