51
|
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: 127] [Impact Index Per Article: 7.9] [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
|
52
|
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. THE JOURNAL OF IMMUNOLOGY 2008; 181:2572-85. [PMID: 18684948 DOI: 10.4049/jimmunol.181.4.2572] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [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
Affiliation(s)
- Lilin Ye
- Laboratory of Immunology, Virginia-Maryland Regional College of Veterinary Medicine, College Park, MD 20742, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
53
|
Earp JC, Dubois DC, Almon RR, Jusko WJ. Quantitative dynamic models of arthritis progression in the rat. Pharm Res 2008; 26:196-203. [PMID: 18758921 DOI: 10.1007/s11095-008-9711-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Accepted: 08/12/2008] [Indexed: 11/28/2022]
Abstract
PURPOSE This comparison employs mathematical disease progression models to identify a rat model of arthritis with the least inter-animal variability and features lending to better study designs. METHODS Arthritis was induced with either collagen (CIA) or mycobacterium (AIA) in either Lewis or Dark Agouti (DA) rats. Disease progression was monitored by paw edema and body weight. Models with production, loss, and feedback components were constructed and population analysis using NONMEM software was employed to identify inter-animal variability in the various disease progression parameters. RESULTS Onset time was the only parameter different within all four groups (DA-AIA 11.5 days, DA-CIA 16.5 days, Lewis-AIA 11.9 days, Lewis-CIA 13.9 days). The loss-of-edema rate constant was 20% slower in DA (0.362 h(-1)) than Lewis (0.466 h(-1)) rats. Most models exhibited peak paw edema 20 days post-induction. Edema in CIA returned to 150% of the initial value after the disease peaked. DA rats displayed more severe overall responses. CONCLUSIONS No statistical differences between groups were observed for inter-animal variation in disease onset, progression and severity parameters. Onset time varies and should be noted in the design of future studies. DA rats may offer a more dynamic range of edema response than Lewis rats.
Collapse
Affiliation(s)
- Justin C Earp
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, SUNY, 565 Hochstetter Hall, Buffalo, New York 14260, USA
| | | | | | | |
Collapse
|
54
|
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. THE JOURNAL OF IMMUNOLOGY 2008; 181:449-63. [PMID: 18566411 DOI: 10.4049/jimmunol.181.1.449] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [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, USA
| | | | | | | | | | | |
Collapse
|
55
|
DeCarlo CA, Escott NG, Werner J, Robinson K, Lambert PF, Law RD, Zehbe I. Gene expression analysis of interferon kappa in laser capture microdissected cervical epithelium. Anal Biochem 2008; 381:59-66. [PMID: 18601893 DOI: 10.1016/j.ab.2008.06.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2008] [Revised: 06/05/2008] [Accepted: 06/06/2008] [Indexed: 11/26/2022]
Abstract
Optimal sample handling techniques for tissue preparation and storage, RNA extraction and quantification, and target gene detection are crucial for reliable gene expression analysis. Methods for measuring low-expressing genes, such as interferons, in human cervical samples are not described in the scientific literature. To detect interferon mRNA in human cervical samples we obtained normal and dysplastic frozen and formalin-fixed cervical biopsies from colposcopy. Histopathological diagnosis was performed by one pathologist. Cervical keratinocytes were isolated using laser capture microdissection. Immortalized keratinocytes transduced with or devoid of an HPV oncogene were used for initial method development. RNA from samples was extracted and integrity tested to compare tissue storage and extraction methods. The expression of five housekeeping genes was analyzed in cell lines and patient tissue to permit normalization between samples using quantitative real-time polymerase chain reaction. The usefulness of cDNA amplification was assessed for the detection of low-expressing interferon kappa in cervical tissue. Here we report optimal tissue storage conditions, RNA extraction, sample normalization, and transcript amplification, as well as the sensitivity of quantitative real-time polymerase chain reaction and laser capture microdissection, for interferon kappa detection in cervical tissue. Without these optimized techniques, interferon kappa detection would be unattainable in cervical samples.
Collapse
Affiliation(s)
- Correne A DeCarlo
- Regional Cancer Centre, Thunder Bay Regional Health Sciences Centre, 980 Oliver Road, Thunder Bay, Ont., Canada P7B 6V4
| | | | | | | | | | | | | |
Collapse
|
56
|
Monach PA, Mathis D, Benoist C. The K/BxN arthritis model. CURRENT PROTOCOLS IN IMMUNOLOGY 2008; Chapter 15:15.22.1-15.22.12. [PMID: 18491295 DOI: 10.1002/0471142735.im1522s81] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mice expressing both the T cell receptor (TCR) transgene KRN and the MHC class II molecule A(g7) (K/BxN mice) develop severe inflammatory arthritis, and serum from these mice causes a similar arthritis in a wide range of mouse strains, due to autoantibodies recognizing glucose-6-phosphate isomerase (GPI). K/BxN transgenic mice have been useful for investigating the development of autoimmunity, and the serum transfer model has been particularly valuable in eliciting mechanisms by which anti-GPI autoantibodies induce joint-specific inflammation. This unit describes detailed methods for the maintenance of a K/BxN colony, induction of arthritis by serum transfer, clinical evaluation of arthritis, and measurement of anti-GPI antibodies.
Collapse
Affiliation(s)
- Paul A Monach
- Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | - Diane Mathis
- Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
57
|
Abstract
The remarkable success story of the therapeutic application of pooled immunoglobulin G (IgG) preparations from thousands of donors, the so-called intravenous IgG (IVIG) therapy, to patients with a variety of hematological and immunological disorders began more than half a century ago. Since then, the use of this primary blood product has increased constantly, resulting in the serious danger of shortages in supply. Despite its widespread use and therapeutic success, the mechanisms of action, especially of the anti-inflammatory activity, are only beginning to be understood. In this review, we summarize the clinical use of IVIG for different diseases and discuss recent data on the molecular mechanisms that might explain how this potent drug mediates its activity in vivo.
Collapse
Affiliation(s)
- Falk Nimmerjahn
- Laboratory of Experimental Immunology and Immunotherapy, Nikolaus-Fiebiger-Center for Molecular Medicine, University of Erlangen-Nuremberg, 91054 Erlangen, Germany.
| | | |
Collapse
|
58
|
Reduction of IgG in nonhuman primates by a peptide antagonist of the neonatal Fc receptor FcRn. Proc Natl Acad Sci U S A 2008; 105:2337-42. [PMID: 18272495 DOI: 10.1073/pnas.0708960105] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The neonatal Fc receptor FcRn provides IgG molecules with their characteristically long half-lives in vivo by protecting them from intracellular catabolism and then returning them to the extracellular space. Other investigators have demonstrated that mice lacking FcRn are protected from induction of various autoimmune diseases, presumably because of the accelerated catabolism of pathogenic IgGs in the animals. Therefore, targeting FcRn with a specific inhibitor may represent a unique approach for the treatment of autoimmune disease or other diseases where the reduction of pathogenic IgG will have a therapeutic benefit. Using phage display peptide libraries, we screened for ligands that bound to human FcRn (hFcRn) and discovered a consensus peptide sequence that binds to hFcRn and inhibits the binding of human IgG (hIgG) in vitro. Chemical optimization of the phage-identified sequences yielded the 26-amino acid peptide dimer SYN1436, which is capable of potent in vitro inhibition of the hIgG-hFcRn interaction. Administration of SYN1436 to mice transgenic for hFcRn induced an increase in the rate of catabolism of hIgG in a dose-dependent manner. Treatment of cynomolgus monkeys with SYN1436 led to a reduction of IgG by up to 80% without reducing serum albumin levels that also binds to FcRn. SYN1436 and related peptides thus represent a previously uncharacterized family of potential therapeutic agents for the treatment of humorally mediated autoimmune and other diseases.
Collapse
|
59
|
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. THE JOURNAL OF IMMUNOLOGY 2007; 179:2999-3011. [PMID: 17709515 PMCID: PMC2667116 DOI: 10.4049/jimmunol.179.5.2999] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [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
- Address correspondence and reprint requests to Dr. Xiaoping Zhu, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, 8075 Greenmead Drive, College Park, MD 20742. E-mail address:
| |
Collapse
|
60
|
Singh Jaggi J, Carrasquillo JA, Seshan SV, Zanzonico P, Henke E, Nagel A, Schwartz J, Beattie B, Kappel BJ, Chattopadhyay D, Xiao J, Sgouros G, Larson SM, Scheinberg DA. Improved tumor imaging and therapy via i.v. IgG-mediated time-sequential modulation of neonatal Fc receptor. J Clin Invest 2007; 117:2422-30. [PMID: 17717602 PMCID: PMC1950462 DOI: 10.1172/jci32226] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2007] [Accepted: 06/12/2007] [Indexed: 11/17/2022] Open
Abstract
The long plasma half-life of IgG, while allowing for enhanced tumor uptake of tumor-targeted IgG conjugates, also results in increased background activity and normal-tissue toxicity. Therefore, successful therapeutic uses of conjugated antibodies have been limited to the highly sensitive and readily accessible hematopoietic tumors. We report a therapeutic strategy to beneficially alter the pharmacokinetics of IgG antibodies via pharmacological inhibition of the neonatal Fc receptor (FcRn) using high-dose IgG therapy. IgG-treated mice displayed enhanced blood and whole-body clearance of radioactivity, resulting in better tumor-to-blood image contrast and protection of normal tissue from radiation. Tumor uptake and the resultant therapeutic response was unaltered. Furthermore, we demonstrated the use of this approach for imaging of tumors in humans and discuss its potential applications in cancer imaging and therapy. The ability to reduce the serum persistence of conjugated IgG antibodies after their infusion can enhance their therapeutic index, resulting in improved therapeutic and diagnostic efficacy.
Collapse
Affiliation(s)
- Jaspreet Singh Jaggi
- Molecular Pharmacology and Chemistry Program and
Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Department of Pathology, Weill Medical College of Cornell University, New York, New York, USA.
Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jorge A. Carrasquillo
- Molecular Pharmacology and Chemistry Program and
Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Department of Pathology, Weill Medical College of Cornell University, New York, New York, USA.
Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Surya V. Seshan
- Molecular Pharmacology and Chemistry Program and
Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Department of Pathology, Weill Medical College of Cornell University, New York, New York, USA.
Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Pat Zanzonico
- Molecular Pharmacology and Chemistry Program and
Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Department of Pathology, Weill Medical College of Cornell University, New York, New York, USA.
Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Erik Henke
- Molecular Pharmacology and Chemistry Program and
Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Department of Pathology, Weill Medical College of Cornell University, New York, New York, USA.
Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew Nagel
- Molecular Pharmacology and Chemistry Program and
Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Department of Pathology, Weill Medical College of Cornell University, New York, New York, USA.
Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jazmin Schwartz
- Molecular Pharmacology and Chemistry Program and
Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Department of Pathology, Weill Medical College of Cornell University, New York, New York, USA.
Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Brad Beattie
- Molecular Pharmacology and Chemistry Program and
Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Department of Pathology, Weill Medical College of Cornell University, New York, New York, USA.
Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Barry J. Kappel
- Molecular Pharmacology and Chemistry Program and
Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Department of Pathology, Weill Medical College of Cornell University, New York, New York, USA.
Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Debjit Chattopadhyay
- Molecular Pharmacology and Chemistry Program and
Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Department of Pathology, Weill Medical College of Cornell University, New York, New York, USA.
Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jing Xiao
- Molecular Pharmacology and Chemistry Program and
Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Department of Pathology, Weill Medical College of Cornell University, New York, New York, USA.
Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - George Sgouros
- Molecular Pharmacology and Chemistry Program and
Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Department of Pathology, Weill Medical College of Cornell University, New York, New York, USA.
Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Steven M. Larson
- Molecular Pharmacology and Chemistry Program and
Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Department of Pathology, Weill Medical College of Cornell University, New York, New York, USA.
Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David A. Scheinberg
- Molecular Pharmacology and Chemistry Program and
Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Department of Pathology, Weill Medical College of Cornell University, New York, New York, USA.
Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
61
|
Nandakumar KS, Holmdahl R. Antibody-induced arthritis: disease mechanisms and genes involved at the effector phase of arthritis. Arthritis Res Ther 2007; 8:223. [PMID: 17254316 PMCID: PMC1794524 DOI: 10.1186/ar2089] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
During the development of rheumatoid arthritis (RA) autoantibodies to IgG-Fc, citrullinated proteins, collagen type II (CII), glucose 6 phosphoisomerase (G6PI) and some other self-antigens appear. Of these, a pathogenic effect of the anti-CII and anti-G6PI antibodies is well demonstrated using animal models. These new antibody mediated arthritis models have proven to be very useful for studies involved in understanding the molecular pathways of the induction of arthritis in joints. Both the complement and FcgammaR systems have been found to play essential roles. Neutrophils and macrophages are important inflammatory cells and the secretion of tumour necrosis factor-alpha and IL-1beta is pathogenic. The identification of the genetic polymorphisms predisposing to arthritis is important for understanding the complexity of arthritis. Disease mechanisms and gene regions studied using the two antibody-induced arthritis mouse models (collagen antibody-induced arthritis and serum transfer-induced arthritis) are compared and discussed for their relevance in RA pathogenesis.
Collapse
MESH Headings
- Animals
- Antibodies/pharmacology
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/metabolism
- Antibody Specificity
- Arthritis, Experimental
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/physiopathology
- Autoantibodies/immunology
- Autoantibodies/metabolism
- B-Lymphocytes/immunology
- Disease Models, Animal
- Genetic Markers/genetics
- Genetic Predisposition to Disease
- Glucose-6-Phosphate Isomerase/genetics
- Glucose-6-Phosphate Isomerase/metabolism
- Mice
- Mice, Inbred DBA
- Mice, Transgenic
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Rheumatoid Factor/metabolism
- Sensitivity and Specificity
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
| | - Rikard Holmdahl
- Medical Inflammation Research, Lund University, Lund 22184, Sweden
| |
Collapse
|
62
|
Negi VS, Elluru S, Sibéril S, Graff-Dubois S, Mouthon L, Kazatchkine MD, Lacroix-Desmazes S, Bayry J, Kaveri SV. Intravenous immunoglobulin: an update on the clinical use and mechanisms of action. J Clin Immunol 2007; 27:233-45. [PMID: 17351760 DOI: 10.1007/s10875-007-9088-9] [Citation(s) in RCA: 182] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2007] [Accepted: 02/21/2007] [Indexed: 01/27/2023]
Abstract
Initially used as a replacement therapy for immunodeficiency diseases, intravenous immunoglobulin (IVIg) is now widely used for a number of autoimmune and inflammatory diseases. Considerable progress has been made in understanding the mechanisms by which IVIg exerts immunomodulatory effects in autoimmune and inflammatory disorders. The mechanisms of action of IVIg are complex, involving modulation of expression and function of Fc receptors, interference with activation of complement and the cytokine network and of idiotype network, regulation of cell growth, and effects on the activation, differentiation, and effector functions of dendritic cells, and T and B cells.
Collapse
Affiliation(s)
- Vir-Singh Negi
- Department of Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry, India
| | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Lecka-Czernik B, Ackert-Bicknell C, Adamo ML, Marmolejos V, Churchill GA, Shockley KR, Reid IR, Grey A, Rosen CJ. Activation of peroxisome proliferator-activated receptor gamma (PPARgamma) by rosiglitazone suppresses components of the insulin-like growth factor regulatory system in vitro and in vivo. Endocrinology 2007; 148:903-11. [PMID: 17122083 PMCID: PMC1851001 DOI: 10.1210/en.2006-1121] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Rosiglitazone (Rosi) belongs to the class of thiazolidinediones (TZDs) that are ligands for peroxisome proliferator-activated receptor gamma (PPARgamma). Stimulation of PPARgamma suppresses bone formation and enhances marrow adipogenesis. We hypothesized that activation of PPARgamma down-regulates components of the IGF regulatory system, leading to impaired osteoblast function. Rosi treatment (1 microm) of a marrow stromal cell line (UAMS-33) transfected with empty vector (U-33/c) or with PPARgamma2 (U-33/gamma2) were analyzed by microarray. Rosi reduced IGF-I, IGF-II, IGFBP-4, and the type I and II IGF receptor (IGF1R and IGF2R) expression at 72 h in U-33/gamma2 compared with U-33/c cells (P < 0.01); these findings were confirmed by RT-PCR. Rosi reduced secreted IGF-I from U-33/gamma2 cells by 75% (P < 0.05). Primary marrow stromal cells (MSCs) extracted from adult (8 months) and old (24 months) C57BL/6J (B6) mice were treated with Rosi (1 microm) for 48 h. IGF-I, IGFBP-4, and IGF1R transcripts were reduced in Rosi-treated MSCs compared with vehicle (P < 0.01) and secreted IGF-I was also suppressed (P < 0.05). B6 mice treated with Rosi (20 mg/kg.d) for short duration (i.e. 4 d), and long term (i.e. 7 wk) had reduced serum IGF-I; this was accompanied by markedly suppressed IGF-I transcripts in the liver and peripheral fat of treated animals. To determine whether Rosi affected circulating IGF-I in humans, we measured serum IGF-I, IGFBP-2, and IGFBP-3 at four time points in 50 postmenopausal women randomized to either Rosi (8 mg/d) or placebo. Rosi-treated subjects had significantly lower IGF-I at 8 wk than baseline (-25%, P < 0.05), and at 16 wk their levels were reduced 14% vs. placebo (P = 0.15). We conclude that Rosi suppresses IGF-I expression in bone and liver; these changes could affect skeletal acquisition through endocrine and paracrine pathways.
Collapse
Affiliation(s)
- B Lecka-Czernik
- St. Joseph Hospital, Maine Center for Osteoporosis Research and Education, 360 Broadway, Bangor, Maine 04401, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Nimmerjahn F, Ravetch JV. The antiinflammatory activity of IgG: the intravenous IgG paradox. ACTA ACUST UNITED AC 2007; 204:11-5. [PMID: 17227911 PMCID: PMC2118416 DOI: 10.1084/jem.20061788] [Citation(s) in RCA: 203] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
How high doses of intravenous IgG (IVIG) suppress autoimmune diseases remains unresolved. We have recently shown that the antiinflammatory activity of IVIG can be attributed to a minor species of IgGs that is modified with terminal sialic acids on their Fc-linked glycans. Here we propose that these Fc-sialylated IgGs engage a unique receptor on macrophages that, in turn, leads to the upregulation of an inhibitory Fcgamma receptor (FcgammaR), thereby protecting against autoantibody-mediated pathology.
Collapse
Affiliation(s)
- Falk Nimmerjahn
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY 10021, USA
| | | |
Collapse
|
65
|
Yu JJ, Ruddy MJ, Wong GC, Sfintescu C, Baker PJ, Smith JB, Evans RT, Gaffen SL. An essential role for IL-17 in preventing pathogen-initiated bone destruction: recruitment of neutrophils to inflamed bone requires IL-17 receptor-dependent signals. Blood 2007; 109:3794-802. [PMID: 17202320 PMCID: PMC1874584 DOI: 10.1182/blood-2005-09-010116] [Citation(s) in RCA: 270] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
IL-17 and its receptor are founding members of a novel family of inflammatory cytokines. IL-17 plays a pathogenic role in rheumatoid arthritis (RA)-associated bone destruction. However, IL-17 is also an important regulator of host defense through granulopoiesis and neutrophil trafficking. Therefore, the role of IL-17 in pathogen-initiated bone loss was not obvious. The most common form of infection-induced bone destruction occurs in periodontal disease (PD). In addition to causing significant morbidity, PD is a risk factor for atherosclerotic heart disease and chronic obstructive pulmonary disease (COPD). Similar to RA, bone destruction in PD is caused by the immune response. However, neutrophils provide critical antimicrobial defense against periodontal organisms. Since IL-17 is bone destructive in RA but a key regulator of neutrophils, we examined its role in inflammatory bone loss induced by the oral pathogen Porphyromonas gingivalis in IL-17RA-deficient mice. These mice showed enhanced periodontal bone destruction, suggesting a bone-protective role for IL-17, reminiscent of a neutrophil deficiency. Although IL-17RA-deficient neutrophils functioned normally ex vivo, IL-17RA knock-out (IL-17RA(KO)) mice exhibited reduced serum chemokine levels and concomitantly reduced neutrophil migration to bone. Consistently, CXCR2(KO) mice were highly susceptible to alveolar bone loss; interestingly, these mice also suggested a role for chemokines in maintaining normal bone homeostasis. These results indicate a nonredundant role for IL-17 in mediating host defense via neutrophil mobilization.
Collapse
Affiliation(s)
- Jeffrey J Yu
- Department of Microbiology and Immunology, School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | | | | | | | | | | | | | | |
Collapse
|
66
|
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: 178] [Impact Index Per Article: 9.4] [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
|
67
|
Scatizzi JC, Hutcheson J, Bickel E, Woods JM, Klosowska K, Moore TL, Haines GK, Perlman H. p21Cip1 is required for the development of monocytes and their response to serum transfer-induced arthritis. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 168:1531-41. [PMID: 16651620 PMCID: PMC1606593 DOI: 10.2353/ajpath.2006.050555] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
One of the central functions of cyclin-dependent kinase inhibitors, such as p21, p27, or p16, is to prevent entry into the cell cycle. However, the question remains as to whether they have other functions in the cell. We previously demonstrated that overexpression of p21 in fibroblasts isolated from patients with rheumatoid arthritis decreases the production of pro-inflammatory molecules. Overexpression of p21 has been also shown to reduce the development of experimental arthritis in mice and rats. To explore the role of endogenous p21 in the development of arthritis, we induced arthritis in p21(-/-) mice using the K/BxN serum transfer model of arthritis. Mice deficient in p21 were more resistant to serum transfer-induced arthritis (K/BxN) than wild-type (wt) control mice. Fewer macrophages were detected in p21(-/-) as compared to wt joints following transfer of K/BxN serum. Chemotaxis assays of bone marrow-derived macrophages from p21(-/-) and wt mice revealed no difference in migration. However, there was a substantial decrease in inflammatory monocytes circulating in peripheral blood and in monocyte precursors in bone marrow of p21(-/-) mice as compared to wt mice. Adoptive transfer of wt bone marrow-derived macrophages into p21(-/-) mice restored the sensitivity to serum transfer-induced arthritis. These data suggest a novel role for p21 in regulating the development and/or differentiation of monocytic populations that are crucial for the induction of inflammatory arthritis.
Collapse
Affiliation(s)
- John C Scatizzi
- Department of Molecular Microbiology and Immunology, Division of Rheumatology, School of Medicine, St. Louis University, St. Louis, Missouri 63104, USA.
| | | | | | | | | | | | | | | |
Collapse
|
68
|
Petkova SB, Konstantinov KN, Sproule TJ, Lyons BL, Awwami MA, Roopenian DC. Human antibodies induce arthritis in mice deficient in the low-affinity inhibitory IgG receptor Fc gamma RIIB. ACTA ACUST UNITED AC 2006; 203:275-80. [PMID: 16476768 PMCID: PMC2118221 DOI: 10.1084/jem.20051951] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Rheumatoid arthritis (RA) is a complex autoimmune disease with a poorly understood pathogenesis. The disease is associated with polyclonal B cell activation and the production of autoantibodies (autoAbs), but there is a longstanding controversy as to whether such Abs contribute to, or are secondary to, the pathogenesis of RA. To address the potential pathogenicity of human RA–associated Abs, we developed a passive transfer model involving mice deficient in the low-affinity inhibitory Fc receptor, FcγRIIB. We report that plasma or serum from patients with active RA can induce inflammation and histological lesions in FcγRIIB−/− mice consistent with arthritis, and that this pathogenic activity is caused by the immunoglobulin G–rich fraction. Our results suggest that humoral autoimmunity can contribute directly to autoimmune arthritis, and that FcγRIIB−/− mice are a promising model to evaluate the arthritogenic potential of human autoAbs.
Collapse
MESH Headings
- Animals
- Arthritis, Experimental/genetics
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/pathology
- Autoantibodies/toxicity
- Binding Sites, Antibody/genetics
- Female
- Humans
- Injections, Intraperitoneal
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, IgG/deficiency
- Receptors, IgG/genetics
Collapse
|
69
|
Nguyen DH, Vadlamudi V, Toshida H, Beuerman RW. Loss of parasympathetic innervation leads to sustained expression of pro-inflammatory genes in the rat lacrimal gland. Auton Neurosci 2006; 124:81-9. [PMID: 16458615 PMCID: PMC2709271 DOI: 10.1016/j.autneu.2005.12.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2005] [Revised: 11/15/2005] [Accepted: 12/02/2005] [Indexed: 11/19/2022]
Abstract
It has been shown that removal of parasympathetic innervation to the lacrimal gland (LG) leads to rapid reduction in tear flow. Additionally, removal of the neural input resulted in disorganization of LG structure and changes in the expression of genes associated with the secretory pathway and inflammation. The goal of this study was to investigate the change in pro-inflammatory and pro-apoptotic gene expression in the rat LG following parasympathetic denervation. Male Long-Evans rats underwent unilateral sectioning of the greater superficial petrosal nerve and were sacrificed 7 days or 2.5 months later. cDNA was synthesized from LG RNA from the contralateral control (Ctla) and parasympathectomized (Px) glands and comparative real-time PCR was performed. Mean threshold cycles (MC(T)) for the Ctla and Px LG genes were normalized to 18S rRNA MC(T) values, and the relative fold change was calculated for each gene using the 2(-DeltaDeltaC)(T) method. The expression of nuclear factor kappa B1, caspase 1, eotaxin, leukocyte antigen MRC-OX44, allograft inflammatory factor-1, MHC class II molecules RT.1B and RT.1D, IgG receptor FcRn, and macrophage metalloelastase was increased and remained elevated in the Px LG, compared with the Ctla LG. Increased expression of the initiator of apoptosis gene, caspase 2, was confirmed, but expression of the executor gene, caspase 6, was not elevated in the Px LG. Reduced expression of genes associated with post-translational protein processing-furin convertase, protein disulfide isomerase, and UDP-gal transporter isozyme 1-was noted in the Px LG. No significant changes in the expression of genes associated with lysosomal and non-lysosomal-mediated protein degradation were found. Removal of parasympathetic input may lead to decreased capacity for protein synthesis and elevated immune responses in the Px LG. These changes occur without increases in expression of the muscarinic acetylcholine receptor subtype 3, and may suggest the early changes in LG acinar cells and the pathophysiology of autoimmune responses.
Collapse
Affiliation(s)
- Doan H Nguyen
- LSU Eye Center/Lions Eye Research Laboratories, Department of Ophthalmology, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite B, New Orleans, LA 70112, USA.
| | | | | | | |
Collapse
|
70
|
Crowley H, Alroy J, Sproule TJ, Roopenian D, Huber BT. The MHC class I-related FcRn ameliorates murine Lyme arthritis. Int Immunol 2006; 18:409-14. [PMID: 16415101 DOI: 10.1093/intimm/dxh380] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The identification of the neonatal FcR (FcRn) as an IgG homeostasis regulator has led to research aimed at delineating a role for FcRn in humorally mediated disease. FcRn is a class I-related molecule that prolongs the half-life of serum IgG by preferentially binding IgG at low pH and inhibiting its degradation. Its role in protective immunity to infectious organisms is unknown. We investigated the function of FcRn in the murine model of Lyme arthritis, caused by infection with Borrelia burgdorferi. We infected FcRn(-/-) and wild-type mice with B. burgdorferi and monitored the development of arthritis. Infected FcRn(-/-) mice demonstrated decreased serum levels of anti-B. burgdorferi antibodies and borreliacidal activity. Moreover, these mutant mice developed increased ankle swelling and joint histopathology following infection. Our data suggest that FcRn ameliorates murine Lyme arthritis by preventing the degradation of protective borreliacidal antibodies.
Collapse
Affiliation(s)
- Helena Crowley
- Department of Pathology, Tufts University School of Medicine, Boston, MA 02111, USA
| | | | | | | | | |
Collapse
|
71
|
Li N, Zhao M, Hilario-Vargas J, Prisayanh P, Warren S, Diaz LA, Roopenian DC, Liu Z. Complete FcRn dependence for intravenous Ig therapy in autoimmune skin blistering diseases. J Clin Invest 2005; 115:3440-50. [PMID: 16284651 PMCID: PMC1280965 DOI: 10.1172/jci24394] [Citation(s) in RCA: 194] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2005] [Accepted: 09/13/2005] [Indexed: 01/16/2023] Open
Abstract
Numerous mechanisms of action have been proposed for intravenous Ig (IVIG). In this study, we used IgG passive transfer murine models of bullous pemphigoid (BP), pemphigus foliaceus (PF), and pemphigus vulgaris (PV) to test the hypothesis that the effect of IVIG in autoantibody-mediated cutaneous bullous diseases is to accelerate the degradation of pathogenic IgG by saturation of the MHC-like Fc receptor neonatal Fc receptor (FcRn). BP, PF, and PV are organ-specific antibody-mediated diseases in which autoantibodies target the hemidesmosomal antigen BP180 and desmosomal antigens Dsg1 and Dsg3, respectively. Antibodies against BP180, Dsg1, and Dsg3, when injected into neonatal mice, induce the BP, PF, and PV disease phenotypes, respectively. We found that FcRn-deficient mice were resistant to experimental BP, PF, and PV. Circulating levels of pathogenic IgG in FcRn-deficient mice were significantly reduced compared with those in WT mice. Administration of high-dose human IgG (HDIG) to WT mice also drastically reduced circulating pathogenic IgG levels and prevented blistering. In FcRn-deficient mice, no additional protective effect with HDIG was realized. These data demonstrate that the therapeutic efficacy of HDIG treatment in the pemphigus and pemphigoid models is dependent on FcRn. Thus, FcRn is a promising therapeutic target for treating such IgG-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Ning Li
- Department of Dermatology, University of North Carolina at Chapel Hill, North Carolina 27599, USA
| | | | | | | | | | | | | | | |
Collapse
|
72
|
Ni H, Chen P, Spring CM, Sayeh E, Semple JW, Lazarus AH, Hynes RO, Freedman J. A novel murine model of fetal and neonatal alloimmune thrombocytopenia: response to intravenous IgG therapy. Blood 2005; 107:2976-83. [PMID: 16317099 PMCID: PMC1895387 DOI: 10.1182/blood-2005-06-2562] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Fetal and neonatal alloimmune thrombo cytopenia (FNAITP) is a life-threatening bleeding disorder caused by maternal antibodies directed against fetal platelet antigens. The immunoreactive epitopes in FNAITP are primarily located in the extracellular regions of the platelet glycoprotein IIIa (beta3 integrin). Here we have established a novel animal model of FNAITP using beta3 integrin-deficient (beta3-/-) mice. We demonstrated first that these mice are immunoresponsive to beta3 integrin; beta3-/- mice transfused with wild-type platelets generated specific anti-beta3 antibodies which were able to induce thrombocytopenia in wild-type mice. Subsequently, beta3-/- female mice (both naive and immunized) were bred with wild-type male mice to recapitulate the features of FNAITP. The titer of generated maternal antibodies correlated with the severity of FNAITP. High titer maternal anti-beta3 anti-bodies caused severe fetal thrombocytopenia, intracranial hemorrhage, and even miscarriage. Furthermore, maternal administration of intravenous immunoglobulin G (IgG) ameliorated FNAITP and down-regulated pathogenic antibodies in both the maternal and fetal circulations.
Collapse
Affiliation(s)
- Heyu Ni
- Canadian Blood Services and Dept of Laboratory Medicine and Pathobiology, St Michael's Hospital, University of Toronto, 30 Bond St Rm 2-006, Bond Wing, Toronto, ON, Canada M5B 1W8.
| | | | | | | | | | | | | | | |
Collapse
|
73
|
Abstract
The activation threshold of cells in the immune system is often tuned by cell surface molecules. The Fc receptors expressed on various hematopoietic cells constitute critical elements for activating or downmodulating immune responses and combines humoral and cell-mediated immunity. Thus, Fc receptors are the intelligent sensors of the immune status in the individual. However, impaired regulation by Fc receptors will lead to unresponsiveness or hyperreactivity to foreign as well as self-antigens. Murine models for autoimmune disease indicate the indispensable roles of the inhibitory Fc receptor in the suppression of such disorders, whereas activating-type FcRs are crucial for the onset and exacerbation of the disease. The development of many autoimmune diseases in humans may be caused by impairment of the human Fc receptor regulatory system. This review is aimed at providing a current overview of the mechanism of Fc receptor-based immune regulation and the possible scenario of how autoimmune disease might result from their dysfunction.
Collapse
Affiliation(s)
- Toshiyuki Takai
- Department of Experimental Immunology and CREST Program of Japan Science and Technology Agency, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan.
| |
Collapse
|
74
|
Solomon S, Kassahn D, Illges H. The role of the complement and the Fc gamma R system in the pathogenesis of arthritis. Arthritis Res Ther 2005; 7:129-35. [PMID: 15987494 PMCID: PMC1175042 DOI: 10.1186/ar1761] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Autoantibodies in sera from patients with autoimmune diseases have long been known and have become diagnostic tools. Analysis of their functional role again became popular with the availability of mice mutant for several genes of the complement and Fcγ receptor (FcγR) systems. Evidence from different inflammatory models suggests that both systems are interconnected in a hierarchical way. The complement system mediators such as complement component 5a (C5a) might be crucial in the communication between the complement system and FcγR-expressing cells. The split complement protein C5a is known to inactivate cells by its G-protein-coupled receptor and to be involved in the transcriptional regulation of FcγRs, thereby contributing to the complex regulation of autoimmune disease.
Collapse
Affiliation(s)
- Samuel Solomon
- Immunology, Department of Biology, Faculty of Sciences, University of Konstanz, Konstanz, Germany
| | - Daniela Kassahn
- Immunology, Department of Biology, Faculty of Sciences, University of Konstanz, Konstanz, Germany
| | - Harald Illges
- Immunology, Department of Biology, Faculty of Sciences, University of Konstanz, Konstanz, Germany
- Biotechnology Institute Thurgau, Tägerwilen, Switzerland
- University of Applied Sciences, Department of Natural Sciences, Immunology and Cell Biology, Rheinbach, Germany
| |
Collapse
|
75
|
Abstract
For several decades, intravenous Ig has been used as treatment for a variety of immune-related diseases, including immune thrombocytopenic purpura (ITP), autoimmune neuropathies, systemic lupus erythematosus, myasthenia gravis, Guillain-Barré syndrome, skin blistering syndromes, and Kawasaki disease. Despite years of use, its mechanism of immunomodulation is still unclear. Recent studies using mouse models of ITP and arthritis, including one reported in this issue of the JCI, now provide some insights into this mechanism and the rationale for the development of Fcgamma receptor-targeted therapeutics.
Collapse
Affiliation(s)
- Raphael Clynes
- Department of Medicine and Microbiology, Columbia University, New York, New York 10032, USA.
| |
Collapse
|
76
|
Siragam V, Brinc D, Crow AR, Song S, Freedman J, Lazarus AH. Can antibodies with specificity for soluble antigens mimic the therapeutic effects of intravenous IgG in the treatment of autoimmune disease? J Clin Invest 2005; 115:155-60. [PMID: 15630455 PMCID: PMC539200 DOI: 10.1172/jci22753] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2004] [Accepted: 11/02/2004] [Indexed: 12/21/2022] Open
Abstract
Intravenous Ig (IVIg) mediates protection from the effects of immune thrombocytopenic purpura (ITP) as well as numerous other autoimmune states; however, the active antibodies within IVIg are unknown. There is some evidence that antibodies specific for a cell-associated antigen on erythrocytes are responsible, at least in part, for the therapeutic effect of IVIg in ITP. Yet whether an IVIg directed to a soluble antigen can likewise be beneficial in ITP or other autoimmune diseases is also unknown. A murine model of ITP was used to determine the effectiveness of IgG specific to soluble antigens in treating immune thrombocytopenic purpura. Mice experimentally treated with soluble OVA + anti-OVA versus mice treated with OVA conjugated to rbcs (OVA-rbcs) + anti-OVA were compared. In both situations, mice were protected from ITP. Both these experimental therapeutic regimes acted in a complement-independent fashion and both also blocked reticuloendothelial function. In contrast to OVA-rbcs + anti-OVA, soluble OVA + anti-OVA (as well as IVIg) did not have any effect on thrombocytopenia in mice lacking the inhibitory receptor FcgammaRIIB (FcgammaRIIB(-/-) mice). Similarly, antibodies reactive with the endogenous soluble antigens albumin and transferrin also ameliorated ITP in an FcgammaRIIB-dependent manner. Finally, broadening the significance of these experiments was the finding that anti-albumin was protective in a K/BxN serum-induced arthritis model. We conclude that IgG antibodies directed to soluble antigens ameliorated 2 disparate IVIg-treatable autoimmune diseases.
Collapse
MESH Headings
- Animals
- Antibodies/immunology
- Antibody Specificity
- Antigens/chemistry
- Antigens/immunology
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Arthritis/immunology
- Immunoglobulin G/administration & dosage
- Immunoglobulin G/immunology
- Immunoglobulin G/therapeutic use
- Immunoglobulins, Intravenous/immunology
- Immunoglobulins, Intravenous/therapeutic use
- Immunotherapy
- Inflammation/immunology
- Mice
- Mice, Knockout
- Ovalbumin/pharmacology
- Purpura, Thrombocytopenic, Idiopathic/chemically induced
- Purpura, Thrombocytopenic, Idiopathic/immunology
- Purpura, Thrombocytopenic, Idiopathic/therapy
- Receptors, IgG/deficiency
- Receptors, IgG/genetics
- Receptors, IgG/immunology
- Receptors, IgG/metabolism
- Solubility
Collapse
Affiliation(s)
- Vinayakumar Siragam
- Transfusion Medicine Research and the Department of Laboratory Medicine and Pathobiology, St. Michael's Hospital, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|