701
|
Kielczewska A, Vidal SM. Enemy at the gates: forward genetics of the mouse antiviral response. Curr Opin Immunol 2006; 18:617-26. [PMID: 16879955 DOI: 10.1016/j.coi.2006.07.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2006] [Accepted: 07/20/2006] [Indexed: 01/20/2023]
Abstract
The environment and the genetic constitution of both the pathogen and the host influence the severity and the outcome of viral infections. Whereas identification of the host component in humans remains challenging, recent progress in defining genes through analysis of mouse models of infection presenting natural or chemically induced variation in host susceptibility mark a fruitful period of gene discovery. This includes recognition that UNC93B1, which encodes an endocytic protein, is a susceptibility gene, providing an unexpected entry point to our understanding of the response against herpesvirus infection. By contrast, elucidation of alternative mechanisms of host resistance against mouse cytomegalovirus in inbred mouse strains has led to new insights regarding molecular recognition of the infected cells by natural killer cell MHC class I receptors. In addition, the conservation of genetic and functional aspects between mouse and human is enabling a rational pursuit of potential cures. With the continuous development of resources for experimental investigation of the genome, the production of new mutant alleles, and the phenotypic characterization of new models of infection, we predict that mouse genetic models will make an increasing contribution to our understanding of the genetic puzzle of host response to virus infection.
Collapse
Affiliation(s)
- Agnieszka Kielczewska
- McGill Centre for the Study of Host Resistance, Department of Human Genetics, McGill University, Montreal, Quebec, H3A 2B4, Canada
| | | |
Collapse
|
702
|
Kagan JC, Medzhitov R. Phosphoinositide-mediated adaptor recruitment controls Toll-like receptor signaling. Cell 2006; 125:943-55. [PMID: 16751103 DOI: 10.1016/j.cell.2006.03.047] [Citation(s) in RCA: 679] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2005] [Revised: 12/08/2005] [Accepted: 03/21/2006] [Indexed: 10/24/2022]
Abstract
Toll-like receptors (TLRs) play a critical role in the immune system as sensors of microbial infection. Signaling downstream from TLRs is initiated by the recruitment of adaptor proteins, including MyD88 and TIRAP. These adaptors play essential roles in TLR signaling, but the mechanism of their function is currently unknown. Here we demonstrate that TIRAP and MyD88 have distinct functions and describe a mechanism of recruitment of TIRAP and MyD88 to TLR4. We find that TIRAP contains a phosphatidylinositol 4,5-bisphosphate (PIP2) binding domain, which mediates TIRAP recruitment to the plasma membrane. TIRAP then functions to facilitate MyD88 delivery to activated TLR4 to initiate signal transduction. These results establish that phosphoinositide-mediated adaptor recruitment initiates a specific signal-transduction pathway.
Collapse
Affiliation(s)
- Jonathan C Kagan
- Section of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | |
Collapse
|
703
|
Ruse M, Knaus UG. New players in TLR-mediated innate immunity: PI3K and small Rho GTPases. Immunol Res 2006; 34:33-48. [PMID: 16720897 DOI: 10.1385/ir:34:1:33] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 12/12/2022]
Abstract
Toll-like receptors (TLRs) play a crucial role in the innate immune system as a first line of defense against pathogens. TLR activation in phagocytes produces pro-inflammatory cytokines and chemokines that contribute directly to elimination of infectious agents and activation of adaptive immune responses. However, a sustained inflammatory response can result in tissue damage and generalized sepsis. This review summarizes the complex and sometimes conflicting links of TLR signaling with two important regulators of immune cells functions: phosphoinositide 3-kinases (PI3Ks) and small GTPases of the Rho family. A unified model of hierarchical organization of these signaling participants is still premature, given that the tools for delineating how control of TLRmediated pathways is achieved are just emerging. Critical progress in our understanding of spatial-temporal propagation of TLR signaling will certainly be provided in the near future by pharmacological targeting of PI3Ks using recently characterized, second-generation PI3K inhibitors in combination with gene-targeting strategies for PI3K subunits and Rho GTPases targeted to the murine myeloid compartment.
Collapse
Affiliation(s)
- Monica Ruse
- Department of Immunology, The Scripps Research Institute La Jolla, California.
| | | |
Collapse
|
704
|
Jiang Z, Georgel P, Li C, Choe J, Crozat K, Rutschmann S, Du X, Bigby T, Mudd S, Sovath S, Wilson IA, Olson A, Beutler B. Details of Toll-like receptor:adapter interaction revealed by germ-line mutagenesis. Proc Natl Acad Sci U S A 2006; 103:10961-6. [PMID: 16832055 PMCID: PMC1544157 DOI: 10.1073/pnas.0603804103] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The immunovariant N-ethyl-N-nitrosourea-induced mutations Pococurante (Poc) and Lackadaisical were found to alter MyD88, creating striking receptor-selective effects. Poc, in particular, prevented sensing of all MyD88-dependent Toll-like receptor (TLR) ligands except diacyl lipopeptides. Furthermore, Poc-site and classical BB loop mutations caused equivalent phenotypes when engrafted into any TLR/IL-1 receptor/resistance (TIR) domain. These observations, complemented by data from docking studies and site-directed mutagenesis, revealed that BB loops and Poc sites interact homotypically across the receptor:adapter signaling interface, whereas the C-terminal alpha(E)-helices support adapter:adapter and receptor:receptor oligomerization. We have thus defined the TIR domain surface that mediates association between TLRs and MyD88 and the surface required for MyD88 or TLR oligomerization. Moreover, MyD88 engages individual TLRs differently, suggesting the feasibility of selective pharmacologic TIR domain receptor blockade.
Collapse
Affiliation(s)
| | | | - Chenglong Li
- Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037; and
| | - Jungwoo Choe
- Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037; and
| | | | | | - Xin Du
- Departments of *Immunology and
| | - Tim Bigby
- Department of Medicine, Veterans Administration San Diego Health Care System, Mail Code 111J, 3350 La Jolla Village Drive, San Diego, CA 92161
| | | | | | - Ian A. Wilson
- Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037; and
| | - Arthur Olson
- Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037; and
| | - Bruce Beutler
- Departments of *Immunology and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
705
|
Hippenstiel S, Opitz B, Schmeck B, Suttorp N. Lung epithelium as a sentinel and effector system in pneumonia--molecular mechanisms of pathogen recognition and signal transduction. Respir Res 2006; 7:97. [PMID: 16827942 PMCID: PMC1533821 DOI: 10.1186/1465-9921-7-97] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2006] [Accepted: 07/08/2006] [Indexed: 12/22/2022] Open
Abstract
Pneumonia, a common disease caused by a great diversity of infectious agents is responsible for enormous morbidity and mortality worldwide. The bronchial and lung epithelium comprises a large surface between host and environment and is attacked as a primary target during lung infection. Besides acting as a mechanical barrier, recent evidence suggests that the lung epithelium functions as an important sentinel system against pathogens. Equipped with transmembranous and cytosolic pathogen-sensing pattern recognition receptors the epithelium detects invading pathogens. A complex signalling results in epithelial cell activation, which essentially participates in initiation and orchestration of the subsequent innate and adaptive immune response. In this review we summarize recent progress in research focussing on molecular mechanisms of pathogen detection, host cell signal transduction, and subsequent activation of lung epithelial cells by pathogens and their virulence factors and point to open questions. The analysis of lung epithelial function in the host response in pneumonia may pave the way to the development of innovative highly needed therapeutics in pneumonia in addition to antibiotics.
Collapse
Affiliation(s)
- Stefan Hippenstiel
- Department of Internal Medicine/Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Bastian Opitz
- Department of Internal Medicine/Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Bernd Schmeck
- Department of Internal Medicine/Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Norbert Suttorp
- Department of Internal Medicine/Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, 13353 Berlin, Germany
| |
Collapse
|
706
|
Johnsen IB, Nguyen TT, Ringdal M, Tryggestad AM, Bakke O, Lien E, Espevik T, Anthonsen MW. Toll-like receptor 3 associates with c-Src tyrosine kinase on endosomes to initiate antiviral signaling. EMBO J 2006; 25:3335-46. [PMID: 16858407 PMCID: PMC1523188 DOI: 10.1038/sj.emboj.7601222] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2004] [Accepted: 06/13/2006] [Indexed: 01/01/2023] Open
Abstract
Double-stranded RNA (dsRNA) is produced during the replication cycle of most viruses and triggers antiviral immune responses through Toll-like receptor 3 (TLR3). However, the molecular mechanisms and subcellular compartments associated with dsRNA-TLR3-mediated signaling are largely unknown. Here we show that c-Src tyrosine kinase is activated by dsRNA in human monocyte-derived dendritic cells, and is recruited to TLR3 in a dsRNA-dependent manner. DsRNA-induced activation of interferon-regulatory factor 3 and signal transducer and activator of transcription 1 was abolished in Src kinase-deficient cells, and restored by adding back c-Src, suggesting a central role of c-Src in antiviral immunity. We also provide evidence that TLR3 is localized in the endoplasmic reticulum of unstimulated cells, moves to dsRNA-containing endosomes in response to dsRNA, and colocalizes with c-Src on endosomes containing dsRNA in the lumen. These results provide novel insight into the molecular mechanisms of TLR3-mediated signaling, which may contribute to the understanding of innate immune responses during viral infections.
Collapse
Affiliation(s)
- Ingvild Bjellmo Johnsen
- Department of Laboratory Medicine, Children's and Women's Health, Institute of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology, Trondheim, Norway.
| | | | | | | | | | | | | | | |
Collapse
|
707
|
Xu Y, Kim SO, Li Y, Han J. Autophagy Contributes to Caspase-independent Macrophage Cell Death. J Biol Chem 2006; 281:19179-87. [PMID: 16702227 DOI: 10.1074/jbc.m513377200] [Citation(s) in RCA: 175] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Macrophage cell death plays a role in many physiological and pathophysiological conditions. Previous work has shown that macrophages can undergo caspase-independent cell death, and this process is associated with Nur77 induction, which is involved in inducing chromatin condensation and DNA fragmentation. Here we show that autophagy is a cytosolic event that controls caspase-independent macrophage cell death. Autophagy was induced in macrophages treated with lipopolysaccharides (LPSs) and the pan-caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp (Z-VAD), and the inhibition of autophagy by either chemical inhibitors or by the RNA interference knockdown of beclin (a protein required for autophagic body formation) inhibited caspase-independent macrophage cell death. We also found an increase in poly(ADP-ribose) (PAR) polymerase (PARP) activation and reactive oxygen species (ROS) production in LPS + Z-VAD-treated macrophages, and both are involved in caspase-independent macrophage cell death. We further determined that the formation of autophagic bodies in macrophages occurs downstream of PARP activation, and PARP activation occurs downstream of ROS production. Using macrophages in which receptor-interacting protein 1 (RIP1) was knocked down by small interfering RNA, and macrophages isolated from Toll/interleukin-1 receptor-domain-containing adaptor inducing IFN-beta (TRIF)-deficient mice, we found that TRIF and RIP1 function upstream of ROS production in LPS + Z-VAD-treated macrophages. We also found that Z-VAD inhibits LPS-induced RIP1 cleavage, which may contribute to ROS over-production in macrophages. This paper reveals that TRIF, RIP1, and ROS production, as well as PARP activation, are involved in inducing autophagy, which contributes to caspase-independent macrophage cell death.
Collapse
Affiliation(s)
- Yue Xu
- Department of Immunology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | |
Collapse
|
708
|
Datta SK, Okamoto S, Hayashi T, Shin SS, Mihajlov I, Fermin A, Guiney DG, Fierer J, Raz E. Vaccination with Irradiated Listeria Induces Protective T Cell Immunity. Immunity 2006; 25:143-52. [PMID: 16860763 DOI: 10.1016/j.immuni.2006.05.013] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2005] [Revised: 04/03/2006] [Accepted: 05/17/2006] [Indexed: 12/14/2022]
Abstract
We evaluated gamma-irradiated Listeria monocytogenes as a killed bacterial vaccine, testing the hypothesis that irradiation preserves antigenic and adjuvant structures destroyed by traditional heat or chemical inactivation. Irradiated Listeria monocytogenes (LM), unlike heat-killed LM, efficiently activated dendritic cells via Toll-like receptors and induced protective T cell responses in mice. Like live LM, irradiated LM induced Toll-like-receptor-independent T cell priming. Cross-presentation of irradiated listerial antigens to CD8(+) T cells involved TAP- and proteasome-dependent cytosolic antigen processing. These results establish that killed LM can induce protective T cell responses, previously thought to require live infection. gamma-irradiation may be potentially applied to numerous bacterial vaccine candidates, and irradiated bacteria could serve as a vaccine platform for recombinant antigens derived from other pathogens, allergens, or tumors.
Collapse
Affiliation(s)
- Sandip K Datta
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, 92093, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
709
|
Beutler B, Jiang Z, Georgel P, Crozat K, Croker B, Rutschmann S, Du X, Hoebe K. Genetic analysis of host resistance: Toll-like receptor signaling and immunity at large. Annu Rev Immunol 2006; 24:353-89. [PMID: 16551253 DOI: 10.1146/annurev.immunol.24.021605.090552] [Citation(s) in RCA: 611] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Classical genetic methods, driven by phenotype rather than hypotheses, generally permit the identification of all proteins that serve nonredundant functions in a defined biological process. Long before this goal is achieved, and sometimes at the very outset, genetics may cut to the heart of a biological puzzle. So it was in the field of mammalian innate immunity. The positional cloning of a spontaneous mutation that caused lipopolysaccharide resistance and susceptibility to Gram-negative infection led directly to the understanding that Toll-like receptors (TLRs) are essential sensors of microbial infection. Other mutations, induced by the random germ line mutagen ENU (N-ethyl-N-nitrosourea), have disclosed key molecules in the TLR signaling pathways and helped us to construct a reasonably sophisticated portrait of the afferent innate immune response. A still broader genetic screen--one that detects all mutations that compromise survival during infection--is permitting fresh insight into the number and types of proteins that mammals use to defend themselves against microbes.
Collapse
Affiliation(s)
- Bruce Beutler
- Department of Immunology, Scripps Research Institute, La Jolla, California 92037, USA.
| | | | | | | | | | | | | | | |
Collapse
|
710
|
Kumar H, Kawai T, Kato H, Sato S, Takahashi K, Coban C, Yamamoto M, Uematsu S, Ishii KJ, Takeuchi O, Akira S. Essential role of IPS-1 in innate immune responses against RNA viruses. ACTA ACUST UNITED AC 2006; 203:1795-803. [PMID: 16785313 PMCID: PMC2118350 DOI: 10.1084/jem.20060792] [Citation(s) in RCA: 399] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
IFN-beta promoter stimulator (IPS)-1 was recently identified as an adapter for retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (Mda5), which recognize distinct RNA viruses. Here we show the critical role of IPS-1 in antiviral responses in vivo. IPS-1-deficient mice showed severe defects in both RIG-I- and Mda5-mediated induction of type I interferon and inflammatory cytokines and were susceptible to RNA virus infection. RNA virus-induced interferon regulatory factor-3 and nuclear factor kappaB activation was also impaired in IPS-1-deficient cells. IPS-1, however, was not essential for the responses to either DNA virus or double-stranded B-DNA. Thus, IPS-1 is the sole adapter in both RIG-I and Mda5 signaling that mediates effective responses against a variety of RNA viruses.
Collapse
Affiliation(s)
- Himanshu Kumar
- Department of Host Defense, Japan Science and Technology Agency, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
711
|
Yang X, Murthy V, Schultz K, Tatro JB, Fitzgerald KA, Beasley D. Toll-like receptor 3 signaling evokes a proinflammatory and proliferative phenotype in human vascular smooth muscle cells. Am J Physiol Heart Circ Physiol 2006; 291:H2334-43. [PMID: 16782847 DOI: 10.1152/ajpheart.00252.2006] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Inflammation plays a key role in atherogenesis, perhaps promoted by bacterial and viral products present within the artery wall. Vascular smooth muscle cells (VSMC) can express certain bacterially responsive Toll-like receptors (TLR), which promote a proinflammatory and proliferative VSMC phenotype when activated, but it is unknown whether virally activated TLR can regulate VSMC phenotype. Here we tested the role in VSMC of TLR3, which is activated by double-stranded (dsRNA), a molecular signature of viruses. VSMC from multiple vessel types, including human coronary artery (HCoASMC) and mouse aorta (MAoSMC), expressed TLR3 constitutively, and HCoASMC were exquisitely sensitive to dsRNA-stimulated release of monocyte chemoattractant protein-1 (MCP-1) and interleukin-6. dsRNA-induced MCP-1 release was abolished by small interfering RNA-mediated TLR3 knockdown in HCoASMC and was absent in TLR3-/- MAoSMC but was unimpaired in TLR2-/- and in TLR4 signaling-deficient MAoSMC. Exposure to dsRNA also activated ERK1/2 and NF-kappaB in both human and murine SMC, but these effects were absent in SMC from TLR3-deficient mice, demonstrating a crucial role of TLR3 signaling. dsRNA also stimulated proliferation of HCoASMC, indicated by increased DNA synthesis, and induced persistent elevations in the intracellular levels of growth-promoting mediators, including interleukin-1alpha and phospho-ERK1/2. We conclude that exposure of HCoASMC to dsRNA elicits dramatic TLR3-mediated proinflammatory and proproliferative phenotypic changes, responses that could potentially be triggered by viral infection of cells within the arterial wall.
Collapse
Affiliation(s)
- Xin Yang
- Tufts-New England Medical Center, Box 8486, 750 Washington St., Boston, MA 02111, USA
| | | | | | | | | | | |
Collapse
|
712
|
Weighardt H, Mages J, Jusek G, Kaiser-Moore S, Lang R, Holzmann B. Organ-specific role of MyD88 for gene regulation during polymicrobial peritonitis. Infect Immun 2006; 74:3618-32. [PMID: 16714594 PMCID: PMC1479251 DOI: 10.1128/iai.01681-05] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Sepsis leads to the rapid induction of proinflammatory signaling cascades by activation of the innate immune system through Toll-like receptors (TLR). To characterize the role of TLR signaling through MyD88 for sepsis-induced transcriptional activation, we investigated gene expression during polymicrobial septic peritonitis by microarray analysis. Comparison of gene expression profiles for spleens and livers from septic wild-type and MyD88-deficient mice revealed striking organ-specific differences. Whereas MyD88 deficiency strongly reduced sepsis-induced gene expression in the liver, gene expression in the spleen was largely independent of MyD88, indicating organ-specific transcriptional regulation during polymicrobial sepsis. In addition to genes regulated by MyD88 in an organ-dependent manner, we also identified genes that exhibited an organ-independent influence of MyD88 and mostly encoded cytokines and chemokines. Notably, the expression of interferon (IFN)-regulated genes was markedly increased in septic MyD88-deficient mice compared to that in septic wild-type controls. Expression of IFN-regulated genes was dependent on the adapter protein TRIF. These results suggest that the influence of MyD88 on gene expression during sepsis strongly depends on the organ compartment affected by inflammation and that the lack of MyD88 may lead to disbalance of the expression of IFN-regulated genes.
Collapse
Affiliation(s)
- Heike Weighardt
- Department of Surgery, Technische Universität München, Ismaningerstrasse 22, 81675 Munich, Germany.
| | | | | | | | | | | |
Collapse
|
713
|
Nemazee D, Gavin A, Hoebe K, Beutler B. Immunology: Toll-like receptors and antibody responses. Nature 2006; 441:E4; discussion E4. [PMID: 16710369 PMCID: PMC3787862 DOI: 10.1038/nature04875] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Microbial components, such as lipopolysaccharides, augment immune responses by activating Toll-like receptors (TLRs). Some have interpreted this to mean that TLR signalling might not only help to initiate the adaptive immune response, but may also be required for it. The expanded view is shared by Pasare and Medzhitov, who conclude from an analysis of mice deficient in MyD88 (a TLR-signalling adaptor protein) that the generation of T-dependent antigen-specific antibody responses requires activation of TLRs in B cells. However, we show here that robust antibody responses can be elicited even in the absence of TLR signals. This appreciable TLR-independence of immune responses should be taken into account in the rational design of immunogenic and toleragenic vaccines.
Collapse
|
714
|
Meylan E, Tschopp J. Toll-like receptors and RNA helicases: two parallel ways to trigger antiviral responses. Mol Cell 2006; 22:561-9. [PMID: 16762830 DOI: 10.1016/j.molcel.2006.05.012] [Citation(s) in RCA: 302] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The early detection by the host of invading microorganisms, including viruses, depends on a limited number of specific receptors that recognize pathogen-associated molecular patterns (PAMPs). A few of these PAMPs, including ssRNA and dsRNA, are recognized by Toll-like receptors (TLR)-7/8 and TLR3, respectively. Activation of an antiviral TLR-dependent signaling cascade leads to the activation of the key transcription factors IRF and NF-kappaB, which promote antiviral responses through induction of specific genes. Recently, a second system has been described, which relies on the cytoplasmic recognition of dsRNA by RNA helicases such as RIG-I. In this review, we discuss the mechanistic aspects of these important arms of the host innate response to dsRNA and a few viral strategies utilized to counteract them.
Collapse
Affiliation(s)
- Etienne Meylan
- Department of Biochemistry, University of Lausanne, BIL Biomedical Research Center, Chemin des Boveresses 155, CH-1066 Epalinges, Switzerland
| | | |
Collapse
|
715
|
Abstract
Toll-like receptors (TLRs) play an important role in innate immunity. Individual TLRs recognise microbial components that are conserved among pathogens. Such recognition initiates necessary inflammatory immune responses and induces subsequent activation of adaptive immunity. Studies in people with polymorphisms in genes encoding TLR signalling can elucidate the relationship between TLRs and human diseases, such as infectious diseases, atherosclerosis and immunodeficiency. Indeed, accumulating data in respect to TLR signalling suggest that TLRs are closely related with the pathogenesis of autoimmune diseases. This review looks at the role of TLRs in various immune disorders, and discusses the pathogenesis of diseases.
Collapse
Affiliation(s)
- Satoshi Uematsu
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Suita Osaka 565-0851, Japan
| | | |
Collapse
|
716
|
Janssen E, Tabeta K, Barnes MJ, Rutschmann S, McBride S, Bahjat KS, Schoenberger SP, Theofilopoulos AN, Beutler B, Hoebe K. Efficient T Cell Activation via a Toll-Interleukin 1 Receptor-Independent Pathway. Immunity 2006; 24:787-799. [PMID: 16782034 DOI: 10.1016/j.immuni.2006.03.024] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2005] [Revised: 03/15/2006] [Accepted: 03/30/2006] [Indexed: 01/09/2023]
Abstract
Here, we describe a previously unrecognized pathway for activation of antigen-specific adaptive immune responses that was independent of Toll-Interleukin 1 Receptor signaling and directed toward detection of antigens expressed by apoptotic cells. This pathway is represented within Flt-3 Ligand-derived dendritic cells (DCs) that represent immature lymphoid DCs, but not within GM-CSF-treated bone marrow-derived dendritic cells. Exposure of these DCs to apoptotic cells resulted in production of type I interferon and favored the development of cytotoxic T cell responses. The N-Ethyl-N-Nitrosourea-induced germline mutation 3d (Unc3b1(3d/3d)) abolished both MHC class I and II responses elicited by this pathway, whereas a null allele of Cd36 selectively abolished class II responses. We propose that this mode of adaptive immune activation evolved to permit the sensitive detection of intracellular microbial infections, particularly viral infections, which frequently induce apoptotic cell death, but may also be important in transplantation, autoimmunity, and vaccine development.
Collapse
Affiliation(s)
- Edith Janssen
- Department of Developmental Immunology, La Jolla Institute for Allergy and Immunology, San Diego, California 92121
| | - Koichi Tabeta
- Department of Immunology, The Scripps Research Institute, La Jolla, California 92037
| | - Michael J Barnes
- Department of Immunology, The Scripps Research Institute, La Jolla, California 92037
| | - Sophie Rutschmann
- Department of Immunology, The Scripps Research Institute, La Jolla, California 92037
| | - Sara McBride
- Department of Developmental Immunology, La Jolla Institute for Allergy and Immunology, San Diego, California 92121
| | | | - Stephen P Schoenberger
- Laboratory of Cellular Immunology, La Jolla Institute for Allergy and Immunology, San Diego, California 92121
| | | | - Bruce Beutler
- Department of Immunology, The Scripps Research Institute, La Jolla, California 92037
| | - Kasper Hoebe
- Department of Immunology, The Scripps Research Institute, La Jolla, California 92037.
| |
Collapse
|
717
|
Szomolanyi-Tsuda E, Liang X, Welsh RM, Kurt-Jones EA, Finberg RW. Role for TLR2 in NK cell-mediated control of murine cytomegalovirus in vivo. J Virol 2006; 80:4286-91. [PMID: 16611887 PMCID: PMC1472014 DOI: 10.1128/jvi.80.9.4286-4291.2006] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Natural killer (NK) cells are essential for the early control of murine cytomegalovirus (MCMV) infection. Here, we demonstrate that toll-like receptor 2 (TLR2) plays a role in the NK cell-mediated control of MCMV. TLR2 knockout (KO) mice had elevated levels of MCMV in the spleen and liver on day 4 postinfection compared to C57BL/6 mice. In vivo depletion of NK cells with anti-NK1.1 antibodies, however, eliminated the differences in viral titers between the two groups, suggesting that the effect of TLR2 on MCMV clearance on day 4 was NK cell mediated. The defect in early antiviral control was associated with a decreased NK cell population in the spleen and liver and reduced amounts of interleukin-18 and alpha/beta interferon secreted in the TLR2 KO mice. Our studies suggest that in addition to the reported involvement of TLR9 and TLR3, TLR2 is also involved in innate immune responses to MCMV infection.
Collapse
Affiliation(s)
- Eva Szomolanyi-Tsuda
- Department of Pathology, University of Massachusetts Medical School, 55 Lake Ave. North, Worcester, Massachusetts 01655, USA.
| | | | | | | | | |
Collapse
|
718
|
Gitlin L, Barchet W, Gilfillan S, Cella M, Beutler B, Flavell RA, Diamond MS, Colonna M. Essential role of mda-5 in type I IFN responses to polyriboinosinic:polyribocytidylic acid and encephalomyocarditis picornavirus. Proc Natl Acad Sci U S A 2006; 103:8459-64. [PMID: 16714379 PMCID: PMC1464000 DOI: 10.1073/pnas.0603082103] [Citation(s) in RCA: 904] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The innate immune system recognizes viral dsRNA through two distinct pathways; the Toll-like receptor 3 (TLR3) pathway detects dsRNA phagocytosed in endosomes; the helicases retinoic acid-induced protein I (RIG-I) and melanoma differentiation-associated gene-5 (mda-5) detect cytoplasmic dsRNA generated during viral replication. Both RIG-I and mda-5 can bind polyriboinosinic:polyribocytidylic acid (polyI:C), the synthetic analog of viral dsRNA, and mediate type I IFN responses to polyI:C and multiple RNA viruses in vitro. We generated mda-5-deficient mice and showed that mda-5 is the dominant receptor mediating type I IFN secretion in response to polyI:C in vitro and in vivo. Moreover, mda-5-/- mice exhibited a selectively impaired antiviral response to encephalomyocarditis picornavirus, indicating functional specialization of mda-5 in vivo.
Collapse
Affiliation(s)
- Leonid Gitlin
- *Department of Pathology and Immunology, Washington University School of Medicine, Campus Box 8118, 660 South Euclid Avenue, St. Louis, MO 63110
| | - Winfried Barchet
- *Department of Pathology and Immunology, Washington University School of Medicine, Campus Box 8118, 660 South Euclid Avenue, St. Louis, MO 63110
| | - Susan Gilfillan
- *Department of Pathology and Immunology, Washington University School of Medicine, Campus Box 8118, 660 South Euclid Avenue, St. Louis, MO 63110
| | - Marina Cella
- *Department of Pathology and Immunology, Washington University School of Medicine, Campus Box 8118, 660 South Euclid Avenue, St. Louis, MO 63110
| | - Bruce Beutler
- Department of Immunology, Scripps Research Institute, La Jolla, CA 92037; and
| | - Richard A. Flavell
- Section of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520
| | | | - Marco Colonna
- *Department of Pathology and Immunology, Washington University School of Medicine, Campus Box 8118, 660 South Euclid Avenue, St. Louis, MO 63110
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
719
|
Dillon S, Agrawal S, Banerjee K, Letterio J, Denning TL, Oswald-Richter K, Kasprowicz DJ, Kellar K, Pare J, van Dyke T, Ziegler S, Unutmaz D, Pulendran B. Yeast zymosan, a stimulus for TLR2 and dectin-1, induces regulatory antigen-presenting cells and immunological tolerance. J Clin Invest 2006; 116:916-28. [PMID: 16543948 PMCID: PMC1401484 DOI: 10.1172/jci27203] [Citation(s) in RCA: 418] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2005] [Accepted: 01/24/2006] [Indexed: 01/08/2023] Open
Abstract
Emerging evidence suggests critical roles for APCs in suppressing immune responses. Here, we show that zymosan, a stimulus for TLR2 and dectin-1, regulates cytokine secretion in DCs and macrophages to induce immunological tolerance. First, zymosan induces DCs to secrete abundant IL-10 but little IL-6 and IL-12(p70). Induction of IL-10 is dependent on TLR2- and dectin-1-mediated activation of ERK MAPK via a mechanism independent of the activation protein 1 (AP-1) transcription factor c-Fos. Such DCs stimulate antigen-specific CD4+ T cells poorly due to IL-10 and the lack of IL-6. Second, zymosan induces F4-80+ macrophages in the splenic red pulp to secrete TGF-beta. Consistent with these effects on APCs, injection of zymosan plus OVA into mice results in OVA-specific T cells that secrete little or no Th1 or Th2 cytokines, but secrete robust levels of IL-10, and are unresponsive to challenge with OVA plus adjuvant. Finally, coinjection of zymosan with OVA plus LPS suppresses the response to OVA via a mechanism dependent on IL-10, TGF-beta, and lack of IL-6. Together, our data demonstrate that zymosan stimulates IL-10+ IL-12(p70)- IL-6low regulatory DCs and TGF-beta+ macrophages to induce immunological tolerance. These data suggest several targets for pharmacological modulation of immune responses in various clinical settings.
Collapse
Affiliation(s)
- Stephanie Dillon
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Sudhanshu Agrawal
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Kaustuv Banerjee
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - John Letterio
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Timothy L. Denning
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Kyra Oswald-Richter
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Deborah J. Kasprowicz
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Kathryn Kellar
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Jeff Pare
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Thomas van Dyke
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | | | - Derya Unutmaz
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Bali Pulendran
- Emory Vaccine Research Center and Department of Pathology, Emory University, Atlanta, Georgia, USA.
Immunoregulation Section, Lab of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology and Immunology, Vanderbilt University Medical School, Nashville, Tennessee, USA.
Immunology Program, Benaroya Research Institute, Seattle, Washington, USA.
Scientific Resources Program, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.
Department of Periodontology and Oral Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
720
|
Chiang E, Dang O, Anderson K, Matsuzawa A, Ichijo H, David M. Cutting edge: apoptosis-regulating signal kinase 1 is required for reactive oxygen species-mediated activation of IFN regulatory factor 3 by lipopolysaccharide. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2006; 176:5720-4. [PMID: 16670275 PMCID: PMC2749679 DOI: 10.4049/jimmunol.176.10.5720] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IFN regulatory factor (IRF) 3 participates in the transcriptional induction of IFN-alpha, IFN-beta, and a subset of IFN-stimulated genes (ISGs) as a result of viral infection. In addition, bacterial cell wall components such as LPS activate IRF3 in a p38-dependent manner. In this study we show that IRF3-mediated ISG induction by LPS requires the production of reactive oxygen species (ROS) by the NADPH-dependent oxidase NOX4. Furthermore, we present evidence that LPS-mediated ROS production leads to activation of apoptosis-regulating-signal kinase (ASK) 1, a MAPK kinase kinase family member capable of activating the MAP kinase 6/p38 axis. ASK1 kinase activity proved essential for IRF3-mediated ISG induction by LPS. Thus, our results presented here suggest a novel role for ROS and ASK1 in the innate immune response as signaling intermediates in the IRF3 activation pathway.
Collapse
Affiliation(s)
- Edward Chiang
- Division of Biological Sciences and University of California San Diego Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Oanh Dang
- Division of Biological Sciences and University of California San Diego Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Keith Anderson
- Division of Biological Sciences and University of California San Diego Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Atsushi Matsuzawa
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Hidenori Ichijo
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Michael David
- Division of Biological Sciences and University of California San Diego Cancer Center, University of California San Diego, La Jolla, CA 92093
| |
Collapse
|
721
|
Harari OA, Alcaide P, Ahl D, Luscinskas FW, Liao JK. Absence of TRAM restricts Toll-like receptor 4 signaling in vascular endothelial cells to the MyD88 pathway. Circ Res 2006; 98:1134-40. [PMID: 16574902 PMCID: PMC2701732 DOI: 10.1161/01.res.0000220105.85182.28] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Mammalian cells respond to bacterial lipopolysaccharide (LPS) through a cognate receptor: Toll-like receptor 4 (TLR4). The signaling pathways, which link TLR4 to the proinflammatory transcription factor nuclear factor kappaB (NF-kappaB), occur through the intracellular docking proteins MyD88 and Trif. We hypothesize that unlike antigen-presenting cells, vascular endothelial cells (ECs) lack the Trif protein TRAM and are therefore incapable of eliciting Trif-dependent immune responses to LPS. Stimulation of wild-type mice with LPS leads to the activation of NF-kappaB in ECs and macrophages in vitro and in vivo. In contrast to macrophages, LPS did not activate endothelial NF-kappaB or NF-kappaB-dependent genes in MyD88(-/-) mice, suggesting the absence of a functional Trif pathway in vascular ECs. Indeed, the Trif-dependent gene cxcl10 was not expressed in ECs after LPS stimulation. This correlated with diminished expression of the Trif accessory TIR protein TRAM in ECs. Overexpression of TRAM cDNA in ECs reconstituted LPS-induced Trif-dependent NF-kappaB activation and cxcl10 promoter activity. The functional absence of TRAM in vascular ECs restricts TLR4 signaling to MyD88-dependent pathway. This is in contrast to macrophages, which respond to LPS via both Trif- and MyD88-dependent pathways. These findings indicate that vascular ECs do not express the Trif-dependent gene subset. This implies that these genes may be dispensable for the endothelial response to bacterial infection and play no role in the endothelial contribution to the development of atherosclerosis.
Collapse
Affiliation(s)
- Olivier A Harari
- Vascular Medicine Research, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | |
Collapse
|
722
|
Ramsey S, Ozinsky A, Clark A, Smith K, de Atauri P, Thorsson V, Orrell D, Bolouri H. Transcriptional noise and cellular heterogeneity in mammalian macrophages. Philos Trans R Soc Lond B Biol Sci 2006; 361:495-506. [PMID: 16524838 PMCID: PMC1609340 DOI: 10.1098/rstb.2005.1808] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Transcriptional noise is known to play a crucial role in heterogeneity in bacteria and yeast. Mammalian macrophages are known to exhibit cell-to-cell variation in their responses to pathogens, but the source of this heterogeneity is not known. We have developed a detailed stochastic model of gene expression that takes into account scaling effects due to cell size and genome complexity. We report the results of applying this model to simulating gene expression variability in mammalian macrophages, demonstrating a possible molecular basis for heterogeneity in macrophage signalling responses. We note that the nature of predicted transcriptional noise in macrophages is different from that in yeast and bacteria. Some molecular interactions in yeast and bacteria are thought to have evolved to minimize the effects of the high-frequency noise observed in these species. Transcriptional noise in macrophages results in slow changes to gene expression levels and would not require the type of spike-filtering circuits observed in yeast and bacteria.
Collapse
Affiliation(s)
- S Ramsey
- Institute for Systems Biology1441 North 34th Street, Seattle, WA 98103-8904, USA
| | - A Ozinsky
- Institute for Systems Biology1441 North 34th Street, Seattle, WA 98103-8904, USA
| | - A Clark
- Institute for Systems Biology1441 North 34th Street, Seattle, WA 98103-8904, USA
| | - K.D Smith
- Institute for Systems Biology1441 North 34th Street, Seattle, WA 98103-8904, USA
- Department of Pathology, University of Washington1959 Pacific Street, Seattle, WA 98195, USA
| | - P de Atauri
- Institute for Systems Biology1441 North 34th Street, Seattle, WA 98103-8904, USA
| | - V Thorsson
- Institute for Systems Biology1441 North 34th Street, Seattle, WA 98103-8904, USA
| | - D Orrell
- Institute for Systems Biology1441 North 34th Street, Seattle, WA 98103-8904, USA
| | - H Bolouri
- Institute for Systems Biology1441 North 34th Street, Seattle, WA 98103-8904, USA
- Author for correspondence ()
| |
Collapse
|
723
|
Affiliation(s)
- Robert F Schwabe
- Department of Medicine, Columbia University, College of Physicians and Surgeons, New York, New York, USA.
| | | | | |
Collapse
|
724
|
Oda K, Kitano H. A comprehensive map of the toll-like receptor signaling network. Mol Syst Biol 2006; 2:2006.0015. [PMID: 16738560 PMCID: PMC1681489 DOI: 10.1038/msb4100057] [Citation(s) in RCA: 244] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2005] [Accepted: 02/23/2006] [Indexed: 12/18/2022] Open
Abstract
Recognition of pathogen-associated molecular signatures is critically important in proper activation of the immune system. The toll-like receptor (TLR) signaling network is responsible for innate immune response. In mammalians, there are 11 TLRs that recognize a variety of ligands from pathogens to trigger immunological responses. In this paper, we present a comprehensive map of TLRs and interleukin 1 receptor signaling networks based on papers published so far. The map illustrates the possible existence of a main network subsystem that has a bow-tie structure in which myeloid differentiation primary response gene 88 (MyD88) is a nonredundant core element, two collateral subsystems with small GTPase and phosphatidylinositol signaling, and MyD88-independent pathway. There is extensive crosstalk between the main bow-tie network and subsystems, as well as feedback and feedforward controls. One obvious feature of this network is the fragility against removal of the nonredundant core element, which is MyD88, and involvement of collateral subsystems for generating different reactions and gene expressions for different stimuli.
Collapse
Affiliation(s)
- Kanae Oda
- The Systems Biology Institute, Tokyo, Japan
- Department of Fundamental Science and Technology, Keio University, Tokyo, Japan
| | - Hiroaki Kitano
- The Systems Biology Institute, Tokyo, Japan
- Department of Fundamental Science and Technology, Keio University, Tokyo, Japan
- Sony Computer Science Laboratories Inc., Tokyo, Japan
- The Systems Biology Institute, Suite 6A, M31 6-31-15 Jingumae, Shibuya, Tokyo 150-0001, Japan. Tel.: +81 3 5468 1661; Fax: +81 3 5468 1664; E-mail:
| |
Collapse
|
725
|
Rowe DC, McGettrick AF, Latz E, Monks BG, Gay NJ, Yamamoto M, Akira S, O’Neill LA, Fitzgerald KA, Golenbock DT. The myristoylation of TRIF-related adaptor molecule is essential for Toll-like receptor 4 signal transduction. Proc Natl Acad Sci U S A 2006; 103:6299-304. [PMID: 16603631 PMCID: PMC1458872 DOI: 10.1073/pnas.0510041103] [Citation(s) in RCA: 213] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2005] [Indexed: 11/18/2022] Open
Abstract
TRIF-related adaptor molecule (TRAM) is the fourth Toll/IL-1 resistance domain-containing adaptor to be described that participates in Toll-like receptor (TLR) signaling. TRAM functions exclusively in the TLR4 pathway. Here we show by confocal microscopy that TRAM is localized in the plasma membrane and the Golgi apparatus, where it colocalizes with TLR4. Membrane localization of TRAM is the result of myristoylation because mutation of a predicted myristoylation site in TRAM (TRAM-G2A) brought about dissociation of TRAM from the membrane and its relocation to the cytosol. Further, TRAM, but not TRAM-G2A, was radiolabeled with [3H]myristate in vivo. Unlike wild-type TRAM, overexpression of TRAM-G2A failed to elicit either IFN regulatory factor 3 or NF-kappaB signaling. Moreover, TRAM-G2A was unable to reconstitute LPS responses in bone marrow-derived macrophages from TRAM-deficient mice. These observations provide clear evidence that the myristoylation of TRAM targets it to the plasma membrane, where it is essential for LPS responses through the TLR4 signal transduction pathway, and suggest a hitherto unappreciated manner in which LPS responses can be regulated.
Collapse
Affiliation(s)
- Daniel C. Rowe
- *Division of Infectious Disease and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | | | - Eicke Latz
- *Division of Infectious Disease and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Brian G. Monks
- *Division of Infectious Disease and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Nicholas J. Gay
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1YP, United Kingdom; and
| | - Masahiro Yamamoto
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Shizuo Akira
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Luke A. O’Neill
- Department of Biochemistry, Trinity College, Dublin 2, Ireland
| | - Katherine A. Fitzgerald
- *Division of Infectious Disease and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Douglas T. Golenbock
- *Division of Infectious Disease and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| |
Collapse
|
726
|
López CB, Yount JS, Moran TM. Toll-like receptor-independent triggering of dendritic cell maturation by viruses. J Virol 2006; 80:3128-34. [PMID: 16537581 PMCID: PMC1440398 DOI: 10.1128/jvi.80.7.3128-3134.2006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Carolina B López
- Department of Microbiology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | |
Collapse
|
727
|
Fredericksen BL, Gale M. West Nile virus evades activation of interferon regulatory factor 3 through RIG-I-dependent and -independent pathways without antagonizing host defense signaling. J Virol 2006; 80:2913-23. [PMID: 16501100 PMCID: PMC1395472 DOI: 10.1128/jvi.80.6.2913-2923.2006] [Citation(s) in RCA: 151] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The ability of viruses to control and/or evade the host antiviral response is critical to the establishment of a productive infection. We have previously shown that West Nile virus NY (WNV-NY) delays activation of interferon regulatory factor 3 (IRF-3), a transcription factor critical to the initiation of the antiviral response. Here we demonstrate that the delayed activation of IRF-3 is essential for WNV-NY to achieve maximum virus production. Furthermore, WNV-NY utilizes a unique mechanism to control activation of IRF-3. In contrast to many other viruses that impose a nonspecific block to the IRF-3 pathway, WNV-NY eludes detection by the host cell at early times postinfection. To better understand this process, we assessed the role of the pathogen recognition receptor (PRR) retinoic acid-inducible gene I (RIG-I) in sensing WNV-NY infection. RIG-I null mouse embryo fibroblasts (MEFs) retained the ability to respond to WNV-NY infection; however, the onset of the host response was delayed compared to wild-type (WT) MEFs. This suggests that RIG-I is involved in initially sensing WNV-NY infection, while other PRRs sustain and/or amplify the host response later in infection. The delayed initiation of the host response correlated with an increase in WNV-NY replication in RIG-I null MEFs compared to WT MEFs. Our data suggest that activation of the host response by RIG-I early in infection is important for controlling replication of WNV-NY. Furthermore, pathogenic strains of WNV may have evolved to circumvent stimulation of the host response until after replication is well under way.
Collapse
Affiliation(s)
- Brenda L Fredericksen
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, USA
| | | |
Collapse
|
728
|
Salaun B, Coste I, Rissoan MC, Lebecque SJ, Renno T. TLR3 Can Directly Trigger Apoptosis in Human Cancer Cells. THE JOURNAL OF IMMUNOLOGY 2006; 176:4894-901. [PMID: 16585585 DOI: 10.4049/jimmunol.176.8.4894] [Citation(s) in RCA: 360] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
TLRs function as molecular sensors to detect pathogen-derived products and trigger protective responses ranging from secretion of cytokines that increase the resistance of infected cells and chemokines that recruit immune cells to cell death that limits microbe spreading. Viral dsRNA participate in virus-infected cell apoptosis, but the signaling pathway involved remains unclear. In this study we show that synthetic dsRNA induces apoptosis of human breast cancer cells in a TLR3-dependent manner, which involves the molecular adaptor Toll/IL-1R domain-containing adapter inducing IFN-beta and type I IFN autocrine signaling, but occurs independently of the dsRNA-activated kinase. Moreover, detailed molecular analysis of dsRNA-induced cell death established the proapoptotic role of IL-1R-associated kinase-4 and NF-kappaB downstream of TLR3 as well as the activation of the extrinsic caspases. The direct proapoptotic activity of endogenous human TLR3 expressed by cancerous cells reveals a novel aspect of the multiple-faced TLR biology, which may open new clinical prospects for using TLR3 agonists as cytotoxic agents in selected cancers.
Collapse
Affiliation(s)
- Bruno Salaun
- Laboratory for Immunological Research Schering-Plough Research Institute, Dardilly, France
| | | | | | | | | |
Collapse
|
729
|
Affiliation(s)
- Fu-Shin X Yu
- Kresge Eye Institute/Department of Ophthalmology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | | |
Collapse
|
730
|
Sun D, Ding A. MyD88-mediated stabilization of interferon-gamma-induced cytokine and chemokine mRNA. Nat Immunol 2006; 7:375-81. [PMID: 16491077 DOI: 10.1038/ni1308] [Citation(s) in RCA: 176] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2005] [Accepted: 01/09/2006] [Indexed: 11/08/2022]
Abstract
The MyD88 adaptor protein is critical in Toll-like receptor and interleukin 1 receptor (IL-1R) signaling, but has not been linked to interferon-gamma receptor (IFN-gammaR) signaling. Here we demonstrate that MyD88 increased the half-life but not the synthesis of IFN-gamma-induced mRNA transcripts encoding tumor necrosis factor and IFN-gamma-inducible protein 10. IFN-gamma stimulation triggered a physical association between the IFN-gammaR1 and MyD88. Transcript stabilization required activation of mixed-lineage kinase 3 and p38 mitogen-activated protein kinase and the presence of an adenine-uridine-rich element in the transcript's 3' untranslated region. These results demonstrate a MyD88-dependent post-transcriptional mechanism through which IFN-gamma can enhance the expression of genes encoding proinflammatory molecules.
Collapse
MESH Headings
- 3' Untranslated Regions
- Adaptor Proteins, Signal Transducing/immunology
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Chemokine CXCL10
- Chemokines, CXC/biosynthesis
- Chemokines, CXC/genetics
- Chemokines, CXC/immunology
- Heterogeneous Nuclear Ribonucleoprotein D0
- Heterogeneous-Nuclear Ribonucleoprotein D/immunology
- Interferon-gamma/immunology
- Interferon-gamma/pharmacology
- MAP Kinase Kinase 3/immunology
- MAP Kinase Kinase Kinases
- Macrophages, Peritoneal/drug effects
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Knockout
- Mitogen-Activated Protein Kinase Kinases/immunology
- Myeloid Differentiation Factor 88
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Receptors, Interferon/immunology
- Receptors, Interferon/metabolism
- Recombinant Proteins
- Signal Transduction
- Tumor Necrosis Factor-alpha/biosynthesis
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/immunology
- p38 Mitogen-Activated Protein Kinases/immunology
- p38 Mitogen-Activated Protein Kinases/metabolism
- Mitogen-Activated Protein Kinase Kinase Kinase 11
Collapse
Affiliation(s)
- Dongxu Sun
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, New York 10021, USA
| | | |
Collapse
|
731
|
Hoyne GF, Goodnow CC. The use of genomewide ENU mutagenesis screens to unravel complex mammalian traits: identifying genes that regulate organ-specific and systemic autoimmunity. Immunol Rev 2006; 210:27-39. [PMID: 16623762 DOI: 10.1111/j.0105-2896.2006.00363.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
T-cell development is perhaps one of the best understood processes of mammalian cell differentiation, as many of the genes and pathways have been identified. By contrast, relatively little is known about the genes and pathways involved in immunological tolerance to self-antigens. Here, we describe the challenges associated with a genomewide screen designed at identifying new immune regulatory genes that uses a model of organ-specific autoimmunity leading to type 1 diabetes. The successful propagation and identification of the new gene variants will shed light on the various developmental checkpoints in lymphocyte development that are crucial for establishing tolerance to self-antigens.
Collapse
Affiliation(s)
- Gerard F Hoyne
- Australian Cancer Research Foundation Genetics Laboratory and Australian Phenomics Facility, John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia.
| | | |
Collapse
|
732
|
Kawai T, Akira S. Innate immune recognition of viral infection. Nat Immunol 2006; 7:131-7. [PMID: 16424890 DOI: 10.1038/ni1303] [Citation(s) in RCA: 1477] [Impact Index Per Article: 77.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2005] [Accepted: 11/14/2005] [Indexed: 11/09/2022]
Abstract
Induction of the antiviral innate immune response depends on recognition of viral components by host pattern-recognition receptors. Members of the Toll-like receptor family have emerged as key sensors that recognize viral components such as nucleic acids. Toll-like receptor signaling results in the production of type I interferon and inflammatory cytokines and leads to dendritic cell maturation and establishment of antiviral immunity. Cells also express cytoplasmic RNA helicases that function as alternative pattern-recognition receptors through recognition of double-stranded RNA produced during virus replication. These two classes of pattern-recognition receptor molecules are expressed in different intracellular compartments and induce type I interferon responses via distinct signaling pathways.
Collapse
Affiliation(s)
- Taro Kawai
- Exploratory Research for Advanced Technology, Japan Science and Technology Agency, Suita, Osaka 565-0871, Japan
| | | |
Collapse
|
733
|
|
734
|
Fischer H, Yamamoto M, Akira S, Beutler B, Svanborg C. Mechanism of pathogen-specific TLR4 activation in the mucosa: fimbriae, recognition receptors and adaptor protein selection. Eur J Immunol 2006; 36:267-77. [PMID: 16385628 DOI: 10.1002/eji.200535149] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The mucosal host defence discriminates pathogens from commensals, and prevents infection while allowing the normal flora to persist. Paradoxically, Toll-like receptors (TLR) control the mucosal defence against pathogens, even though the TLR recognise conserved molecules like LPS, which are shared between pathogens and commensals. This study proposes a mechanism of pathogen-specific mucosal TLR4 activation, involving adhesive ligands and their host cell receptors. TLR4 signalling was activated in CD14-negative, LPS-unresponsive epithelial cells by P fimbriated, uropathogenic Escherichia coli but not by a mutant lacking fimbriae. Epithelial TLR4 signalling in vivo involved the glycosphingolipid receptors for P fimbriae and the adaptor proteins Toll/IL-1R (TIR) domain-containing adaptor inducing IFN-beta (TRIF)/TRIF-related adaptor molecule (TRAM), but myeloid differentiation protein 88 (MyD88)/TIR domain-containing adaptor protein were not required for the epithelial response. Substituting the P fimbriae with type 1 fimbriae changed TLR4 signalling from the TRIF to the MyD88 adaptor pathway. In addition, the adaptor proteins and the fimbrial type were found to influence bacterial clearance. Trif(-/-) and Tram(-/-) mice remained infected with P fimbriated E. coli but cleared the type 1 fimbriated strain, while Myd88(-/-) mice became carriers of both the P and the type 1 fimbriated bacteria. Thus, TLR4 may be engaged specifically by pathogens, when the proper cell surface receptors are engaged by virulence ligands.
Collapse
Affiliation(s)
- Hans Fischer
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, S-22362 Lund, Sweden.
| | | | | | | | | |
Collapse
|
735
|
Abstract
Microorganisms that invade a vertebrate host are initially recognized by the innate immune system through germline-encoded pattern-recognition receptors (PRRs). Several classes of PRRs, including Toll-like receptors and cytoplasmic receptors, recognize distinct microbial components and directly activate immune cells. Exposure of immune cells to the ligands of these receptors activates intracellular signaling cascades that rapidly induce the expression of a variety of overlapping and unique genes involved in the inflammatory and immune responses. New insights into innate immunity are changing the way we think about pathogenesis and the treatment of infectious diseases, allergy, and autoimmunity.
Collapse
Affiliation(s)
- Shizuo Akira
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan.
| | | | | |
Collapse
|
736
|
Yasutomi M, Ohshima Y, Omata N, Yamada A, Iwasaki H, Urasaki Y, Mayumi M. Erythromycin differentially inhibits lipopolysaccharide- or poly(I:C)-induced but not peptidoglycan-induced activation of human monocyte-derived dendritic cells. THE JOURNAL OF IMMUNOLOGY 2006; 175:8069-76. [PMID: 16339544 DOI: 10.4049/jimmunol.175.12.8069] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Erythromycin (EM) has attracted attention because of its anti-inflammatory effect. Because dendritic cells (DCs) are the most potent APCs involved in numerous pathologic processes including innate immunity, we examined effects of EM on the activation of human DCs by pathogen-derived stimuli. Monocyte-derived DCs were pretreated with EM and subsequently stimulated with peptidoglycan, polyriboinosinic:polyribocytidylic acid (poly(I:C)), or LPS. The activation of DCs was assessed by surface molecule expression and cytokine production. To reveal the signaling pathways affected by EM, TLR expression, NF-kappaB, IFN regulatory factor-3, and AP-1 activation were examined. EM inhibited costimulatory molecule expression and cytokine production that was induced by poly(I:C) and LPS but not by peptidoglycan. EM pretreatment down- and up-regulated mRNA levels of TLR3 and TLR2, respectively, but did not affect that of TLR4. EM suppressed IFN regulatory factor-3 activation and IFN-beta production but not AP-1 activation induced by poly(I:C) and LPS. The inhibitory effect of EM on NF-kappaB activation was observed only in poly(I:C)-stimulated DCs. EM selectively suppressed activation of DCs induced by LPS and poly(I:C) in different ways, suggesting that the immuno-modulating effects of EM depend on the nature of pathogens. These results might explain why EM prevents the virus-induced exacerbation in the chronic inflammatory respiratory diseases and give us the clue to design new drugs to treat these diseases.
Collapse
Affiliation(s)
- Motoko Yasutomi
- Department of Pediatrics, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | | | | | | | | | | | | |
Collapse
|
737
|
Haller D. Intestinal epithelial cell signalling and host-derived negative regulators under chronic inflammation: to be or not to be activated determines the balance towards commensal bacteria. Neurogastroenterol Motil 2006; 18:184-99. [PMID: 16487409 DOI: 10.1111/j.1365-2982.2006.00762.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Advancing knowledge regarding the cellular mechanisms of intestinal inflammation has led to a better understanding of the disease pathology in patients with chronic disorders of the gut including inflammatory bowel disease, coeliac disease, lymphocytic colitis and irritable bowel syndrome. An emerging new paradigm suggests that changes in the homeostasis of bacteria- and host-derived signal transduction at the epithelial cell level may lead to functional and immune disturbances of the intestinal epithelium. It has become clear from numerous studies that enteric bacteria are a critical component in the development and prevention/treatment of chronic intestinal inflammation. Signal-specific activation of mitogen-activated protein kinases (MAPK), interferon-regulated factors (IRF) and the transcription factor NF-kappaB through pattern recognition receptor signalling effectively induce inflammatory defence mechanisms. Unbalanced activation of these innate signalling pathways because of host genetic predispositions and/or the lack of adequate anti-inflammatory feedback mechanisms may turn a physiological response into a pathological situation including failure of bacterial clearance and development of chronic inflammation. Host-derived regulators from the immune and enteric nerve system crosstalk to the innate signalling network of the intestinal epithelium in order to shape the extent and duration of inflammatory processes.
Collapse
Affiliation(s)
- D Haller
- Technical University of Munich, Else-Kroener-Fresenius Centre for Experimental Nutritional Medicine, Freising-Weihenstephan, Germany.
| |
Collapse
|
738
|
Abstract
Innate immunity is a primordial system that has a primary role in lung antimicrobial defenses. Recent advances in understanding the recognition systems by which cells of the innate immune system recognize and respond to microbial products have revolutionized the understanding of host defenses in the lungs and other tissues. The innate immune system includes lung leukocytes and also epithelial cells lining the alveolar surface and the conducting airways. The innate immune system drives adaptive immunity in the lungs and has important interactions with other systems, including apoptosis pathways and signaling pathways induced by mechanical stretch. Human diversity in innate immune responses could explain some of the variability seen in the responses of patients to bacterial, fungal, and viral infections in the lungs. New strategies to modify innate immune responses could be useful in limiting the adverse consequences of some inflammatory reactions in the lungs.
Collapse
Affiliation(s)
- Thomas R Martin
- Pulmonary Research Laboratories, VA Puget Sound Health Care System, 151L, 1660 South Columbian Way, Seattle, WA 98108, USA.
| | | |
Collapse
|
739
|
Eckert M, Wittmann I, Röllinghoff M, Gessner A, Schnare M. Endotoxin-induced expression of murine bactericidal permeability/increasing protein is mediated exclusively by toll/IL-1 receptor domain-containing adaptor inducing IFN-beta-dependent pathways. THE JOURNAL OF IMMUNOLOGY 2006; 176:522-8. [PMID: 16365446 DOI: 10.4049/jimmunol.176.1.522] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Antimicrobial effector proteins are a key mechanism for the innate immune system to combat pathogens once they infect the host. We report the identification and cloning of the mouse homologue of human bactericidal permeability/increasing protein (BPI). Mouse BPI is constitutively expressed in lymphatic organs and tissues as well as in mouse testis. Upon stimulation with different TLR ligands, mouse BPI is strongly expressed in granulocytes and, surprisingly, in bone marrow-derived dendritic cells. Mouse BPI is most strongly induced by bacterial LPS through a signaling pathway that is completely dependent on TLR4-Toll/IL-1R domain-containing adaptor inducing IFN-beta. Functional studies revealed that mouse BPI does have the potential to neutralize LPS and inhibits bacterial growth. Mouse BPI is expressed in granulocytes and bone marrow-derived dendritic cells, and the transcriptional activation is controlled by TLRs.
Collapse
Affiliation(s)
- Melanie Eckert
- Institute for Clinical Microbiology, Immunology, and Hygiene, University of Erlangen-Nuremberg, Erlangen, Germany
| | | | | | | | | |
Collapse
|
740
|
Chi H, Barry SP, Roth RJ, Wu JJ, Jones EA, Bennett AM, Flavell RA. Dynamic regulation of pro- and anti-inflammatory cytokines by MAPK phosphatase 1 (MKP-1) in innate immune responses. Proc Natl Acad Sci U S A 2006; 103:2274-9. [PMID: 16461893 PMCID: PMC1413743 DOI: 10.1073/pnas.0510965103] [Citation(s) in RCA: 471] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Engagement of Toll-like receptors (TLRs) on macrophages leads to activation of the mitogen-activated protein kinases (MAPKs), which contribute to innate immune responses. MAPK activity is regulated negatively by MAPK phosphatases (MKPs). MKP-1, the founding member of this family of dual-specificity phosphatases, has been implicated in regulating lipopolysaccharide (LPS) responses, but its role in TLR-mediated immune responses in vivo has not been defined. Here, we show that mice deficient in MKP-1 were highly susceptible to endotoxic shock in vivo, associated with enhanced production of proinflammatory cytokines TNF-alpha and IL-6 and an anti-inflammatory cytokine, IL-10. We further examined the regulation and function of MKP-1 in macrophages, a major cell type involved in endotoxic shock. MKP-1 was transiently induced by TLR stimulation through pathways mediated by both myeloid differentiation factor 88 (MyD88) and TIR domain-containing adaptor inducing IFN-beta (TRIF). MKP-1 deficiency led to sustained activation of p38 MAPK and c-Jun N-terminal kinase (JNK) in LPS-treated macrophages. In response to TLR signals, MKP-1-deficient macrophages produced 5- to 10-fold higher IL-10, which could be blocked by a p38 MAPK inhibitor. Thus, p38 MAPK plays a critical role in mediating IL-10 synthesis in TLR signaling. TNF-alpha was found to be more abundant in MKP-1-deficient macrophages within 2 hours of TLR stimulation, but its production was rapidly down-regulated by IL-10. Our studies demonstrate that MKP-1 attenuates the activities of p38 MAPK and JNK to regulate both pro- and anti-inflammatory cytokines in TLR signaling. These results highlight the complex mechanisms by which the MAPKs regulate innate immunity.
Collapse
Affiliation(s)
| | - Sean P. Barry
- *Section of Immunobiology
- Medical Molecular Biology Unit, Institute of Child Health, University College London, London WC1N 1EH, United Kingdom
| | | | | | | | | | - Richard A. Flavell
- *Section of Immunobiology
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520; and
- To whom correspondence should be addressed at:
Section of Immunobiology, Yale University School of Medicine, 300 Cedar Street, TAC S-569, New Haven, CT 06520. E-mail:
| |
Collapse
|
741
|
Querec T, Bennouna S, Alkan S, Laouar Y, Gorden K, Flavell R, Akira S, Ahmed R, Pulendran B. Yellow fever vaccine YF-17D activates multiple dendritic cell subsets via TLR2, 7, 8, and 9 to stimulate polyvalent immunity. ACTA ACUST UNITED AC 2006; 203:413-24. [PMID: 16461338 PMCID: PMC2118210 DOI: 10.1084/jem.20051720] [Citation(s) in RCA: 423] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The live attenuated yellow fever vaccine 17D (YF-17D) is one of the most effective vaccines available, with a 65-yr history of use in >400 million people globally. Despite this efficacy, there is presently no information about the immunological mechanisms by which YF-17D acts. Here, we present data that suggest that YF-17D activates multiple Toll-like receptors (TLRs) on dendritic cells (DCs) to elicit a broad spectrum of innate and adaptive immune responses. Specifically, YF-17D activates multiple DC subsets via TLRs 2, 7, 8, and 9 to elicit the proinflammatory cytokines interleukin (IL)-12p40, IL-6, and interferon-α. Interestingly, the resulting adaptive immune responses are characterized by a mixed T helper cell (Th)1/Th2 cytokine profile and antigen-specific CD8+ T cells. Furthermore, distinct TLRs appear to differentially control the Th1/Th2 balance; thus, whilst MyD88-deficient mice show a profound impairment of Th1 cytokines, TLR2-deficient mice show greatly enhanced Th1 and Tc1 responses to YF-17D. Together, these data enhance our understanding of the molecular mechanism of action of YF-17D, and highlight the potential of vaccination strategies that use combinations of different TLR ligands to stimulate polyvalent immune responses.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/physiology
- Amino Acid Sequence
- Animals
- Cell Line
- Cells, Cultured
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Humans
- Immunity, Active
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Molecular Sequence Data
- Myeloid Differentiation Factor 88
- Receptors, Interleukin-1/genetics
- Receptors, Interleukin-1/physiology
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Th2 Cells/immunology
- Th2 Cells/metabolism
- Toll-Like Receptor 2/physiology
- Toll-Like Receptor 7/physiology
- Toll-Like Receptor 8/physiology
- Toll-Like Receptor 9/physiology
- Toll-Like Receptors/physiology
- Vaccines, Attenuated/immunology
- Yellow Fever Vaccine/immunology
Collapse
Affiliation(s)
- Troy Querec
- Emory Vaccine Center, Emory University, Atlanta, GA 30329, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
742
|
Mahieu T, Park JM, Revets H, Pasche B, Lengeling A, Staelens J, Wullaert A, Vanlaere I, Hochepied T, van Roy F, Karin M, Libert C. The wild-derived inbred mouse strain SPRET/Ei is resistant to LPS and defective in IFN-beta production. Proc Natl Acad Sci U S A 2006; 103:2292-7. [PMID: 16455798 PMCID: PMC1413734 DOI: 10.1073/pnas.0510874103] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Although activation of Toll-like receptor 4 (TLR4)-positive cells is essential for eliminating Gram-negative bacteria, overactivation of these cells by the TLR4 ligand LPS initiates a systemic inflammatory reaction and shock. Here we demonstrate that SPRET/Ei mice, derived from Mus spretus, exhibit a dominant resistance against LPS-induced lethality. This resistance is mediated by bone marrow-derived cells. Macrophages from these mice exhibit normal signaling and gene expression responses that depend on the myeloid differentiation factor 88 adaptor protein, but they are impaired in IFN-beta production. The defect appears to be specific for IFN-beta, although the SPRET/Ei IFN-beta promoter is normal. In vivo IFN-beta induction by LPS or influenza virus is very low in SPRET/Ei mice, but IFN-beta-treatment restores the sensitivity to LPS, and IFN type 1 receptor-deficient mice are also resistant to LPS. Because of the defective induction of IFN-beta, these mice are completely resistant to Listeria monocytogenes and highly sensitive to Leishmania major infection. Stimulation of SPRET/Ei macrophages leads to rapid down-regulation of IFN type 1 receptor mRNA expression, which is reflected in poor induction of IFN-beta-dependent genes. This finding indicates that the resistance of SPRET/Ei mice to LPS is due to disruption of a positive-feedback loop that amplifies IFN-beta production. In contrast to TLR4-deficient mice, SPRET/Ei mice resist both LPS and sepsis induced with Klebsiella pneumoniae.
Collapse
Affiliation(s)
- Tina Mahieu
- *Department for Molecular Biomedical Research, Flanders Interuniversity Institute for Biotechnology and Ghent University, Technologiepark 927, B-9052 Ghent, Belgium
| | - Jin Mo Park
- Department of Pharmacology, University of California at San Diego, La Jolla, CA 92093
| | - Hilde Revets
- Department for Molecular and Cellular Interactions, Flanders Interuniversity Institute for Biotechnology and Free University Brussels, B-1640 Brussels, Belgium; and
| | - Bastian Pasche
- Young Investigator Group Infection Genetics, German Research Centre for Biotechnology, 38124 Braunschweig, Germany
| | - Andreas Lengeling
- Young Investigator Group Infection Genetics, German Research Centre for Biotechnology, 38124 Braunschweig, Germany
| | - Jan Staelens
- *Department for Molecular Biomedical Research, Flanders Interuniversity Institute for Biotechnology and Ghent University, Technologiepark 927, B-9052 Ghent, Belgium
| | - Andy Wullaert
- *Department for Molecular Biomedical Research, Flanders Interuniversity Institute for Biotechnology and Ghent University, Technologiepark 927, B-9052 Ghent, Belgium
| | - Ineke Vanlaere
- *Department for Molecular Biomedical Research, Flanders Interuniversity Institute for Biotechnology and Ghent University, Technologiepark 927, B-9052 Ghent, Belgium
| | - Tino Hochepied
- *Department for Molecular Biomedical Research, Flanders Interuniversity Institute for Biotechnology and Ghent University, Technologiepark 927, B-9052 Ghent, Belgium
| | - Frans van Roy
- *Department for Molecular Biomedical Research, Flanders Interuniversity Institute for Biotechnology and Ghent University, Technologiepark 927, B-9052 Ghent, Belgium
| | - Michael Karin
- Department of Pharmacology, University of California at San Diego, La Jolla, CA 92093
- **To whom correspondence may be addressed. E-mail:
or
| | - Claude Libert
- *Department for Molecular Biomedical Research, Flanders Interuniversity Institute for Biotechnology and Ghent University, Technologiepark 927, B-9052 Ghent, Belgium
- **To whom correspondence may be addressed. E-mail:
or
| |
Collapse
|
743
|
Silliman CC, Wang M. The merits of in vitro versus in vivo modeling in investigation of the immune system. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2006; 21:123-134. [PMID: 21783649 DOI: 10.1016/j.etap.2005.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Immunity is vital for determining self and for the recognition and swift eradication of foreign antigens without harming the host. Innate immunity developed in metazoan, multi-cellular organisms under overwhelming selection pressure of invasive microbes and, although imperfect, has performed admirably to enable the evolution of higher eukaryotes. Adaptive immunity developed within an existing innate immune system to more effectively eradicate foreign antigens, whether from pathogens, malignant cells, or microbial toxins, such that repeated stimulations with foreign antigens are more efficiently excluded. Investigation of the immune system requires both in vivo and in vitro experimentation, not only because of the inherent complexity of immunity and the required pertinence of using higher mammals to not falsely disrupt the immune system, but also to use isolates of the specific cellular and humoral components to determine function, signal transduction, and a possible role of these constituents without the complexity and redundancy of immunity in intact animals. The hypotheses of well-designed in vitro experiments must also be tested in intact in vivo models to determine relevance and to discard artifactual findings secondary to the in vitro environment. The following review outlines the basic constituents and functions of both adaptive and innate immunity to demonstrate the importance of both in vivo and in vitro investigation of immunity in our attempt to define host defense and to decrease morbidity and mortality in humans.
Collapse
Affiliation(s)
- Christopher C Silliman
- Bonfils Blood Center, 717 Yosemite Circle, Denver, CO 80230, USA; Department of Pediatrics, University of Colorado School of Medicine, Denver, CO 80262, USA; Department of Surgery, University of Colorado School of Medicine, Denver, CO 80262, USA
| | | |
Collapse
|
744
|
Abstract
Toll-like receptors (TLR) are critical sentinels of the host innate immune system. Prior evidence has clearly demonstrated that these receptors are essential to immune recognition of invading pathogens. However, there is emerging evidence that TLR signaling participates in inflammation that is not driven by microorganisms. In the setting of solid organ transplantation, there is accumulating evidence, both in experimental and clinical studies, that TLR signaling is involved in the immune recognition of allografts. Further investigation of how innate immunity impacts solid organ transplantation will likely lead to improved therapeutics for transplant recipients.
Collapse
Affiliation(s)
- Jagdeep Obhrai
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | | |
Collapse
|
745
|
Papathanasiou P, Goodnow CC. Connecting mammalian genome with phenome by ENU mouse mutagenesis: gene combinations specifying the immune system. Annu Rev Genet 2006; 39:241-62. [PMID: 16053407 DOI: 10.1146/annurev.genet.39.110304.095817] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The human and mouse genome sequences bring closer the goal of understanding how characteristics of adult mammalian physiology and pathology are encoded by DNA. Here we review the challenge of understanding how genes specify mammalian traits, with particular focus on the cells and behavior of the immune system. Summarized is the emerging experience, advantages, and limitations of using ethylnitrosourea (ENU) to modify the mouse genome and select informative variants by phenotypic screens, yielding two main conclusions. First, ENU-induced variation provides an eminently feasible route to understanding how the genome encodes important mammalian processes without any prior assumptions about genes, their chromosomal locations, or expression patterns. Second, ENU alleles match those arising by natural variation. By changing individual protein domains or splice products, these alleles reveal separate gene functions specified through protein combinations.
Collapse
Affiliation(s)
- Peter Papathanasiou
- Australian Cancer Research Foundation Genetics Laboratory and Australian Phenomics Facility, John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601 Australia.
| | | |
Collapse
|
746
|
Sasai M, Matsumoto M, Seya T. The Kinase Complex Responsible for IRF-3–Mediated IFN-β Production in Myeloid Dendritic Cells (mDC). ACTA ACUST UNITED AC 2006; 139:171-5. [PMID: 16452304 DOI: 10.1093/jb/mvj025] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Type I interferons (IFN) IFN-alpha and -beta play a central role in the induction of antiviral immunity, which involves up-regulation or activation of a large number of IFN-inducible genes in host immune competent cells. Initial events in the antiviral response may occur in myeloid dendritic cells (mDCs), and the proteins expressed provoke early responses to cope with concomitant infection in the host. The participation of transcription factors IRF-3/7, AP1 and NF-kappaB in IFN-beta promoter activation in mDCs is well established. An initial trigger of this event is a viral dsRNA that is recognized by proteins with an RNA-binding motif. Toll-like receptor (TLR) 3 on membranes and RIG-Iin the cytoplasm are molecules with dsRNA-recognition ability. Our main aim in the present review is to describe how IRF-3 and/or NF-kappaB are activated through the initial recognition of dsRNA by these pattern-recognition receptors. By analogy to the trimolecular complex of IKKgamma, IKKalpha and IKKbeta, thus far, IRF-3-activated kinases have been reported to be kinase complexes with trimolecular assembly. Two kinases, TBK1 and IKKepsilon, are thought to be linked to regulatory subunit TANK or NAP1 with no kinase activity like IKKgamma. The TLR3 and RIG-I pathways converge upstream of IRF-3, possibly at NAP1, the regulatory subunit of IRF-3-activating kinase. Thus, a novel function of the regulatory subunit has emerged. These proteins are involved in the TLR3 and RIG-I pathways, and act as adapters bridging on the dsRNA-recognition unit and IRF-3-activating kinases in addition to their kinase-regulatory function. Here, we summarize the properties of regulatory subunits NAP1 and TANK, and the mode of activation of NF-kappaB and IRF-3 in conjunction with the unique properties of the TLR3 function.
Collapse
Affiliation(s)
- Miwa Sasai
- Department of Microbiology and Immunology, Hokkaido University Graduate School of Medicine, Kita-15, Nishi-7, Sapporo 060-8638, Japan
| | | | | |
Collapse
|
747
|
Wang T, Chuang TH, Ronni T, Gu S, Du YC, Cai H, Sun HQ, Yin HL, Chen X. Flightless I Homolog Negatively Modulates the TLR Pathway. THE JOURNAL OF IMMUNOLOGY 2006; 176:1355-62. [PMID: 16424162 DOI: 10.4049/jimmunol.176.3.1355] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
To date, much of our knowledge about the signaling networks involved in the innate immune response has come from studies using nonphysiologic model systems rather than actual immune cells. In this study, we used a dual-tagging proteomic strategy to identify the components of the MyD88 signalosome in murine macrophages stimulated with lipid A. This systems approach revealed 16 potential MyD88-interacting partners, one of which, flightless I homolog (Fliih) was verified to interact with MyD88 and was further characterized as a negative regulator of the TLR4-MyD88 pathway. Conversely, a reduction in endogenous Fliih by small-interfering RNA enhanced the activation of NF-kappaB, as well as cytokine production by LPS. Results from immunoprecipitation and a two-hybrid assay further indicated that Fliih directly interfered with the formation of the TLR4-MyD88 signaling complex. These results in turn suggest a new basis for the regulation of the TLR pathway by Fliih.
Collapse
Affiliation(s)
- Tianyi Wang
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
748
|
Abstract
Vaccinia virus, a member of the Poxviridae, expresses many proteins involved in immune evasion. In this review, we present a brief characterisation of the virus and its effects on host cells and discuss representative secreted and intracellular proteins expressed by vaccinia virus that are involved in modulation of innate immunity. These proteins target different aspects of the innate response by binding cytokines and interferons, inhibiting cytokine synthesis, opposing apoptosis or interfering with different signalling pathways, including those triggered by interferons and toll-like receptors.
Collapse
Affiliation(s)
- I R Haga
- Department of Biochemistry, Trinity College, Dublin 2, Ireland.
| | | |
Collapse
|
749
|
Tabeta K, Hoebe K, Janssen EM, Du X, Georgel P, Crozat K, Mudd S, Mann N, Sovath S, Goode J, Shamel L, Herskovits AA, Portnoy DA, Cooke M, Tarantino LM, Wiltshire T, Steinberg BE, Grinstein S, Beutler B. The Unc93b1 mutation 3d disrupts exogenous antigen presentation and signaling via Toll-like receptors 3, 7 and 9. Nat Immunol 2006; 7:156-64. [PMID: 16415873 DOI: 10.1038/ni1297] [Citation(s) in RCA: 528] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2005] [Accepted: 12/01/2005] [Indexed: 11/08/2022]
Abstract
Here we have identified 'triple D' (3d), a recessive N-ethyl-N-nitrosourea-induced mutation and phenotype in which no signaling occurs via the intracellular Toll-like receptors 3, 7 and 9 (sensors for double-stranded RNA, single-stranded RNA and unmethylated DNA, respectively). The 3d mutation also prevented cross-presentation and diminished major histocompatibility complex class II presentation of exogenous antigen; it also caused hypersusceptibility to infection by mouse cytomegalovirus and other microbes. By positional identification, we found 3d to be a missense allele of Unc93b1, which encodes the 12-membrane-spanning protein UNC-93B, a highly conserved molecule found in the endoplasmic reticulum with multiple paralogs in mammals. Innate responses to nucleic acids and exogenous antigen presentation, which both initiate in endosomes, thus seem to depend on an endoplasmic reticulum-resident protein, which suggests communication between these organellar systems.
Collapse
Affiliation(s)
- Koichi Tabeta
- Department of Immunology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
750
|
Moynagh PN. TLR signalling and activation of IRFs: revisiting old friends from the NF-kappaB pathway. Trends Immunol 2006; 26:469-76. [PMID: 16006187 DOI: 10.1016/j.it.2005.06.009] [Citation(s) in RCA: 255] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2005] [Revised: 06/17/2005] [Accepted: 06/29/2005] [Indexed: 01/05/2023]
Abstract
Toll-like receptors (TLRs) are crucially important in the sensing of infectious agents. They serve to recognize pathogen-associated molecules and respond by triggering the induction of specific profiles of proteins that are tailored to the successful removal of the invading pathogens. The induction of TLR-responsive genes is mediated by the activation of transcription factors, and most interest has focussed on NF-kappaB, a transcription factor that is universally used by all TLRs. However, there has recently been a burgeoning effort to increase our appreciation of the importance of members of the interferon-regulatory factor (IRF) family in TLR signalling. This review will discuss the most recent findings relating to the regulation of IRF activity by TLRs and will highlight the rapidly increasing complexity of TLR signalling pathways.
Collapse
Affiliation(s)
- Paul N Moynagh
- Department of Pharmacology, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|