201
|
Single-nucleotide polymorphism of Toll-like receptor 4 and interleukin-10 in response to interferon-based therapy in Egyptian chronic hepatitis C patients. Arch Virol 2015; 160:2181-95. [DOI: 10.1007/s00705-015-2493-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 06/09/2015] [Indexed: 02/07/2023]
|
202
|
CD14 dependence of TLR4 endocytosis and TRIF signaling displays ligand specificity and is dissociable in endotoxin tolerance. Proc Natl Acad Sci U S A 2015; 112:8391-6. [PMID: 26106158 DOI: 10.1073/pnas.1424980112] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Dimerization of Toll-like receptor 4 (TLR4)/myeloid differentiation factor 2 (MD2) heterodimers is critical for both MyD88- and TIR-domain-containing adapter-inducing IFN-β (TRIF)-mediated signaling pathways. Recently, Zanoni et al. [(2011) Cell 147(4):868-880] reported that cluster of differentiation 14 (CD14) is required for LPS-/Escherichia coli- induced TLR4 internalization into endosomes and activation of TRIF-mediated signaling in macrophages. We confirmed their findings with LPS but report here that CD14 is not required for receptor endocytosis and downstream signaling mediated by TLR4/MD2 agonistic antibody (UT12) and synthetic small-molecule TLR4 ligands (1Z105) in murine macrophages. CD14 deficiency completely ablated the LPS-induced TBK1/IRF3 signaling axis that mediates production of IFN-β in murine macrophages without affecting MyD88-mediated signaling, including NF-κB, MAPK activation, and TNF-α and IL-6 production. However, neither the MyD88- nor TRIF-signaling pathways and their associated cytokine profiles were altered in the absence of CD14 in UT12- or 1Z105-treated murine macrophages. Eritoran (E5564), a lipid A antagonist that binds the MD2 "pocket," completely blocked LPS- and 1Z105-driven, but not UT12-induced, TLR4 dimerization and endocytosis. Furthermore, TLR4 endocytosis is induced in macrophages tolerized by exposure to either LPS or UT12 and is independent of CD14. These data indicate that TLR4 receptor endocytosis and the TRIF-signaling pathway are dissociable and that TLR4 internalization in macrophages can be induced by UT12, 1Z105, and during endotoxin tolerance in the absence of CD14.
Collapse
|
203
|
van Lieshout MHP, Florquin S, Vanʼt Veer C, de Vos AF, van der Poll T. TIR-Domain-Containing Adaptor-Inducing Interferon-β (TRIF) Mediates Antibacterial Defense during Gram-Negative Pneumonia by Inducing Interferon-x03B3. J Innate Immun 2015; 7:637-46. [PMID: 26065469 DOI: 10.1159/000430913] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/25/2015] [Indexed: 12/13/2022] Open
Abstract
Klebsiella pneumoniae is an important cause of Gram-negative pneumonia and sepsis. Mice deficient for TIR-domain-containing adaptor-inducing interferon-β (TRIF) demonstrate enhanced bacterial growth and dissemination during Klebsiella pneumonia. We show here that the impaired antibacterial defense of TRIF mutant mice is associated with absent interferon (IFN)-x03B3; production in the lungs. IFN-x03B3; production by splenocytes in response to K. pneumoniae in vitro was critically dependent on Toll-like receptor 4 (TLR4), the common TLR adaptor myeloid differentiation primary response gene (MyD88) and TRIF. Reconstitution of TRIF mutant mice with recombinant IFN-x03B3; via the airways reduced bacterial loads in lungs and distant body sites to levels measured in wild-type mice, and partially restored pulmonary cytokine levels. The IFN-x03B3;-induced, improved, enhanced antibacterial response in TRIF mutant mice occurred at the expense of increased hepatocellular injury. These data indicate that TRIF mediates antibacterial defense during Gram-negative pneumonia, at least in part, by inducing IFN-x03B3; at the primary site of infection.
Collapse
Affiliation(s)
- Miriam H P van Lieshout
- Center of Infection and Immunity Amsterdam, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
204
|
Mager LF, Riether C, Schürch CM, Banz Y, Wasmer MH, Stuber R, Theocharides AP, Li X, Xia Y, Saito H, Nakae S, Baerlocher GM, Manz MG, McCoy KD, Macpherson AJ, Ochsenbein AF, Beutler B, Krebs P. IL-33 signaling contributes to the pathogenesis of myeloproliferative neoplasms. J Clin Invest 2015; 125:2579-91. [PMID: 26011644 DOI: 10.1172/jci77347] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 04/23/2015] [Indexed: 12/16/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) are characterized by the clonal expansion of one or more myeloid cell lineage. In most cases, proliferation of the malignant clone is ascribed to defined genetic alterations. MPNs are also associated with aberrant expression and activity of multiple cytokines; however, the mechanisms by which these cytokines contribute to disease pathogenesis are poorly understood. Here, we reveal a non-redundant role for steady-state IL-33 in supporting dysregulated myelopoiesis in a murine model of MPN. Genetic ablation of the IL-33 signaling pathway was sufficient and necessary to restore normal hematopoiesis and abrogate MPN-like disease in animals lacking the inositol phosphatase SHIP. Stromal cell-derived IL-33 stimulated the secretion of cytokines and growth factors by myeloid and non-hematopoietic cells of the BM, resulting in myeloproliferation in SHIP-deficient animals. Additionally, in the transgenic JAK2V617F model, the onset of MPN was delayed in animals lacking IL-33 in radio-resistant cells. In human BM, we detected increased numbers of IL-33-expressing cells, specifically in biopsies from MPN patients. Exogenous IL-33 promoted cytokine production and colony formation by primary CD34+ MPN stem/progenitor cells from patients. Moreover, IL-33 improved the survival of JAK2V617F-positive cell lines. Together, these data indicate a central role for IL-33 signaling in the pathogenesis of MPNs.
Collapse
|
205
|
A20 restricts ubiquitination of pro-interleukin-1β protein complexes and suppresses NLRP3 inflammasome activity. Immunity 2015; 42:55-67. [PMID: 25607459 DOI: 10.1016/j.immuni.2014.12.031] [Citation(s) in RCA: 233] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 11/20/2014] [Indexed: 11/23/2022]
Abstract
Inappropriate inflammasome activation contributes to multiple human diseases, but the mechanisms by which inflammasomes are suppressed are poorly understood. The NF-κB inhibitor A20 is a ubiquitin-modifying enzyme that might be critical in preventing human inflammatory diseases. Here, we report that A20-deficient macrophages, unlike normal cells, exhibit spontaneous NLRP3 inflammasome activity to LPS alone. The kinase RIPK3, but not the adaptor MyD88, is required for this response. In normal cells, A20 constitutively associates with caspase-1 and pro-IL-1β, and NLRP3 activation further promotes A20 recruitment to the inflammasome. Pro-IL-1β also co-immunoprecipitates with RIPK1, RIPK3, caspase-1, and caspase-8 in a complex that is modified with K63-linked and unanchored polyubiquitin. In A20-deficient macrophages, this pro-IL-1β-associated ubiquitination is markedly increased in a RIPK3-dependent manner. Mass spectrometric and mutational analyses reveal that K133 of pro-IL-1β is a physiological ubiquitination site that supports processing. Our study reveals a mechanism by which A20 prevents inflammatory diseases.
Collapse
|
206
|
Kropp KA, Hsieh WY, Isern E, Forster T, Krause E, Brune W, Angulo A, Ghazal P. A temporal gate for viral enhancers to co-opt Toll-like-receptor transcriptional activation pathways upon acute infection. PLoS Pathog 2015; 11:e1004737. [PMID: 25856589 PMCID: PMC4391941 DOI: 10.1371/journal.ppat.1004737] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 02/09/2015] [Indexed: 12/27/2022] Open
Abstract
Viral engagement with macrophages activates Toll-Like-Receptors (TLRs) and viruses must contend with the ensuing inflammatory responses to successfully complete their replication cycle. To date, known counter-strategies involve the use of viral-encoded proteins that often employ mimicry mechanisms to block or redirect the host response to benefit the virus. Whether viral regulatory DNA sequences provide an opportunistic strategy by which viral enhancer elements functionally mimic innate immune enhancers is unknown. Here we find that host innate immune genes and the prototypical viral enhancer of cytomegalovirus (CMV) have comparable expression kinetics, and positively respond to common TLR agonists. In macrophages but not fibroblasts we show that activation of NFκB at immediate-early times of infection is independent of virion-associated protein, M45. We find upon virus infection or transfection of viral genomic DNA the TLR-agonist treatment results in significant enhancement of the virus transcription-replication cycle. In macrophage time-course infection experiments we demonstrate that TLR-agonist stimulation of the viral enhancer and replication cycle is strictly delimited by a temporal gate with a determined half-maximal time for enhancer-activation of 6 h; after which TLR-activation blocks the viral transcription-replication cycle. By performing a systematic siRNA screen of 149 innate immune regulatory factors we identify not only anticipated anti-viral and pro-viral contributions but also new factors involved in the CMV transcription-replication cycle. We identify a central convergent NFκB-SP1-RXR-IRF axis downstream of TLR-signalling. Activation of the RXR component potentiated direct and indirect TLR-induced activation of CMV transcription-replication cycle; whereas chromatin binding experiments using wild-type and enhancer-deletion virus revealed IRF3 and 5 as new pro-viral host transcription factor interactions with the CMV enhancer in macrophages. In a series of pharmacologic, siRNA and genetic loss-of-function experiments we determined that signalling mediated by the TLR-adaptor protein MyD88 plays a vital role for governing the inflammatory activation of the CMV enhancer in macrophages. Downstream TLR-regulated transcription factor binding motif disruption for NFκB, AP1 and CREB/ATF in the CMV enhancer demonstrated the requirement of these inflammatory signal-regulated elements in driving viral gene expression and growth in cells as well as in primary infection of neonatal mice. Thus, this study shows that the prototypical CMV enhancer, in a restricted time-gated manner, co-opts through DNA regulatory mimicry elements, innate-immune transcription factors to drive viral expression and replication in the face of on-going pro-inflammatory antiviral responses in vitro and in vivo and; suggests an unexpected role for inflammation in promoting acute infection and has important future implications for regulating latency. Here we discover how inflammatory signalling may unintentionally promote infection, as a result of viruses evolving DNA sequences, known as enhancers, which act as a bait to prey on the infected cell transcription factors induced by inflammation. The major inflammatory transcription factors activated are part of the TLR-signalling pathway. We find the prototypical viral enhancer of cytomegalovirus can be paradoxically boosted by activation of inflammatory “anti-viral” TLR-signalling independent of viral structural proteins. This leads to an increase in viral gene expression and replication in cell-culture and upon infection of mice. We identify an axis of inflammatory transcription factors, acting downstream of TLR-signalling but upstream of interferon inhibition. Mechanistically, the central TLR-adapter protein MyD88 is shown to play a critical role in promoting viral enhancer activity in the first 6h of infection. The co-option of TLR-signalling exceeds the usage of NFκB, and we identify IRF3 and 5 as newly found viral-enhancer interacting inflammatory transcription factors. Taken together this study reveals how virus enhancers, employ a path of least resistance by directly harnessing within a short temporal window, the activation of anti-viral signalling in macrophages to drive viral gene expression and replication to an extent that has not been recognised before.
Collapse
Affiliation(s)
- Kai A. Kropp
- Division of Pathway Medicine, Edinburgh Infectious Diseases, University of Edinburgh, Edinburgh, United Kingdom
- * E-mail: (KAK); (PG)
| | - Wei Yuan Hsieh
- Division of Pathway Medicine, Edinburgh Infectious Diseases, University of Edinburgh, Edinburgh, United Kingdom
| | - Elena Isern
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | - Thorsten Forster
- Division of Pathway Medicine, Edinburgh Infectious Diseases, University of Edinburgh, Edinburgh, United Kingdom
| | - Eva Krause
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Wolfram Brune
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Ana Angulo
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | - Peter Ghazal
- Division of Pathway Medicine, Edinburgh Infectious Diseases, University of Edinburgh, Edinburgh, United Kingdom
- SynthSys, University of Edinburgh, The King’s Buildings, Edinburgh, United Kingdom
- * E-mail: (KAK); (PG)
| |
Collapse
|
207
|
Günther C, Buchen B, He GW, Hornef M, Torow N, Neumann H, Wittkopf N, Martini E, Basic M, Bleich A, Watson AJM, Neurath MF, Becker C. Caspase-8 controls the gut response to microbial challenges by Tnf-α-dependent and independent pathways. Gut 2015; 64:601-10. [PMID: 25379949 PMCID: PMC4392221 DOI: 10.1136/gutjnl-2014-307226] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Intestinal epithelial cells (IEC) express toll-like receptors (TLR) that facilitate microbial recognition. Stimulation of TLR ligands induces a transient increase in epithelial cell shedding, a mechanism that serves the antibacterial and antiviral host defence of the epithelium and promotes elimination of intracellular pathogens. Although activation of the extrinsic apoptosis pathway has been described during inflammatory shedding, its functional involvement is currently unclear. DESIGN We investigated the functional involvement of caspase-8 signalling in microbial-induced intestinal cell shedding by injecting Lipopolysaccharide (LPS) to mimic bacterial pathogens and poly(I:C) as a probe for RNA viruses in vivo. RESULTS TLR stimulation of IEC was associated with a rapid activation of caspase-8 and increased epithelial cell shedding. In mice with an epithelial cell-specific deletion of caspase-8 TLR stimulation caused Rip3-dependent epithelial necroptosis instead of apoptosis. Mortality and tissue damage were more severe in mice in which IECs died by necroptosis than apoptosis. Inhibition of receptor-interacting protein (Rip) kinases rescued the epithelium from TLR-induced gut damage. TLR3-induced necroptosis was directly mediated via TRIF-dependent pathways, independent of Tnf-α and type III interferons, whereas TLR4-induced tissue damage was critically dependent on Tnf-α. CONCLUSIONS Together, our data demonstrate an essential role for caspase-8 in maintaining the gut barrier in response to mucosal pathogens by permitting inflammatory shedding and preventing necroptosis of infected cells. These data suggest that therapeutic strategies targeting the cell death machinery represent a promising new option for the treatment of inflammatory and infective enteropathies.
Collapse
Affiliation(s)
- Claudia Günther
- Medical Clinic 1, Friedrich Alexander University, Erlangen, Germany
| | - Barbara Buchen
- Medical Clinic 1, Friedrich Alexander University, Erlangen, Germany
| | - Gui-Wei He
- Medical Clinic 1, Friedrich Alexander University, Erlangen, Germany
| | - Mathias Hornef
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Natalia Torow
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Helmut Neumann
- Medical Clinic 1, Friedrich Alexander University, Erlangen, Germany
| | - Nadine Wittkopf
- Medical Clinic 1, Friedrich Alexander University, Erlangen, Germany
| | - Eva Martini
- Medical Clinic 1, Friedrich Alexander University, Erlangen, Germany
| | - Marijana Basic
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - André Bleich
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | | | - Markus F Neurath
- Medical Clinic 1, Friedrich Alexander University, Erlangen, Germany
| | - Christoph Becker
- Medical Clinic 1, Friedrich Alexander University, Erlangen, Germany
| |
Collapse
|
208
|
Abstract
The mammalian Toll-like receptor (TLR) family consists of 13 members, and recognizes specific patterns of microbial components, called pathogen-associated molecular patterns (PAMPs). TLR-dependent recognition of PAMPs leads to activation of the innate immune system, which subsequently leads to activation of antigen-specific adaptive immunity. The TLR-mediated signaling pathways consist of the MyD88-dependent pathway and TRIF-dependent pathway, both of which induce gene expression. This unit discusses mammalian TLRs (TLR1 to 13) that have an essential role in the innate immune recognition of microorganisms. Also discussed are TLR-mediated signaling pathways and antibodies that are available to detect specific TLRs.
Collapse
Affiliation(s)
- Kiyoshi Takeda
- Graduate School of Medicine, Osaka University, Osaka, Japan.,WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Shizuo Akira
- WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| |
Collapse
|
209
|
Abstract
All aspects of the pathogenesis of atherosclerosis are critically influenced by the inflammatory response in vascular plaques. Research in the field of innate immunity from the past 2 decades has uncovered many novel mechanisms elucidating how immune cells sense microbes, tissue damage, and metabolic derangements. Here, we summarize which triggers of innate immunity appear during atherogenesis and by which pathways they can contribute to inflammation in atherosclerotic plaques. The increased understanding gained from studies assessing how immune activation is associated with the pathogenesis of atherosclerosis has provided many novel targets for potential therapeutic intervention. Excitingly, the concept that inflammation may be the core of cardiovascular disease is currently being clinically evaluated and will probably encourage further studies in this area.
Collapse
Affiliation(s)
- Sebastian Zimmer
- From the Medizinische Klinik und Poliklinik II (S.Z.) and Institute of Innate Immunity (A.G., E.L.), University Hospitals Bonn, Bonn, Germany; Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester (E.L.); and German Center of Neurodegenerative Diseases (DZNE), Bonn, Germany (E.L.)
| | - Alena Grebe
- From the Medizinische Klinik und Poliklinik II (S.Z.) and Institute of Innate Immunity (A.G., E.L.), University Hospitals Bonn, Bonn, Germany; Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester (E.L.); and German Center of Neurodegenerative Diseases (DZNE), Bonn, Germany (E.L.)
| | - Eicke Latz
- From the Medizinische Klinik und Poliklinik II (S.Z.) and Institute of Innate Immunity (A.G., E.L.), University Hospitals Bonn, Bonn, Germany; Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester (E.L.); and German Center of Neurodegenerative Diseases (DZNE), Bonn, Germany (E.L.).
| |
Collapse
|
210
|
Brinkmann MM, Dağ F, Hengel H, Messerle M, Kalinke U, Čičin-Šain L. Cytomegalovirus immune evasion of myeloid lineage cells. Med Microbiol Immunol 2015; 204:367-82. [PMID: 25776081 DOI: 10.1007/s00430-015-0403-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 02/28/2015] [Indexed: 12/23/2022]
Abstract
Cytomegalovirus (CMV) evades the immune system in many different ways, allowing the virus to grow and its progeny to spread in the face of an adverse environment. Mounting evidence about the antiviral role of myeloid immune cells has prompted the research of CMV immune evasion mechanisms targeting these cells. Several cells of the myeloid lineage, such as monocytes, dendritic cells and macrophages, play a role in viral control, but are also permissive for CMV and are naturally infected by it. Therefore, CMV evasion of myeloid cells involves mechanisms that qualitatively differ from the evasion of non-CMV-permissive immune cells of the lymphoid lineage. The evasion of myeloid cells includes effects in cis, where the virus modulates the immune signaling pathways within the infected myeloid cell, and those in trans, where the virus affects somatic cells targeted by cytokines released from myeloid cells. This review presents an overview of CMV strategies to modulate and evade the antiviral activity of myeloid cells in cis and in trans.
Collapse
Affiliation(s)
- Melanie M Brinkmann
- Viral Immune Modulation Research Group, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124, Brunswick, Germany
| | | | | | | | | | | |
Collapse
|
211
|
Manček-Keber M, Jerala R. Postulates for validating TLR4 agonists. Eur J Immunol 2015; 45:356-70. [DOI: 10.1002/eji.201444462] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 10/20/2014] [Accepted: 12/01/2014] [Indexed: 02/03/2023]
Affiliation(s)
- Mateja Manček-Keber
- Department of Biotechnology; National Institute of Chemistry; Ljubljana Slovenia
- EN-FIST Centre of Excellence; Ljubljana Slovenia
| | - Roman Jerala
- Department of Biotechnology; National Institute of Chemistry; Ljubljana Slovenia
- EN-FIST Centre of Excellence; Ljubljana Slovenia
| |
Collapse
|
212
|
Narayanan KB, Park HH. Toll/interleukin-1 receptor (TIR) domain-mediated cellular signaling pathways. Apoptosis 2015; 20:196-209. [DOI: 10.1007/s10495-014-1073-1] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
213
|
Synthetic Toll-like receptor 4 (TLR4) and TLR7 ligands as influenza virus vaccine adjuvants induce rapid, sustained, and broadly protective responses. J Virol 2015; 89:3221-35. [PMID: 25568203 DOI: 10.1128/jvi.03337-14] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Current vaccines against influenza virus infection rely on the induction of neutralizing antibodies targeting the globular head of the viral hemagglutinin (HA). Protection against seasonal antigenic drift or sporadic pandemic outbreaks requires further vaccine development to induce cross-protective humoral responses, potentially to the more conserved HA stalk region. Here, we present a novel viral vaccine adjuvant comprised of two synthetic ligands for Toll-like receptor 4 (TLR4) and TLR7. 1Z105 is a substituted pyrimido[5,4-b]indole specific for the TLR4-MD2 complex, and 1V270 is a phospholipid-conjugated TLR7 agonist. Separately, 1Z105 induces rapid Th2-associated IgG1 responses, and 1V270 potently generates Th1 cellular immunity. 1Z105 and 1V270 in combination with recombinant HA from the A/Puerto Rico/8/1934 strain (rPR/8 HA) effectively induces rapid and sustained humoral immunity that is protective against lethal challenge with a homologous virus. More importantly, immunization with the combined adjuvant and rPR/8 HA, a commercially available split vaccine, or chimeric rHA antigens significantly improves protection against both heterologous and heterosubtypic challenge viruses. Heterosubtypic protection is associated with broadly reactive antibodies to HA stalk epitopes. Histological examination and cytokine profiling reveal that intramuscular (i.m.) administration of 1Z105 and 1V270 is less reactogenic than a squalene-based adjuvant, AddaVax. In summary, the combination of 1Z105 and 1V270 with a recombinant HA induces rapid, long-lasting, and balanced Th1- and Th2-type immunity; demonstrates efficacy in a variety of murine influenza virus vaccine models assaying homologous, heterologous, and heterosubtypic challenge viruses; and has an excellent safety profile. IMPORTANCE Novel adjuvants are needed to enhance immunogenicity and increase the protective breadth of influenza virus vaccines to reduce the seasonal disease burden and ensure pandemic preparedness. We show here that the combination of synthetic Toll-like receptor 4 (TLR4) and TLR7 ligands is a potent adjuvant for recombinant influenza virus hemagglutinin, inducing rapid and sustained immunity that is protective against influenza viruses in homologous, heterologous, and heterosubtypic challenge models. Combining TLR4 and TLR7 ligands balances Th1- and Th2-type immune responses for long-lived cellular and neutralizing humoral immunity against the viral hemagglutinin. The combined adjuvant has an attractive safety profile and the potential to augment seasonal-vaccine breadth, contribute to a broadly neutralizing universal vaccine formulation, and improve response time in an emerging pandemic.
Collapse
|
214
|
Chintala SK, Putris N, Geno M. Activation of TLR3 promotes the degeneration of retinal ganglion cells by upregulating the protein levels of JNK3. Invest Ophthalmol Vis Sci 2015; 56:505-14. [PMID: 25564448 DOI: 10.1167/iovs.14-15539] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
PURPOSE To investigate whether activation of Toll-like receptor 3 (TLR3) promotes the degeneration of retinal ganglion cells (RGCs) by upregulating the protein levels of c-jun N-terminal kinase 3 (JNK3). METHODS Toll-like receptor 3-specific activator, Poly(I:C) (polyinosinic-polycytidylic acid), or PBS was injected into the vitreous humor of Thy1-YFP mice. At 24, 48, and 72 hours after treatments, degeneration of RGCs was assessed by using antibodies against brain-specific homeobox/POU domain protein 3a (Brn3a). A TLR3-specific inhibitor was injected into the vitreous humor with or without Poly(I:C). Western blot assays were performed to determine relative levels of TLR3, JNK3, pJNK3, and sterile alpha and HEAT/Armadillo motif-containing 1 (SARM1) proteins in retinal protein extracts, and immunohistochemistry assays were performed to determine their cellular localization in the retina. Mouse eyes were treated with Poly(I:C) or PBS along with MitoTracker Red, and colocalization of MitoTracker Red and JNK3 in the retinas was determined by using antibodies against JNK3. RESULTS Poly(I:C) activated TLR3 and upregulated its downstream target protein JNK3 but not SARM1 in the retina. Poly(I:C) activated TLR3 and upregulated JNK3 specifically in RGCs and promoted a significant degeneration of RGCs over a 72-hour time period. Toll-like receptor 3 upregulated the levels of JNK3 protein in the cytoplasm of RGCs, but not in the mitochondria. Toll-like receptor 3-specific inhibitor downregulated Poly(I:C)-mediated upregulation of JNK3 protein, and, in turn, significantly attenuated TLR3-induced degeneration of RGCs. CONCLUSIONS Results presented in this study show that the activation of TLR3 alone promotes the degeneration of RGCs by upregulating the protein levels of JNK3.
Collapse
Affiliation(s)
- Shravan K Chintala
- Laboratory of Ophthalmic Neurobiology, Eye Research Institute of Oakland University, Rochester, Michigan, United States
| | - Nahrain Putris
- Laboratory of Ophthalmic Neurobiology, Eye Research Institute of Oakland University, Rochester, Michigan, United States
| | - Mason Geno
- Laboratory of Ophthalmic Neurobiology, Eye Research Institute of Oakland University, Rochester, Michigan, United States
| |
Collapse
|
215
|
Park H, Noh ALSM, Kang JH, Sim JS, Lee DS, Yim M. Peroxiredoxin II negatively regulates lipopolysaccharide-induced osteoclast formation and bone loss via JNK and STAT3. Antioxid Redox Signal 2015; 22:63-77. [PMID: 25074339 PMCID: PMC4270137 DOI: 10.1089/ars.2013.5748] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
AIMS Lipopolysaccharide (LPS) is considered a prominent pathogenic factor in inflammatory bone diseases. LPS challenge contributes to the production of reactive oxygen species (ROS) in diverse inflammatory diseases. However, its mechanism remains to be clarified in bone. Thus, we investigated the critical mechanism of ROS in LPS-induced osteoclastogenesis and bone loss. RESULTS Antioxidant prevented LPS-induced osteoclast formation via inhibition of nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1) and c-Fos expression in preosteoclasts. Moreover, LPS-induced osteoclast formation via ROS was attenuated by treatment with c-Jun N-terminal protein kinase (JNK) inhibitor. Interestingly, LPS also activated signal transducer and activator of transcription 3 (STAT3), which is suppressed by antioxidants. We found that knockdown of STAT3 or use of a STAT3 inhibitor resulted in a significant reduction in interleukin-1 beta (IL-1β), interleukin-6 (IL-6), and nitric oxide (NO) production, followed by decreased osteoclast formation by LPS. Peroxiredoxin II (PrxII) is a member of the antioxidant enzyme family, and it plays a protective role against oxidative damage caused by ROS. In our study, ROS production and osteoclast formation by LPS was significantly enhanced in PrxII(-/-) cells. Moreover, JNK-mediated c-Fos and NFATc1 expression was promoted in PrxII(-/-) cells. Furthermore, STAT3 activation and accompanying IL-1β, IL-6, and NO production was also increased in PrxII(-/-) cells. Consistent with the in vitro result, PrxII-deficient mice showed increased osteoclast formation and bone loss by LPS challenge compared with wild-type mice. INNOVATION For the first time, we showed that LPS-induced ROS signaling is dependent on the coordinated mechanism of JNK and STAT3 during osteoclastogenesis, which is negatively regulated by PrxII. CONCLUSION We suggest that PrxII could be useful in the development of a novel target for inflammatory bone loss.
Collapse
Affiliation(s)
- Hyojung Park
- 1 College of Pharmacy, Sookmyung Women's University , Seoul, Republic of Korea
| | | | | | | | | | | |
Collapse
|
216
|
Coquenlorge S, Duchalais E, Chevalier J, Cossais F, Rolli-Derkinderen M, Neunlist M. Modulation of lipopolysaccharide-induced neuronal response by activation of the enteric nervous system. J Neuroinflammation 2014; 11:202. [PMID: 25497784 PMCID: PMC4279994 DOI: 10.1186/s12974-014-0202-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 11/14/2014] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Evidence continues to mount concerning the importance of the enteric nervous system (ENS) in controlling numerous intestinal functions in addition to motility and epithelial functions. Nevertheless, little is known concerning the direct participation of the ENS in the inflammatory response of the gut during infectious or inflammatory insults. In the present study we analyzed the ENS response to bacterial lipopolysaccharide, in particular the production of a major proinflammatory cytokine, tumor necrosis factor-alpha (TNF-α). METHODS TNF-α expression (measured by qPCR, quantitative Polymerase Chain Reaction) and production (measured by ELISA) were measured in human longitudinal muscle-myenteric plexus (LMMP) and rat ENS primary cultures (rENSpc). They were either treated or not treated with lipopolysaccharide (LPS) in the presence or not of electrical field stimulation (EFS). Activation of extracellular signal-regulated kinase (ERK) and 5'-adenosine monophosphate-activated protein kinase (AMPK) pathways was analyzed by immunocytochemistry and Western blot analysis. Their implications were studied using specific inhibitors (U0126, mitogen-activated protein kinase kinase, MEK, inhibitor and C compound, AMPK inhibitor). We also analyzed toll-like receptor 2 (TLR2) expression and interleukin-6 (IL-6) production after LPS treatment simultaneously with EFS or TNF-α-neutralizing antibody. RESULTS Treatment of human LMMP or rENSpc with LPS induced an increase in TNF-α production. Activation of the ENS by EFS significantly inhibited TNF-α production. This regulation occurred at the transcriptional level. Signaling analyses showed that LPS induced activation of ERK but not AMPK, which was constitutively activated in rENSpc neurons. Both U0126 and C compound almost completely prevented LPS-induced TNF-α production. In the presence of LPS, EFS inhibited the ERK and AMPK pathways. In addition, we demonstrated using TNF-α-neutralizing antibody that LPS-induced TNF-α production increased TLR2 expression and reduced IL-6 production. CONCLUSIONS Our results show that LPS induced TNF-α production by enteric neurons through activation of the canonical ERK pathway and also in an AMPK-dependent manner. ENS activation through the inhibition of these pathways decreased TNF-α production, thereby modulating the inflammatory response induced by endotoxin.
Collapse
Affiliation(s)
- Sabrina Coquenlorge
- Neuropathies of the enteric nervous system and digestive diseases, INSERM UMR913, School of Medicine, University of Nantes, 1, rue Gaston Veil, Nantes, F-44035, France. .,University of Nantes, 1 quai de Tourville, BP 13522, Nantes, Cedex 1, F-44035, France. .,Institut des Maladies de l'Appareil Digestif, Centre Hospitalier Universitaire, Nantes, Hopital Hôtel-Dieu, 1 place Alexis Ricordeau, Nantes, F-44093, France. .,Centre de Recherche en Nutrition Humaine, Hopital Hôtel-Dieu, 1 place Alexis Ricordeau, Nantes, F-44093, France.
| | - Emilie Duchalais
- Neuropathies of the enteric nervous system and digestive diseases, INSERM UMR913, School of Medicine, University of Nantes, 1, rue Gaston Veil, Nantes, F-44035, France. .,University of Nantes, 1 quai de Tourville, BP 13522, Nantes, Cedex 1, F-44035, France. .,Institut des Maladies de l'Appareil Digestif, Centre Hospitalier Universitaire, Nantes, Hopital Hôtel-Dieu, 1 place Alexis Ricordeau, Nantes, F-44093, France. .,Centre de Recherche en Nutrition Humaine, Hopital Hôtel-Dieu, 1 place Alexis Ricordeau, Nantes, F-44093, France.
| | - Julien Chevalier
- Neuropathies of the enteric nervous system and digestive diseases, INSERM UMR913, School of Medicine, University of Nantes, 1, rue Gaston Veil, Nantes, F-44035, France. .,University of Nantes, 1 quai de Tourville, BP 13522, Nantes, Cedex 1, F-44035, France. .,Institut des Maladies de l'Appareil Digestif, Centre Hospitalier Universitaire, Nantes, Hopital Hôtel-Dieu, 1 place Alexis Ricordeau, Nantes, F-44093, France. .,Centre de Recherche en Nutrition Humaine, Hopital Hôtel-Dieu, 1 place Alexis Ricordeau, Nantes, F-44093, France.
| | - Francois Cossais
- Neuropathies of the enteric nervous system and digestive diseases, INSERM UMR913, School of Medicine, University of Nantes, 1, rue Gaston Veil, Nantes, F-44035, France. .,University of Nantes, 1 quai de Tourville, BP 13522, Nantes, Cedex 1, F-44035, France. .,Institut des Maladies de l'Appareil Digestif, Centre Hospitalier Universitaire, Nantes, Hopital Hôtel-Dieu, 1 place Alexis Ricordeau, Nantes, F-44093, France. .,Centre de Recherche en Nutrition Humaine, Hopital Hôtel-Dieu, 1 place Alexis Ricordeau, Nantes, F-44093, France.
| | - Malvyne Rolli-Derkinderen
- Neuropathies of the enteric nervous system and digestive diseases, INSERM UMR913, School of Medicine, University of Nantes, 1, rue Gaston Veil, Nantes, F-44035, France. .,University of Nantes, 1 quai de Tourville, BP 13522, Nantes, Cedex 1, F-44035, France. .,Institut des Maladies de l'Appareil Digestif, Centre Hospitalier Universitaire, Nantes, Hopital Hôtel-Dieu, 1 place Alexis Ricordeau, Nantes, F-44093, France. .,Centre de Recherche en Nutrition Humaine, Hopital Hôtel-Dieu, 1 place Alexis Ricordeau, Nantes, F-44093, France.
| | - Michel Neunlist
- Neuropathies of the enteric nervous system and digestive diseases, INSERM UMR913, School of Medicine, University of Nantes, 1, rue Gaston Veil, Nantes, F-44035, France. .,University of Nantes, 1 quai de Tourville, BP 13522, Nantes, Cedex 1, F-44035, France. .,Institut des Maladies de l'Appareil Digestif, Centre Hospitalier Universitaire, Nantes, Hopital Hôtel-Dieu, 1 place Alexis Ricordeau, Nantes, F-44093, France. .,Centre de Recherche en Nutrition Humaine, Hopital Hôtel-Dieu, 1 place Alexis Ricordeau, Nantes, F-44093, France.
| |
Collapse
|
217
|
Nilsen NJ, Vladimer GI, Stenvik J, Orning MPA, Zeid-Kilani MV, Bugge M, Bergstroem B, Conlon J, Husebye H, Hise AG, Fitzgerald KA, Espevik T, Lien E. A role for the adaptor proteins TRAM and TRIF in toll-like receptor 2 signaling. J Biol Chem 2014; 290:3209-22. [PMID: 25505250 DOI: 10.1074/jbc.m114.593426] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Toll-like receptors (TLRs) are involved in sensing invading microbes by host innate immunity. TLR2 recognizes bacterial lipoproteins/lipopeptides, and lipopolysaccharide activates TLR4. TLR2 and TLR4 signal via the Toll/interleukin-1 receptor adaptors MyD88 and MAL, leading to NF-κB activation. TLR4 also utilizes the adaptors TRAM and TRIF, resulting in activation of interferon regulatory factor (IRF) 3. Here, we report a new role for TRAM and TRIF in TLR2 regulation and signaling. Interestingly, we observed that TLR2-mediated induction of the chemokine Ccl5 was impaired in TRAM or TRIF deficient macrophages. Inhibition of endocytosis reduced Ccl5 release, and the data also suggested that TRAM and TLR2 co-localize in early endosomes, supporting the hypothesis that signaling may occur from an intracellular compartment. Ccl5 release following lipoprotein challenge additionally involved the kinase Tbk-1 and Irf3, as well as MyD88 and Irf1. Induction of Interferon-β and Ccl4 by lipoproteins was also partially impaired in cells lacking TRIF cells. Our results show a novel function of TRAM and TRIF in TLR2-mediated signal transduction, and the findings broaden our understanding of how Toll/interleukin-1 receptor adaptor proteins may participate in signaling downstream from TLR2.
Collapse
Affiliation(s)
- Nadra J Nilsen
- From the Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, and the KG Jebsen Center for Myeloma Research, Norwegian University of Science and Technology, N-7489 Trondheim, Norway,
| | - Gregory I Vladimer
- the Department of Medicine, Division of Infectious Diseases and Immunology, Program in Innate Immunity, University of Massachusetts Medical School, Worcester, Massachusetts 01605, and
| | - Jørgen Stenvik
- From the Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, and
| | - M Pontus A Orning
- From the Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, and the Department of Medicine, Division of Infectious Diseases and Immunology, Program in Innate Immunity, University of Massachusetts Medical School, Worcester, Massachusetts 01605, and
| | - Maria V Zeid-Kilani
- From the Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, and
| | - Marit Bugge
- From the Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, and
| | - Bjarte Bergstroem
- From the Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, and
| | - Joseph Conlon
- the Department of Medicine, Division of Infectious Diseases and Immunology, Program in Innate Immunity, University of Massachusetts Medical School, Worcester, Massachusetts 01605, and
| | - Harald Husebye
- From the Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, and
| | - Amy G Hise
- the Center for Global Health and Diseases and Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106
| | - Katherine A Fitzgerald
- From the Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, and the Department of Medicine, Division of Infectious Diseases and Immunology, Program in Innate Immunity, University of Massachusetts Medical School, Worcester, Massachusetts 01605, and
| | - Terje Espevik
- From the Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, and the KG Jebsen Center for Myeloma Research, Norwegian University of Science and Technology, N-7489 Trondheim, Norway
| | - Egil Lien
- From the Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, and the Department of Medicine, Division of Infectious Diseases and Immunology, Program in Innate Immunity, University of Massachusetts Medical School, Worcester, Massachusetts 01605, and
| |
Collapse
|
218
|
Role of toll-like receptors in multiple myeloma and recent advances. Exp Hematol 2014; 43:158-67. [PMID: 25462020 DOI: 10.1016/j.exphem.2014.11.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 11/02/2014] [Accepted: 11/10/2014] [Indexed: 12/22/2022]
Abstract
Multiple myeloma (MM) is a hematologic malignancy characterized as an abnormal proliferation and invasion of plasma cells into the bone marrow. Toll-like receptors (ТLRs) connect the innate and adaptive immune responses and represent a significant and potentially linking element between inflammation and cancer. When TLRs bind to their ligands, they trigger two major signaling pathways such that both share overlapping downstream signals: one is a myeloid differentiation primary response 88 (MyD88)-dependent production and activation of nuclear factor-κB, whereas the other is a MyD88-independent production of type-I interferon. Whereas the MyD88 pathway results in proinflammatory cytokine production, the other pathway stimulates cell proliferation. Dysregulations of these pathways may eventually lead to abnormal cell proliferation and MM. Despite recent biomedical advances, MM continues to be an incurable disease. There are an increasing number of TLR-based therapeutic approaches currently being tested in a number of preclinical and clinical studies. We here attempt to outline in detail the currently available information on TLRs in various types of cancer.
Collapse
|
219
|
Kolb JP, Casella CR, SenGupta S, Chilton PM, Mitchell TC. Type I interferon signaling contributes to the bias that Toll-like receptor 4 exhibits for signaling mediated by the adaptor protein TRIF. Sci Signal 2014; 7:ra108. [PMID: 25389373 PMCID: PMC4459894 DOI: 10.1126/scisignal.2005442] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Signaling by Toll-like receptor 4 (TLR4) is mediated by either of two adaptor proteins: myeloid differentiation marker 88 (MyD88) or Toll-interleukin-1 (IL-1) receptor (TIR) domain-containing adaptor inducing interferon-β (TRIF). Whereas MyD88-mediated signaling leads to proinflammatory responses, TRIF-mediated signaling leads to less toxic immunostimulatory responses that are beneficial in boosting vaccine responses. The hypothesis that monophosphorylated lipid A structures act as TRIF-biased agonists of TLR4 offered a potential mechanism to explain their clinical value as vaccine adjuvants, but studies of TRIF-biased agonists have been contradictory. In experiments with mouse dendritic cells, we found that irrespective of the agonist used, TLR4 functioned as a TRIF-biased signaling system through a mechanism that depended on the autocrine and paracrine effects of type I interferons. The TLR4 agonist synthetic lipid A induced expression of TRIF-dependent genes at lower concentrations than were necessary to induce the expression of genes that depend on MyD88-mediated signaling. Blockade of type I interferon signaling selectively decreased the potency of lipid A (increased the concentration required) in inducing the expression of TRIF-dependent genes, thereby eliminating adaptor bias. These data may explain how high-potency TLR4 agonists can act as clinically useful vaccine adjuvants by selectively activating TRIF-dependent signaling events required for immunostimulation, without or only weakly activating potentially harmful MyD88-dependent inflammatory responses.
Collapse
Affiliation(s)
- Joseph P Kolb
- Department of Microbiology and Immunology, University of Louisville School of Medicine, 570 South Preston Street, Louisville, KY 40202, USA. Institute for Cellular Therapeutics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Carolyn R Casella
- Institute for Cellular Therapeutics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Shuvasree SenGupta
- Department of Microbiology and Immunology, University of Louisville School of Medicine, 570 South Preston Street, Louisville, KY 40202, USA. Institute for Cellular Therapeutics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Paula M Chilton
- Department of Microbiology and Immunology, University of Louisville School of Medicine, 570 South Preston Street, Louisville, KY 40202, USA. Institute for Cellular Therapeutics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Thomas C Mitchell
- Department of Microbiology and Immunology, University of Louisville School of Medicine, 570 South Preston Street, Louisville, KY 40202, USA. Institute for Cellular Therapeutics, University of Louisville School of Medicine, Louisville, KY 40202, USA.
| |
Collapse
|
220
|
Kuriakose T, Rada B, Watford WT. Tumor progression locus 2-dependent oxidative burst drives phosphorylation of extracellular signal-regulated kinase during TLR3 and 9 signaling. J Biol Chem 2014; 289:36089-100. [PMID: 25378393 DOI: 10.1074/jbc.m114.587121] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Signal transduction via NFκB and MAP kinase cascades is a universal response initiated upon pathogen recognition by Toll-like receptors (TLRs). How activation of these divergent signaling pathways is integrated to dictate distinct immune responses to diverse pathogens is still incompletely understood. Herein, contrary to current perception, we demonstrate that a signaling pathway defined by the inhibitor of κB kinase β (IKKβ), MAP3 kinase tumor progression locus 2 (Tpl2/MAP3K8), and MAP kinase ERK is differentially activated by TLRs. TLRs 2, 4, and 7 directly activate this inflammatory axis, inducing immediate ERK phosphorylation and early TNFα secretion. In addition to TLR adaptor proteins, IKKβ-Tpl2-ERK activation by TLR4 is regulated by the TLR4 co-receptor CD14 and the tyrosine kinase Syk. Signals from TLRs 3 and 9 do not initiate early activation of IKKβ-Tpl2-ERK pathway but instead induce delayed, NADPH-oxidase-dependent ERK phosphorylation and TNFα secretion via autocrine reactive oxygen species signaling. Unexpectedly, Tpl2 is an essential regulator of ROS production during TLR signaling. Overall, our study reveals distinct mechanisms activating a common inflammatory signaling cascade and delineates differences in MyD88-dependent signaling between endosomal TLRs 7 and 9. These findings further confirm the importance of Tpl2 in innate host defense mechanisms and also enhance our understanding of how the immune system tailors pathogen-specific gene expression patterns.
Collapse
Affiliation(s)
- Teneema Kuriakose
- From the Department of Infectious Diseases, The University of Georgia, College of Veterinary Medicine, Athens, Georgia 30602
| | - Balázs Rada
- From the Department of Infectious Diseases, The University of Georgia, College of Veterinary Medicine, Athens, Georgia 30602
| | - Wendy T Watford
- From the Department of Infectious Diseases, The University of Georgia, College of Veterinary Medicine, Athens, Georgia 30602
| |
Collapse
|
221
|
Lawrence H, Deehan D, Holland J, Kirby J, Tyson-Capper A. The immunobiology of cobalt: demonstration of a potential aetiology for inflammatory pseudotumours after metal-on-metal replacement of the hip. Bone Joint J 2014; 96-B:1172-7. [PMID: 25183586 DOI: 10.1302/0301-620x.96b9.33476] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Abnormal wear of cobalt-containing metal-on-metal joints is associated with inflammatory pseudotumours. Cobalt ions activate human toll-like receptor 4 (TLR4), which normally responds to bacterial lipopolysaccharide (LPS) in sepsis. Activation of TLR4 by LPS increases the expression of chemokines IL-8 and CXCL10, which recruit leukocytes and activated T-cells, respectively. This study was designed to determine whether cobalt induces a similar inflammatory response to LPS by promoting the expression of IL-8 and CXCL10. A human monocytic cell line, derived from acute monocytic leukaemia, was treated with cobalt ions and expression of IL-8 and CXCL10 measured at mRNA and protein levels. Cobalt-treated macrophages showed a 60-fold increase in IL-8 mRNA, and an eightfold increase in production of the mature chemokine (both p < 0.001); expression of the CXCL10 gene and protein was also significantly increased by cobalt (both p < 0.001). Experiments were also performed in the presence of CLI-095, a TLR4-specific antagonist which abrogated the cobalt-mediated increase in IL-8 and CXCL10 expression. These findings suggest that cobalt ions induce inflammation similar to that observed during sepsis by the simultaneous activation of two TLR4-mediated signalling pathways. These pathways result in increased production of IL-8 and CXCL10, and may be implicated in pseudotumour formation following metal-on-metal replacement.
Collapse
Affiliation(s)
- H Lawrence
- Newcastle University, Institute of Cellular Medicine, 3rd Floor, William Leech Building, Faculty of Medical Sciences, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - D Deehan
- Freeman Hospital, Department of Orthopaedics, Freeman Hospital, Freeman Road, Newcastle upon Tyne, NE7 7DN, UK
| | - J Holland
- Freeman Hospital, Department of Orthopaedics, Freeman Hospital, Freeman Road, Newcastle upon Tyne, NE7 7DN, UK
| | - J Kirby
- Newcastle University, Institute of Cellular Medicine, 3rd Floor, William Leech Building, Faculty of Medical Sciences, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - A Tyson-Capper
- Newcastle University, Institute of Cellular Medicine, 3rd Floor, William Leech Building, Faculty of Medical Sciences, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| |
Collapse
|
222
|
Schwenk R, DeBot M, Porter M, Nikki J, Rein L, Spaccapelo R, Crisanti A, Wightman PD, Ockenhouse CF, Dutta S. IgG2 antibodies against a clinical grade Plasmodium falciparum CSP vaccine antigen associate with protection against transgenic sporozoite challenge in mice. PLoS One 2014; 9:e111020. [PMID: 25343487 PMCID: PMC4208815 DOI: 10.1371/journal.pone.0111020] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 09/19/2014] [Indexed: 12/26/2022] Open
Abstract
The availability of a highly purified and well characterized circumsporozoite protein (CSP) is essential to improve upon the partial success of recombinant CSP-based malaria vaccine candidates. Soluble, near full-length, Plasmodium falciparum CSP vaccine antigen (CS/D) was produced in E. coli under bio-production conditions that comply with current Good Manufacturing Practices (cGMP). A mouse immunogenicity study was conducted using a stable oil-in-water emulsion (SE) of CS/D in combination with the Toll-Like Receptor 4 (TLR4) agonist Glucopyranosyl Lipid A (GLA/SE), or one of two TLR7/8 agonists: R848 (un-conjugated) or 3M-051 (covalently conjugated). Compared to Alum and SE, GLA/SE induced higher CS/D specific antibody response in Balb/c mice. Subclass analysis showed higher IgG2:IgG1 ratio of GLA/SE induced antibodies as compared to Alum and SE. TLR synergy was not observed when soluble R848 was mixed with GLA/SE. Antibody response of 3M051 formulations in Balb/c was similar to GLA/SE, except for the higher IgG2:IgG1 ratio and a trend towards higher T cell responses in 3M051 containing groups. However, no synergistic enhancement of antibody and T cell response was evident when 3M051 conjugate was mixed with GLA/SE. In C57Bl/6 mice, CS/D adjuvanted with 3M051/SE or GLA/SE induced higher CSP repeat specific titers compared to SE. While, 3M051 induced antibodies had high IgG2c:IgG1 ratio, GLA/SE promoted high levels of both IgG1 and IgG2c. GLA/SE also induced more potent T-cell responses compared to SE in two independent C57/BL6 vaccination studies, suggesting a balanced and productive TH1/TH2 response. GLA and 3M-051 similarly enhanced the protective efficacy of CS/D against challenge with a transgenic P. berghei parasite and most importantly, high levels of cytophilic IgG2 antibodies were associated with protection in this model. Our data indicated that the cGMP-grade, soluble CS/D antigen combined with the TLR4-containing adjuvant GLA/SE warrants further evaluation for protective responses in humans.
Collapse
Affiliation(s)
- Robert Schwenk
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - Margot DeBot
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - Michael Porter
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - Jennifer Nikki
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - Lisa Rein
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - Roberta Spaccapelo
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Paul D. Wightman
- 3M Drug Delivery Systems, St. Paul, MN, United States of America
| | - Christian F. Ockenhouse
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - Sheetij Dutta
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- * E-mail:
| |
Collapse
|
223
|
Pérez-Cano FJ, Massot-Cladera M, Rodríguez-Lagunas MJ, Castell M. Flavonoids Affect Host-Microbiota Crosstalk through TLR Modulation. Antioxidants (Basel) 2014; 3:649-70. [PMID: 26785232 PMCID: PMC4665504 DOI: 10.3390/antiox3040649] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 08/07/2014] [Accepted: 09/26/2014] [Indexed: 12/18/2022] Open
Abstract
Interaction between host cells and microbes is known as crosstalk. Among other mechanisms, this takes place when certain molecules of the micro-organisms are recognized by the toll-like receptors (TLRs) in the body cells, mainly in the intestinal epithelial cells and in the immune cells. TLRs belong to the pattern-recognition receptors and represent the first line of defense against pathogens, playing a pivotal role in both innate and adaptive immunity. Dysregulation in the activity of such receptors can lead to the development of chronic and severe inflammation as well as immunological disorders. Among components present in the diet, flavonoids have been suggested as antioxidant dietary factors able to modulate TLR-mediated signaling pathways. This review focuses on the molecular targets involved in the modulatory action of flavonoids on TLR-mediated signaling pathways, providing an overview of the mechanisms involved in such action. Particular flavonoids have been able to modify the composition of the microbiota, to modulate TLR gene and protein expression, and to regulate the downstream signaling molecules involved in the TLR pathway. These synergistic mechanisms suggest the role of some flavonoids in the preventive effect on certain chronic diseases.
Collapse
Affiliation(s)
- Francisco J Pérez-Cano
- Departament de Fisiologia, Facultat de Farmàcia, Universitat de Barcelona, Spain.
- Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB), E-08028 Barcelona, Spain.
| | - Malen Massot-Cladera
- Departament de Fisiologia, Facultat de Farmàcia, Universitat de Barcelona, Spain.
- Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB), E-08028 Barcelona, Spain.
| | - Maria J Rodríguez-Lagunas
- Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB), E-08028 Barcelona, Spain.
- Departament de Ciències Fisiològiques II, Facultat de Medicina, Universitat de Barcelona, Feixa Llarga s/n, L'Hospitalet de Llobregat, E-08907 Barcelona, Spain.
| | - Margarida Castell
- Departament de Fisiologia, Facultat de Farmàcia, Universitat de Barcelona, Spain.
- Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB), E-08028 Barcelona, Spain.
| |
Collapse
|
224
|
Cipriano C, Maiti A, Hale G, Jiranek W. The host response: Toll-like receptor expression in periprosthetic tissues as a biomarker for deep joint infection. J Bone Joint Surg Am 2014; 96:1692-8. [PMID: 25320195 DOI: 10.2106/jbjs.m.01295] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Toll-like receptors (TLRs) 1 and 6 are consistent molecular indicators of the host inflammatory response against bacterial infection. Our aims were to determine whether TLR elevation could be detected in infected periprosthetic tissues and to assess the utility of these biomarkers as tests for detecting a periprosthetic joint infection. METHODS Fifty-nine patients undergoing revision total joint arthroplasty (twenty-seven hips and thirty-two knees) were prospectively evaluated for periprosthetic joint infection according to currently recommended diagnostic criteria. Nine patients were excluded because of insufficient work-up, leaving fifty available for study. Of these, twenty-one were categorized as infected and twenty-nine as noninfected. Periprosthetic tissues were collected intraoperatively, and total RNA was extracted by standard techniques. Expression of TLR messenger RNAs was assessed by first-strand complementary DNA synthesis from 1 μg of total RNA followed by real-time PCR (polymerase chain reaction). Results were normalized relative to the housekeeping gene GAPDH (glyceraldehyde 3-phosphate dehydrogenase). Expression of TLRs 1, 6, and 10 in the infected and noninfected groups was compared with use of the Student t test. The receiver operating characteristic curve, area under the curve (AUC), sensitivity, specificity, positive likelihood ratio (LR+), and negative likelihood ratio (LR-) were calculated to determine the accuracy of each TLR for predicting periprosthetic joint infection at its optimal diagnostic threshold. RESULTS Mean TLR1 mRNA expression was significantly elevated in infected compared with noninfected samples (0.600 compared with 0.005, p = 0.0003); the same was true of TLR6 (0.208 compared with 0.0165, p = 0.0059) but not of TLR10 (0.00019 compared with 0.00014, p = 0.6238). The AUC was 0.995 for TLR1, 0.883 for TLR6, and 0.546 for TLR10. The optimal threshold for diagnosing periprosthetic joint infection was 0.0924 for TLR1 (sensitivity = 95.2%, specificity = 100%, LR+ = 13.80, LR- = 0.91) and 0.0215 for TLR6 (sensitivity = 85.7%, specificity = 82.8%, LR+ = 4.98, LR- = 0.83). CONCLUSIONS In our pilot study, TLR1 expression in periprosthetic tissues most accurately predicted periprosthetic joint infection. This measure of the host response may be particularly helpful in detecting culture-negative infections and avoiding false positives resulting from contamination. LEVEL OF EVIDENCE Diagnostic Level III. See Instructions for Authors for a complete description of levels of evidence.
Collapse
Affiliation(s)
- Cara Cipriano
- Department of Orthopaedic Surgery, Virginia Commonwealth University, 1112 East Clay Street, McGuire Hall Annex, Richmond, VA 23298. E-mail address for C. Cipriano: . E-mail address for A. Maiti:
| | - Aparna Maiti
- Department of Orthopaedic Surgery, Virginia Commonwealth University, 1112 East Clay Street, McGuire Hall Annex, Richmond, VA 23298. E-mail address for C. Cipriano: . E-mail address for A. Maiti:
| | - Gregory Hale
- Department of Orthopaedic Surgery, Virginia Commonwealth University, 1112 East Clay Street, McGuire Hall Annex, Richmond, VA 23298. E-mail address for C. Cipriano: . E-mail address for A. Maiti:
| | - William Jiranek
- Department of Orthopaedic Surgery, Virginia Commonwealth University, 1112 East Clay Street, McGuire Hall Annex, Richmond, VA 23298. E-mail address for C. Cipriano: . E-mail address for A. Maiti:
| |
Collapse
|
225
|
Mouse ENU Mutagenesis to Understand Immunity to Infection: Methods, Selected Examples, and Perspectives. Genes (Basel) 2014; 5:887-925. [PMID: 25268389 PMCID: PMC4276919 DOI: 10.3390/genes5040887] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 08/19/2014] [Accepted: 08/21/2014] [Indexed: 12/30/2022] Open
Abstract
Infectious diseases are responsible for over 25% of deaths globally, but many more individuals are exposed to deadly pathogens. The outcome of infection results from a set of diverse factors including pathogen virulence factors, the environment, and the genetic make-up of the host. The completion of the human reference genome sequence in 2004 along with technological advances have tremendously accelerated and renovated the tools to study the genetic etiology of infectious diseases in humans and its best characterized mammalian model, the mouse. Advancements in mouse genomic resources have accelerated genome-wide functional approaches, such as gene-driven and phenotype-driven mutagenesis, bringing to the fore the use of mouse models that reproduce accurately many aspects of the pathogenesis of human infectious diseases. Treatment with the mutagen N-ethyl-N-nitrosourea (ENU) has become the most popular phenotype-driven approach. Our team and others have employed mouse ENU mutagenesis to identify host genes that directly impact susceptibility to pathogens of global significance. In this review, we first describe the strategies and tools used in mouse genetics to understand immunity to infection with special emphasis on chemical mutagenesis of the mouse germ-line together with current strategies to efficiently identify functional mutations using next generation sequencing. Then, we highlight illustrative examples of genes, proteins, and cellular signatures that have been revealed by ENU screens and have been shown to be involved in susceptibility or resistance to infectious diseases caused by parasites, bacteria, and viruses.
Collapse
|
226
|
Smith SM. Role of Toll-like receptors in Helicobacter pylori infection and immunity. World J Gastrointest Pathophysiol 2014; 5:133-146. [PMID: 25133016 PMCID: PMC4133513 DOI: 10.4291/wjgp.v5.i3.133] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 02/25/2014] [Accepted: 05/16/2014] [Indexed: 02/07/2023] Open
Abstract
The gram-negative bacterium Helicobacter pylori (H. pylori) infects the stomachs of approximately half of the world's population. Although infection induces an immune response that contributes to chronic gastric inflammation, the response is not sufficient to eliminate the bacterium. H. pylori infection causes peptic ulcers, gastric cancer and mucosa-associated lymphoid tissue lymphoma. Disease outcome is linked to the severity of the host inflammatory response. Gastric epithelial cells represent the first line of innate immune defence against H. pylori, and respond to infection by initiating numerous cell signalling cascades, resulting in cytokine induction and the subsequent recruitment of inflammatory cells to the gastric mucosa. Pathogen recognition receptors of the Toll-like receptor (TLR) family mediate many of these cell signalling events. This review discusses recent findings on the role of various TLRs in the recognition of H. pylori in distinct cell types, describes the TLRs responsible for the recognition of individual H. pylori components and outlines the influence of innate immune activation on the subsequent development of the adaptive immune response. The mechanistic identification of host mediators of H. pylori-induced pathogenesis has the potential to reveal drug targets and opportunities for therapeutic intervention or prevention of H. pylori-associated disease by means of vaccines or immunomodulatory therapy.
Collapse
|
227
|
Spanier J, Lienenklaus S, Paijo J, Kessler A, Borst K, Heindorf S, Baker DP, Kröger A, Weiss S, Detje CN, Staeheli P, Kalinke U. Concomitant TLR/RLH signaling of radioresistant and radiosensitive cells is essential for protection against vesicular stomatitis virus infection. THE JOURNAL OF IMMUNOLOGY 2014; 193:3045-54. [PMID: 25127863 DOI: 10.4049/jimmunol.1400959] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Several studies indicated that TLR as well as retinoic acid-inducible gene I-like helicase (RLH) signaling contribute to vesicular stomatitis virus (VSV)-mediated triggering of type I IFN (IFN-I) responses. Nevertheless, TLR-deficient MyD88(-/-)Trif(-/-) mice and RLH-deficient caspase activation and recruitment domain adaptor inducing IFN-β (Cardif)(-/-) mice showed only marginally enhanced susceptibility to lethal VSV i.v. infection. Therefore, we addressed whether concomitant TLR and RLH signaling, or some other additional mechanism, played a role. To this end, we generated MyD88(-/-)Trif(-/-)Cardif(-/-) (MyTrCa(-/-)) mice that succumbed to low-dose i.v. VSV infection with similar kinetics as IFN-I receptor-deficient mice. Three independent approaches (i.e., analysis of IFN-α/β serum levels, experiments with IFN-β reporter mice, and investigation of local IFN-stimulated gene induction) revealed that MyTrCa(-/-) mice did not mount IFN-I responses following VSV infection. Of note, treatment with rIFN-α protected the animals, qualifying MyTrCa(-/-) mice as a model to study the contribution of different immune cell subsets to the production of antiviral IFN-I. Upon adoptive transfer of wild-type plasmacytoid dendritic cells and subsequent VSV infection, MyTrCa(-/-) mice displayed significantly reduced viral loads in peripheral organs and showed prolonged survival. On the contrary, adoptive transfer of wild-type myeloid dendritic cells did not have such effects. Analysis of bone marrow chimeric mice revealed that TLR and RLH signaling of radioresistant and radiosensitive cells was required for efficient protection. Thus, upon VSV infection, plasmacytoid dendritic cell-derived IFN-I primarily protects peripheral organs, whereas concomitant TLR and RLH signaling of radioresistant stroma cells as well as of radiosensitive immune cells is needed to effectively protect against lethal disease.
Collapse
Affiliation(s)
- Julia Spanier
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research, Braunschweig, and the Hannover Medical School, 30625 Hannover, Germany
| | - Stefan Lienenklaus
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Jennifer Paijo
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research, Braunschweig, and the Hannover Medical School, 30625 Hannover, Germany
| | - Annett Kessler
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research, Braunschweig, and the Hannover Medical School, 30625 Hannover, Germany
| | - Katharina Borst
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research, Braunschweig, and the Hannover Medical School, 30625 Hannover, Germany
| | - Sabrina Heindorf
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research, Braunschweig, and the Hannover Medical School, 30625 Hannover, Germany
| | | | - Andrea Kröger
- Research Group on Innate Immunity and Infection, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; and
| | - Siegfried Weiss
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Claudia N Detje
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research, Braunschweig, and the Hannover Medical School, 30625 Hannover, Germany
| | - Peter Staeheli
- Department of Virology, Institute for Medical Microbiology and Hygiene, Albert-Ludwigs-Universität, 79104 Freiburg, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research, Braunschweig, and the Hannover Medical School, 30625 Hannover, Germany;
| |
Collapse
|
228
|
Datta B, Njau F, Thalmann J, Haller H, Wagner AD. Differential infection outcome of Chlamydia trachomatis in human blood monocytes and monocyte-derived dendritic cells. BMC Microbiol 2014; 14:209. [PMID: 25123797 PMCID: PMC4236547 DOI: 10.1186/s12866-014-0209-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 07/21/2014] [Indexed: 02/05/2023] Open
Abstract
Background Chlamydia trachomatis is an intracellular bacteria which consist of three biovariants; trachoma (serovars A-C), urogenital (serovars D-K) and lymphogranuloma venereum (L1-L3), causing a wide spectrum of disease in humans. Monocytes are considered to disseminate this pathogen throughout the body while dendritic cells (DCs) play an important role in mediating immune response against bacterial infection. To determine the fate of C. trachomatis within human peripheral blood monocytes and monocyte-derived DCs, these two sets of immune cells were infected with serovars Ba, D and L2, representative of the three biovariants of C. trachomatis. Results Our study revealed that the different serovars primarily infect monocytes and DCs in a comparable fashion, however undergo differential infection outcome, serovar L2 being the only candidate to inflict active infection. Moreover, the C. trachomatis serovars Ba and D become persistent in monocytes while the serovars predominantly suffer degradation within DCs. Effects of persistence gene Indoleamine 2, 3-dioxygenase (IDO) was not clearly evident in the differential infection outcome. The heightened levels of inflammatory cytokines secreted by the chlamydial infection in DCs compared to monocytes seemed to be instrumental for this consequence. The immune genes induced in monocytes and DCs against chlamydial infection involves a different set of Toll-like receptors, indicating that distinct intracellular signalling pathways are adopted for immune response. Conclusion Our results demonstrate that the host pathogen interaction in chlamydia infection is not only serovar specific but manifests cell specific features, inducing separate immune response cascade in monocytes and DCs.
Collapse
|
229
|
Aranda F, Vacchelli E, Obrist F, Eggermont A, Galon J, Sautès-Fridman C, Cremer I, Henrik ter Meulen J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Toll-like receptor agonists in oncological indications. Oncoimmunology 2014; 3:e29179. [PMID: 25083332 PMCID: PMC4091055 DOI: 10.4161/onci.29179] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 05/09/2014] [Indexed: 12/20/2022] Open
Abstract
Toll-like receptors (TLRs) are an evolutionarily conserved group of enzymatically inactive, single membrane-spanning proteins that recognize a wide panel of exogenous and endogenous danger signals. Besides constituting a crucial component of the innate immune response to bacterial and viral pathogens, TLRs appear to play a major role in anticancer immunosurveillance. In line with this notion, several natural and synthetic TLR ligands have been intensively investigated for their ability to boost tumor-targeting immune responses elicited by a variety of immunotherapeutic and chemotherapeutic interventions. Three of these agents are currently approved by the US Food and Drug Administration (FDA) or equivalent regulatory agencies for use in cancer patients: the so-called bacillus Calmette-Guérin, monophosphoryl lipid A, and imiquimod. However, the number of clinical trials testing the therapeutic potential of both FDA-approved and experimental TLR agonists in cancer patients is stably decreasing, suggesting that drug developers and oncologists are refocusing their interest on alternative immunostimulatory agents. Here, we summarize recent findings on the use of TLR agonists in cancer patients and discuss how the clinical evaluation of FDA-approved and experimental TLR ligands has evolved since the publication of our first Trial Watch dealing with this topic.
Collapse
Affiliation(s)
- Fernando Aranda
- Gustave Roussy; Villejuif, France
- INSERM, UMRS1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France
- Université Paris-Sud/Paris XI; Paris, France
| | - Erika Vacchelli
- Gustave Roussy; Villejuif, France
- INSERM, UMRS1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France
- Université Paris-Sud/Paris XI; Paris, France
| | - Florine Obrist
- Gustave Roussy; Villejuif, France
- INSERM, UMRS1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France
- Université Paris-Sud/Paris XI; Paris, France
| | | | - Jérôme Galon
- INSERM, UMRS1138; Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France
- Laboratory of Integrative Cancer Immunology, Centre de Recherche des Cordeliers; Paris, France
| | - Catherine Sautès-Fridman
- INSERM, UMRS1138; Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
- Equipe 13, Centre de Recherche des Cordeliers; Paris, France
| | - Isabelle Cremer
- INSERM, UMRS1138; Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
- Equipe 13, Centre de Recherche des Cordeliers; Paris, France
| | | | - Laurence Zitvogel
- Gustave Roussy; Villejuif, France
- INSERM, U1015; CICBT507; Villejuif, France
| | - Guido Kroemer
- INSERM, UMRS1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP; Villejuif, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy; Villejuif, France
| | - Lorenzo Galluzzi
- Gustave Roussy; Villejuif, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France
| |
Collapse
|
230
|
Nan Y, Ma Z, Wang R, Yu Y, Kannan H, Fredericksen B, Zhang YJ. Enhancement of interferon induction by ORF3 product of hepatitis E virus. J Virol 2014; 88:8696-705. [PMID: 24850742 PMCID: PMC4135948 DOI: 10.1128/jvi.01228-14] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 05/18/2014] [Indexed: 02/06/2023] Open
Abstract
UNLABELLED Hepatitis E virus (HEV) causes both the endemic and epidemic spread of acute hepatitis in many parts of the world. HEV open reading frame 3 (ORF3) encodes a 13-kDa multifunctional protein (vp13) that is essential for HEV infection of animals. The exact role of vp13 in HEV infection remains unclear. In this study, vp13 was found to enhance interferon (IFN) production induced by poly(I · C), a synthetic analog of double-stranded RNA. Poly(I · C) treatment induced a higher level of IFN-β mRNA in HeLa cells stably expressing vp13 than in control cells. Using a luciferase reporter construct driven by the IFN-β promoter, we demonstrated that vp13 enhanced retinoic acid-inducible gene I (RIG-I)-dependent luciferase expression. This enhancement was found to be due to both an increased level of RIG-I protein and its activation. The levels of both endogenous and exogenous RIG-I were increased by vp13 by extension of the half-life of RIG-I. Additionally, vp13 interacts with the RIG-I N-terminal domain and enhances its K63-linked ubiquitination, which is essential for RIG-I activation. Analysis of vp13 deletion constructs suggested that the C-terminal domain of vp13 was essential for the enhancement of RIG-I signaling. In HEV-infected hepatoma cells, wild-type HEV led to a higher level of RIG-I and more poly(I · C)-induced IFN-β expression than did ORF3-null mutants. Analysis of vp13 from four HEV genotypes showed that vp13 from genotype I and III strains boosted RIG-I signaling, while vp13 from genotype II and IV strains had a minimal effect. These results indicate that vp13 enhances RIG-I signaling, which may play a role in HEV invasion. IMPORTANCE Hepatitis E virus (HEV) is a significant pathogen causing hepatitis in many parts of the world, yet it is understudied compared with other viral hepatitis pathogens. Here we found that the HEV open reading frame 3 product, vp13, enhances interferon induction stimulated by a synthetic analog of double-stranded RNA. This enhancement may play a role in HEV invasion, as vp13 is essential for HEV infection in vivo. The results of this study provide insights into virus-cell interactions during HEV infection. In addition to revealing its possible roles in HEV interference with cellular signaling, these results suggest that the second half of the vp13 sequence can be ligated into the genomes of attenuated live viruses to induce an innate immune response for better protective immunity, as well as a marker for differentiation of vaccinated animals from those infected with the corresponding wild-type viruses.
Collapse
Affiliation(s)
- Yuchen Nan
- Molecular Virology Laboratory, Virginia-Maryland Regional College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland
| | - Zexu Ma
- Molecular Virology Laboratory, Virginia-Maryland Regional College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland
| | - Rong Wang
- Molecular Virology Laboratory, Virginia-Maryland Regional College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland
| | - Ying Yu
- Molecular Virology Laboratory, Virginia-Maryland Regional College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland
| | - Harilakshmi Kannan
- Molecular Virology Laboratory, Virginia-Maryland Regional College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland
| | - Brenda Fredericksen
- Department of Cell Biology & Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland
| | - Yan-Jin Zhang
- Molecular Virology Laboratory, Virginia-Maryland Regional College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland
| |
Collapse
|
231
|
Frenz T, Graalmann L, Detje CN, Döring M, Grabski E, Scheu S, Kalinke U. Independent of Plasmacytoid Dendritic Cell (pDC) infection, pDC Triggered by Virus-Infected Cells Mount Enhanced Type I IFN Responses of Different Composition as Opposed to pDC Stimulated with Free Virus. THE JOURNAL OF IMMUNOLOGY 2014; 193:2496-503. [DOI: 10.4049/jimmunol.1400215] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
232
|
Luu K, Greenhill CJ, Majoros A, Decker T, Jenkins BJ, Mansell A. STAT1 plays a role in TLR signal transduction and inflammatory responses. Immunol Cell Biol 2014; 92:761-9. [DOI: 10.1038/icb.2014.51] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 05/18/2014] [Accepted: 05/19/2014] [Indexed: 12/18/2022]
Affiliation(s)
- Kevin Luu
- Centre for Innate Immunity and Infectious Diseases, MIMR‐PHI Institute of Medical Research, Monash UniversityClaytonVictoriaAustralia
| | - Claire J Greenhill
- Centre for Innate Immunity and Infectious Diseases, MIMR‐PHI Institute of Medical Research, Monash UniversityClaytonVictoriaAustralia
| | - Andrea Majoros
- Max F. Perutz Laboratories, Department of Genetics, Microbiology and Immunobiology, University of Vienna, Dr Bohr‐Gasse 9/4ViennaAustria
| | - Thomas Decker
- Max F. Perutz Laboratories, Department of Genetics, Microbiology and Immunobiology, University of Vienna, Dr Bohr‐Gasse 9/4ViennaAustria
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, MIMR‐PHI Institute of Medical Research, Monash UniversityClaytonVictoriaAustralia
| | - Ashley Mansell
- Centre for Innate Immunity and Infectious Diseases, MIMR‐PHI Institute of Medical Research, Monash UniversityClaytonVictoriaAustralia
| |
Collapse
|
233
|
Yang Q, Fu S, Wang J. Hepatitis C virus infection decreases the expression of Toll-like receptors 3 and 7 via upregulation of miR-758. Arch Virol 2014; 159:2997-3003. [PMID: 25008898 DOI: 10.1007/s00705-014-2167-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 06/27/2014] [Indexed: 12/22/2022]
Abstract
Chronic infection with hepatotropic viruses is the main cause of chronic liver disease and cirrhosis worldwide. Toll-like receptor 3 (TLR3) and Toll-like receptor 7 (TLR7) are pathogen-recognition receptors that are expressed on innate immune cells. They recognize viral RNA, which induces their activation, with a subsequent increase in type I interferon transcription. Hepatitis C virus (HCV) infection inhibits the expression of TLR3 and TLR7; however, the mechanism by which this occurs is unclear. MicroRNAs (miRNAs) are small RNAs that posttranscriptionally regulate gene expression. Their aberrant expression is commonly correlated with disease status, as is the case with HCV infection. Here, we found that miR-758 levels were increased in patients with HCV infection and were correlated with TLR3 and TLR7 expression levels in the patients with HCV infection, and bioinformatics analysis predicted that TLR3 and TLR7 are targets of miR-758. Therefore, we postulate that HCV may increase the level of miR-758, which inhibits the expression of TLR3 and TLR7, resulting in a loss of antiviral effect. In order to test our hypothesis, we constructed an HCV core protein expression plasmid and used it to transfect liver cells. The results showed that HCV infection increased miR-758 levels and decreased TLR3/TLR7 expression. Furthermore, using RT-PCR and luciferase reporter analysis, we found that miR-758 targets TLR3 and TLR7, with a subsequent decrease in IFNα and IFNβ production. In conclusion, our results highlight the upregulation of miR-758 expression by HCV as a novel mechanism contributing to downregulation of TLR3 and TLR7 in patients with HCV infection.
Collapse
Affiliation(s)
- Qian Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Rd., Hangzhou, 310003, People's Republic of China
| | | | | |
Collapse
|
234
|
Abstract
Respiratory infections and diseases are among the leading causes of death worldwide, and effective treatments probably require manipulating the inflammatory response to pathogenic microbes or allergens. Here, we review mechanisms controlling the production and functions of interleukin-17 (IL-17) and IL-22, cytokines that direct several aspects of lung immunity. Innate lymphocytes (γδ T cells, natural killer cells, innate lymphoid cells) are the major source of IL-17 and IL-22 during acute infections, while CD4(+) T-helper 17 (Th17) cells contribute to vaccine-induced immunity. The characterization of dendritic cell (DC) subsets has revealed their central roles in T-cell activation. CD11b(+) DCs stimulated with bacteria or fungi secrete IL-1β and IL-23, potent inducers of IL-17 and IL-22. On the other hand, recognition of viruses by plasmacytoid DCs inhibits IL-1β and IL-23 release, increasing susceptibility to bacterial superinfections. IL-17 and IL-22 primarily act on the lung epithelium, inducing antimicrobial proteins and neutrophil chemoattractants. Recent studies found that stimulation of macrophages and DCs with IL-17 also contributes to antibacterial immunity, while IL-22 promotes epithelial proliferation and repair following injury. Chronic diseases such as asthma and chronic obstructive pulmonary disease have been associated with IL-17 and IL-22 responses directed against innocuous antigens. Future studies will evaluate the therapeutic efficacy of targeting the IL-17/IL-22 pathway in pulmonary inflammation.
Collapse
Affiliation(s)
- Jeremy P. McAleer
- Richard King Mellon Foundation Institute for Pediatric Research, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224, USA
| | - Jay K. Kolls
- Richard King Mellon Foundation Institute for Pediatric Research, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224, USA
| |
Collapse
|
235
|
Murine cytomegalovirus virion-associated protein M45 mediates rapid NF-κB activation after infection. J Virol 2014; 88:9963-75. [PMID: 24942588 DOI: 10.1128/jvi.00684-14] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED Murine cytomegalovirus (MCMV) rapidly induces activation of nuclear factor κB (NF-κB) upon infection of host cells. After a transient phase of activation, the MCMV M45 protein blocks all canonical NF-κB-activating pathways by inducing the degradation of the gamma subunit of the inhibitor of κB kinase complex (IKKγ; commonly referred to as the NF-κB essential modulator [NEMO]). Here we show that the viral M45 protein also mediates rapid NF-κB activation immediately after infection. MCMV mutants lacking M45 or expressing C-terminally truncated M45 proteins induced neither NF-κB activation nor transcription of NF-κB-dependent genes within the first 3 h of infection. Rapid NF-κB activation was absent in MCMV-infected NEMO-deficient fibroblasts, indicating that activation occurs at or upstream of the IKK complex. NF-κB activation was strongly reduced in murine fibroblasts lacking receptor-interacting protein 1 (RIP1), a known M45-interacting protein, but was restored upon complementation with murine RIP1. However, the ability of M45 to interact with RIP1 and NEMO was not sufficient to induce NF-κB activation upon infection. In addition, incorporation of the M45 protein into virions was required. This was dependent on a C-terminal region of M45, which is not required for interaction with RIP1 and NEMO. We propose a model in which M45 delivered by viral particles activates NF-κB, presumably involving an interaction with RIP1 and NEMO. Later in infection, expression of M45 induces the degradation of NEMO and the shutdown of canonical NF-κB activation. IMPORTANCE Transcription factor NF-κB is an important regulator of innate and adaptive immunity. Its activation can be beneficial or detrimental for viral pathogens. Therefore, many viruses interfere with NF-κB signaling by stimulating or inhibiting the activation of this transcription factor. Cytomegaloviruses, opportunistic pathogens that cause lifelong infections in their hosts, activate NF-κB rapidly and transiently upon infection but block NF-κB signaling soon thereafter. Here we report the surprising finding that the murine cytomegalovirus protein M45, a component of viral particles, plays a dual role in NF-κB signaling. It not only blocks NF-κB signaling later in infection but also triggers the rapid activation of NF-κB immediately following virus entry into host cells. Both activation and inhibition involve M45 interaction with the cellular signaling mediators RIP1 and NEMO. Similar dual functions in NF-κB signaling are likely to be found in other viral proteins.
Collapse
|
236
|
The gammaherpesviruses Kaposi's sarcoma-associated herpesvirus and murine gammaherpesvirus 68 modulate the Toll-like receptor-induced proinflammatory cytokine response. J Virol 2014; 88:9245-59. [PMID: 24899179 DOI: 10.1128/jvi.00841-14] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED The human pathogen Kaposi's sarcoma-associated herpesvirus (KSHV), the etiological agent of Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease, establishes lifelong latency upon infection. Murine gammaherpesvirus 68 (MHV68) is a well-established model for KSHV. Toll-like receptors (TLRs) play a crucial role for the innate immune response to pathogens. Although KSHV and MHV68 are detected by TLRs, studies suggest they modulate TLR4 and TLR9 signaling, respectively. In this study, we show that in bone marrow-derived macrophages (BMDMs), MHV68 did not induce a detectable proinflammatory cytokine response. Furthermore, MHV68 abrogated the response to TLR2, -4, -7, and -9 agonists in BMDMs. Similarly to observations with MHV68, infection with KSHV efficiently inhibited TLR2 signaling in THP-1 monocytes. Using a KSHV open reading frame (ORF) library, we found that K4.2, ORF21, ORF31, and the replication and transcription activator protein (RTA)/ORF50 inhibited TLR2-dependent nuclear factor kappa B (NF-κB) activation in HEK293 TLR2-yellow fluorescent protein (YFP)- and Flag-TLR2-transfected HEK293T cells. Of the identified ORFs, RTA/ORF50 strongly downregulated TLR2 and TLR4 signaling by reducing TLR2 and TLR4 protein expression. Confocal microscopy revealed that TLR2 and TLR4 were no longer localized to the plasma membrane in cells expressing RTA/ORF50. In this study, we have shown that the gammaherpesviruses MHV68 and KSHV efficiently downmodulate TLR signaling in macrophages and have identified a novel function of RTA/ORF50 in modulation of the innate immune response. IMPORTANCE The Toll-like receptors (TLRs) are an important class of pattern recognition receptors of the innate immune system. They induce a potent proinflammatory cytokine response upon detection of a variety of pathogens. In this study, we found that the gammaherpesviruses murine gammaherpesvirus 68 (MHV68) and Kaposi's sarcoma-associated herpesvirus (KSHV) efficiently inhibit the TLR-mediated innate immune response. We further identified the KSHV-encoded replication and transcription activator protein (RTA) as a novel modulator of TLR signaling. Our data suggest that the gammaherpesviruses MHV68 and KSHV prevent activation of the innate immune response by targeting TLR signaling.
Collapse
|
237
|
Censoplano N, Epting CL, Coates BM. The Role of the Innate Immune System in Sepsis. CLINICAL PEDIATRIC EMERGENCY MEDICINE 2014. [DOI: 10.1016/j.cpem.2014.04.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
238
|
Dutra FF, Bozza MT. Heme on innate immunity and inflammation. Front Pharmacol 2014; 5:115. [PMID: 24904418 PMCID: PMC4035012 DOI: 10.3389/fphar.2014.00115] [Citation(s) in RCA: 235] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Accepted: 04/29/2014] [Indexed: 12/30/2022] Open
Abstract
Heme is an essential molecule expressed ubiquitously all through our tissues. Heme plays major functions in cellular physiology and metabolism as the prosthetic group of diverse proteins. Once released from cells and from hemeproteins free heme causes oxidative damage and inflammation, thus acting as a prototypic damage-associated molecular pattern. In this context, free heme is a critical component of the pathological process of sterile and infectious hemolytic conditions including malaria, hemolytic anemias, ischemia-reperfusion, and hemorrhage. The plasma scavenger proteins hemopexin and albumin reduce heme toxicity and are responsible for transporting free heme to intracellular compartments where it is catabolized by heme-oxygenase enzymes. Upon hemolysis or severe cellular damage the serum capacity to scavenge heme may saturate and increase free heme to sufficient amounts to cause tissue damage in various organs. The mechanism by which heme causes reactive oxygen generation, activation of cells of the innate immune system and cell death are not fully understood. Although heme can directly promote lipid peroxidation by its iron atom, heme can also induce reactive oxygen species generation and production of inflammatory mediators through the activation of selective signaling pathways. Heme activates innate immune cells such as macrophages and neutrophils through activation of innate immune receptors. The importance of these events has been demonstrated in infectious and non-infectious diseases models. In this review, we will discuss the mechanisms behind heme-induced cytotoxicity and inflammation and the consequences of these events on different tissues and diseases.
Collapse
Affiliation(s)
- Fabianno F. Dutra
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Marcelo T. Bozza
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| |
Collapse
|
239
|
Shaik-Dasthagirisaheb YB, Huang N, Weinberg EO, Shen SS, Genco CA, Gibson FC. Aging and contribution of MyD88 and TRIF to expression of TLR pathway-associated genes following stimulation with Porphyromonas gingivalis. J Periodontal Res 2014; 50:89-102. [PMID: 24862405 DOI: 10.1111/jre.12185] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2014] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND OBJECTIVE Periodontal disease is a highly complex chronic inflammatory disease of the oral cavity. Multiple factors influence periodontal disease, including socio-economic status, genetics and age; however, inflammation elicited by the presence of specific bacteria in the subgingival space is thought to drive the majority of soft- and hard-tissue destruction. Porphyromonas gingivalis is closely associated with periodontal disease. Toll-like receptors (TLRs) and their intracellular signaling pathways play roles in the host response to P. gingivalis. The focus of the current study was to use microarray analysis to define the contributions of the TLR adaptor molecules myeloid differentiation factor 88 (MyD88) and Toll/interleukin-1 receptor domain-containing adaptor inducing interferon-beta (TRIF), and aging, on the expression of TLR pathway-associated mRNAs in response to P. gingivalis. MATERIAL AND METHODS Bone marrow-derived macrophages (BMØ) from wild-type (Wt), MyD88 knockout (MyD88-KO) and Trif(Lps2) [i.e. containing a point mutation in the lipopolysaccharide 2 (Lps2) gene rendering the Toll/interleukin (IL)-1 receptor domain-containing adaptor inducing interferon-beta (TRIF) protein nonfunctional] mice, at 2-and 12-mo of age, were cultured with P. gingivalis. Expression of genes in BMØ cultured with P. gingivalis was determined in comparison with expression of genes in BMØ cultured in medium only. RESULTS Using, as criteria, a twofold increase or decrease in mRNA expression, differential expression of 32 genes was observed when Wt BMØ from 2-mo-old mice were cultured with P. gingivalis compared with the medium-only control. When compared with 2-mo-old Wt mice, 21 and 12 genes were differentially expressed (p < 0.05) as a result of the mutations in MyD88 or TRIF, respectively. The expression of five genes was significantly (p < 0.05) reduced in Wt BMØ from 12-mo-old mice compared with those from 2-mo-old mice following culture with P. gingivalis. Age also influenced the expression of genes in MyD88-KO and Trif(Lps2) mice challenged with P. gingivalis. CONCLUSIONS Our results indicate that P. gingivalis induces differential expression of TLR pathway-associated genes, and both MyD88 and TRIF play roles in the expression of these genes. Age also played a role in the expression of TLR-associated genes following stimulation of BMØ with P. gingivalis.
Collapse
Affiliation(s)
- Y B Shaik-Dasthagirisaheb
- Section of Infectious Diseases, Department of Medicine, Boston University Medical Center, Boston, MA, USA
| | | | | | | | | | | |
Collapse
|
240
|
Siggs OM. Dissecting mammalian immunity through mutation. Immunol Cell Biol 2014; 92:392-9. [PMID: 24518983 PMCID: PMC4038135 DOI: 10.1038/icb.2014.8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 01/15/2014] [Accepted: 01/15/2014] [Indexed: 12/27/2022]
Abstract
Although mutation and natural selection have given rise to our immune system, a well-placed mutation can also cripple it, and within an expanding population we are recognizing more and more cases of single-gene mutations that compromise immunity. These mutations are an ideal tool for understanding human immunology, and there are more ways than ever to measure their physiological effects. There are also more ways to create mutations in the laboratory, and to use these resources to systematically define the function of every gene in our genome. This review focuses on the discovery and creation of mutations in the context of mammalian immunity, with an emphasis on the use of genome-wide chemical and CRISPR/Cas9 mutagenesis to reveal gene function.
Collapse
Affiliation(s)
- Owen M Siggs
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| |
Collapse
|
241
|
Dalod M, Chelbi R, Malissen B, Lawrence T. Dendritic cell maturation: functional specialization through signaling specificity and transcriptional programming. EMBO J 2014; 33:1104-16. [PMID: 24737868 DOI: 10.1002/embj.201488027] [Citation(s) in RCA: 309] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Dendritic cells (DC) are key regulators of both protective immune responses and tolerance to self-antigens. Soon after their discovery in lymphoid tissues by Steinman and Cohn, as cells with the unique ability to prime naïve antigen-specific T cells, it was realized that DC can exist in at least two distinctive states characterized by morphological, phenotypic and functional changes-this led to the description of DC maturation. It is now well appreciated that there are several subsets of DC in both lymphoid and non-lymphoid tissues of mammals, and these cells show remarkable functional specialization and specificity in their roles in tolerance and immunity. This review will focus on the specific characteristics of DC subsets and how their functional specialization may be regulated by distinctive gene expression programs and signaling responses in both steady-state and in the context of inflammation. In particular, we will highlight the common and distinctive genes and signaling pathways that are associated with the functional maturation of DC subsets.
Collapse
Affiliation(s)
- Marc Dalod
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University UM2, Marseille, France Institut National de la Santé et de la Recherche Médicale (INSERM) U1104, Marseille, France Centre National de la Recherche Scientifique (CNRS), UMR7280, Marseille, France
| | - Rabie Chelbi
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University UM2, Marseille, France Institut National de la Santé et de la Recherche Médicale (INSERM) U1104, Marseille, France Centre National de la Recherche Scientifique (CNRS), UMR7280, Marseille, France
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University UM2, Marseille, France Institut National de la Santé et de la Recherche Médicale (INSERM) U1104, Marseille, France Centre National de la Recherche Scientifique (CNRS), UMR7280, Marseille, France
| | - Toby Lawrence
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University UM2, Marseille, France Institut National de la Santé et de la Recherche Médicale (INSERM) U1104, Marseille, France Centre National de la Recherche Scientifique (CNRS), UMR7280, Marseille, France
| |
Collapse
|
242
|
Rowley SM, Kuriakose T, Dockery LM, Tran-Nguyen T, Gingerich AD, Wei L, Watford WT. Tumor progression locus 2 (Tpl2) kinase promotes chemokine receptor expression and macrophage migration during acute inflammation. J Biol Chem 2014; 289:15788-97. [PMID: 24713702 DOI: 10.1074/jbc.m114.559344] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In autoimmune diseases, the accumulation of activated leukocytes correlates with inflammation and disease progression, and, therefore, the disruption of leukocyte trafficking is an active area of research. The serine/threonine protein kinase Tpl2 (MAP3K8) regulates leukocyte inflammatory responses and is also being investigated for therapeutic inhibition during autoimmunity. Here we addressed the contribution of Tpl2 to the regulation of macrophage chemokine receptor expression and migration in vivo using a mouse model of Tpl2 ablation. LPS stimulation of bone marrow-derived macrophages induced early CCR1 chemokine receptor expression but repressed CCR2 and CCR5 expression. Notably, early induction of CCR1 expression by LPS was dependent upon a signaling pathway involving Tpl2, PI3K, and ERK. On the contrary, Tpl2 was required to maintain the basal expression of CCR2 and CCR5 as well as to stabilize CCR5 mRNA expression. Consistent with impairments in chemokine receptor expression, tpl2(-/-) macrophages were defective in trafficking to the peritoneal cavity following thioglycollate-induced inflammation. Overall, this study demonstrates a Tpl2-dependent mechanism for macrophage expression of select chemokine receptors and provides further insight into how Tpl2 inhibition may be used therapeutically to disrupt inflammatory networks in vivo.
Collapse
Affiliation(s)
- Sean M Rowley
- From the Department of Infectious Diseases, The University of Georgia, College of Veterinary Medicine, Athens, Georgia 30602 and
| | - Teneema Kuriakose
- From the Department of Infectious Diseases, The University of Georgia, College of Veterinary Medicine, Athens, Georgia 30602 and
| | - Lee M Dockery
- From the Department of Infectious Diseases, The University of Georgia, College of Veterinary Medicine, Athens, Georgia 30602 and
| | - Thi Tran-Nguyen
- From the Department of Infectious Diseases, The University of Georgia, College of Veterinary Medicine, Athens, Georgia 30602 and
| | - Aaron D Gingerich
- From the Department of Infectious Diseases, The University of Georgia, College of Veterinary Medicine, Athens, Georgia 30602 and
| | - Lai Wei
- the Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China
| | - Wendy T Watford
- From the Department of Infectious Diseases, The University of Georgia, College of Veterinary Medicine, Athens, Georgia 30602 and
| |
Collapse
|
243
|
Samarajiwa SA, Mangan NE, Hardy MP, Najdovska M, Dubach D, Braniff SJ, Owczarek CM, Hertzog PJ. Soluble IFN receptor potentiates in vivo type I IFN signaling and exacerbates TLR4-mediated septic shock. THE JOURNAL OF IMMUNOLOGY 2014; 192:4425-35. [PMID: 24696235 DOI: 10.4049/jimmunol.1302388] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Circulating levels of a soluble type I IFNR are elevated in diseases, such as chronic inflammation, infections, and cancer, but whether it functions as an antagonist, agonist, or transporter is unknown. In this study, we elucidate the in vivo importance of the soluble type I IFNAR, soluble (s)IFNAR2a, which is generated by alternative splicing of the Ifnar2 gene. A transgenic mouse model was established to mimic the 10-15-fold elevated expression of sIFNAR2a observed in some human diseases. We generated transgenic mouse lines, designated SolOX, in which the transgene mRNA and protein-expression patterns mirrored the expression patterns of the endogenous gene. SolOX were demonstrated to be more susceptible to LPS-mediated septic shock, a disease model in which type I IFN plays a crucial role. This effect was independent of "classical" proinflammatory cytokines, such as TNF-α and IL-6, whose levels were unchanged. Because the increased levels of sIFNAR2a did not affect the kinetics of the increased interferonemia, this soluble receptor does not potentiate its ligand signaling by improving IFN pharmacokinetics. Mechanistically, increased levels of sIFNAR2a are likely to facilitate IFN signaling, as demonstrated in spleen cells overexpressing sIFNAR2a, which displayed quicker, higher, and more sustained activation of STAT1 and STAT3. Thus, the soluble IFNR is an important agonist of endogenous IFN actions in pathophysiological processes and also is likely to modulate the therapeutic efficacy of clinically administered IFNs.
Collapse
Affiliation(s)
- Shamith A Samarajiwa
- Centre for Innate Immunity and Infectious Diseases, Monash Institute of Medical Research, Monash University, Clayton, Victoria 3168, Australia
| | | | | | | | | | | | | | | |
Collapse
|
244
|
Moresco EMY, Li X, Beutler B. Going forward with genetics: recent technological advances and forward genetics in mice. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 182:1462-73. [PMID: 23608223 DOI: 10.1016/j.ajpath.2013.02.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 02/05/2013] [Indexed: 12/24/2022]
Abstract
Forward genetic analysis is an unbiased approach for identifying genes essential to defined biological phenomena. When applied to mice, it is one of the most powerful methods to facilitate understanding of the genetic basis of human biology and disease. The speed at which disease-causing mutations can be identified in mutagenized mice has been markedly increased by recent advances in DNA sequencing technology. Creating and analyzing mutant phenotypes may therefore become rate-limiting in forward genetic experimentation. We review the forward genetic approach and its future in the context of recent technological advances, in particular massively parallel DNA sequencing, induced pluripotent stem cells, and haploid embryonic stem cells.
Collapse
Affiliation(s)
- Eva Marie Y Moresco
- Center for Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75235-8505, USA
| | | | | |
Collapse
|
245
|
Fleige H, Ravens S, Moschovakis GL, Bölter J, Willenzon S, Sutter G, Häussler S, Kalinke U, Prinz I, Förster R. IL-17-induced CXCL12 recruits B cells and induces follicle formation in BALT in the absence of differentiated FDCs. ACTA ACUST UNITED AC 2014; 211:643-51. [PMID: 24663215 PMCID: PMC3978277 DOI: 10.1084/jem.20131737] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The requirements for BALT formation are pathogen-dependent and, in the absence of FDC maturation, IL-17 can drive BALT formation via CXCL12 B cell recruitment. Ectopic lymphoid tissue, such as bronchus-associated lymphoid tissue (BALT) in the lung, develops spontaneously at sites of chronic inflammation or during infection. The molecular mechanisms underlying the neogenesis of such tertiary lymphoid tissue are still poorly understood. We show that the type of inflammation-inducing pathogen determines which key factors are required for the formation and maturation of BALT. Thus, a single intranasal administration of the poxvirus modified vaccinia virus Ankara (MVA) is sufficient to induce highly organized BALT with densely packed B cell follicles containing a network of CXCL13-expressing follicular DCs (FDCs), as well as CXCL12-producing follicular stromal cells. In contrast, mice treated with P. aeruginosa (P.a.) develop BALT but B cell follicles lack FDCs while still harboring CXCL12-positive follicular stromal cells. Furthermore, in IL-17–deficient mice, P.a.-induced BALT largely lacks B cells as well as CXCL12-expressing stromal cells, and only loose infiltrates of T cells are present. We show that Toll-like receptor pathways are required for BALT induction by P.a., but not MVA, and provide evidence that IL-17 drives the differentiation of lung stroma toward podoplanin-positive CXCL12-expressing cells that allow follicle formation even in the absence of FDCs. Taken together, our results identify distinct pathogen-dependent induction and maturation pathways for BALT formation.
Collapse
Affiliation(s)
- Henrike Fleige
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
246
|
Huang N, Gibson FC. Immuno-pathogenesis of Periodontal Disease: Current and Emerging Paradigms. ACTA ACUST UNITED AC 2014; 1:124-132. [PMID: 24839590 DOI: 10.1007/s40496-014-0017-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Periodontal disease (PD) is a highly complex disease involving many factors; however, two principal facets central to initiation and progression of the majority of PD are the composition of the microbes in the sub-gingival plaque, and the host immune response to these organisms. Numerous studies point to the complexity of PD, and to the fact that despite innate and adaptive immune activation, and resultant inflammation, our immune response fails to cure disease. Stunning new findings have begun to clarify several complexities of the host-pathogen interaction of PD pointing to key roles for microbial dysboisis and immune imbalance in the pathogenesis of disease. Furthermore, these investigations have identified novel translational opportunities to intercede in PD treatment. In this review we will highlight a select few recent findings in innate and adaptive immunity, and host pathogen interactions of PD at a micro-environmental level that may have profound impact on PD progression.
Collapse
Affiliation(s)
- Nasi Huang
- Section of Infectious Diseases, Department of Medicine, Boston University School of Medicine, Boston, MA, 02118
| | - Frank C Gibson
- Section of Infectious Diseases, Department of Medicine, Boston University School of Medicine, Boston, MA, 02118
| |
Collapse
|
247
|
Nguyen VP, Chen J, Petrus MN, Goldman CK, Kruhlak MJ, Bamford RN, Waldmann TA. A new domain in the Toll/IL-1R domain-containing adaptor inducing interferon-β factor protein amino terminus is important for tumor necrosis factor-α receptor-associated factor 3 association, protein stabilization and interferon signaling. J Innate Immun 2014; 6:377-93. [PMID: 24577058 DOI: 10.1159/000356408] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 10/01/2013] [Indexed: 01/16/2023] Open
Abstract
Toll/IL-1R domain-containing adaptor inducing interferon-β (IFN-β) factor (TRIF) is a key adaptor for Toll-like receptor (TLR) 3 and TLR4 signaling. Using a novel cDNA isolate encoding a TRIF protein with a 21-residue deletion (Δ160-181) from its amino-terminal half, we investigated the impact of this deletion on TRIF functions. Transfection studies consistently showed higher expression levels of the (Δ160-181) TRIF compared to wild-type (wt) TRIF, an effect unrelated to apoptosis, cell lines or plasmid amplification. Colocalization of wt and (Δ160-181) TRIF proteins led to a dramatic reduction of their respective expressions, suggesting that wt/(Δ160-181) TRIF heterocomplexes are targeted for degradation. We demonstrated that wt TRIF associates with tumor necrosis factor-α receptor-associated factor 3 (TRAF3) better than (Δ160-181) TRIF, culminating in its greater ubiquitination and proteolysis. This explains, in part, the differential expression levels of the two TRIF proteins. Despite higher expression levels in transfected cells, (Δ160-181) TRIF inefficiently transactivated the IFN pathway, whereas the nuclear factor-κB (NF-κB) pathway activation remained similar to that by wt TRIF. In coexpression studies, (Δ160-181) TRIF marginally contributed to the IFN pathway activation, but still enhanced NF-κB signaling with wt TRIF. Therefore, this 21 amino acid sequence is crucial for TRAF3 association, modulation of TRIF stability and activation of the IFN pathway.
Collapse
Affiliation(s)
- Vinh-Phuc Nguyen
- Metabolism Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Md., USA
| | | | | | | | | | | | | |
Collapse
|
248
|
Beiting DP, Peixoto L, Akopyants NS, Beverley SM, Wherry EJ, Christian DA, Hunter CA, Brodsky IE, Roos DS. Differential induction of TLR3-dependent innate immune signaling by closely related parasite species. PLoS One 2014; 9:e88398. [PMID: 24505488 PMCID: PMC3914978 DOI: 10.1371/journal.pone.0088398] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 12/31/2013] [Indexed: 12/20/2022] Open
Abstract
The closely related protozoan parasites Toxoplasma gondii and Neospora caninum display similar life cycles, subcellular ultrastructure, invasion mechanisms, metabolic pathways, and genome organization, but differ in their host range and disease pathogenesis. Type II (γ) interferon has long been known to be the major mediator of innate and adaptive immunity to Toxoplasma infection, but genome-wide expression profiling of infected host cells indicates that Neospora is a potent activator of the type I (α/β) interferon pathways typically associated with antiviral responses. Infection of macrophages from mice with targeted deletions in various innate sensing genes demonstrates that host responses to Neospora are dependent on the toll-like receptor Tlr3 and the adapter protein Trif. Consistent with this observation, RNA from Neospora elicits TLR3-dependent type I interferon responses when targeted to the host endo-lysosomal system. Although live Toxoplasma fail to induce type I interferon, heat-killed parasites do trigger this response, albeit much weaker than Neospora, and co-infection studies reveal that T. gondii actively suppresses the production of type I interferon. These findings reveal that eukaryotic pathogens can be potent inducers of type I interferon and that related parasite species interact with this pathway in distinct ways.
Collapse
Affiliation(s)
- Daniel P. Beiting
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| | - Lucia Peixoto
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Natalia S. Akopyants
- Department of Molecular Microbiology, Washington University, St. Louis, Missouri, United States of America
| | - Stephen M. Beverley
- Department of Molecular Microbiology, Washington University, St. Louis, Missouri, United States of America
| | - E. John Wherry
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - David A. Christian
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Christopher A. Hunter
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Igor E. Brodsky
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - David S. Roos
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
249
|
Han JE, Wui SR, Kim KS, Cho YJ, Cho WJ, Lee NG. Characterization of the structure and immunostimulatory activity of a vaccine adjuvant, de-O-acylated lipooligosaccharide. PLoS One 2014; 9:e85838. [PMID: 24465739 PMCID: PMC3899070 DOI: 10.1371/journal.pone.0085838] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 12/02/2013] [Indexed: 12/20/2022] Open
Abstract
Lipopolysaccharide (LPS) is a major component of the outer membrane of Gram-negative bacteria. LPS elicits strong immunopathological responses during bacterial infection, and the lipid A moiety of LPS is responsible for this immunostimulatory activity. Lipid A exerts its biological activity by sending signals via TLR4 present on immune cells, and TLR4 agonists have been a target for vaccine adjuvant. Previously, we demonstrated an adjuvant activity of deacylated lipooligosaccharide (dLOS) to viral and bacterial antigens. In this study, we characterized the chemical structure of dLOS and evaluated its immunostimulatory activity on mouse and human immune cells in comparison with monophosphoryl lipid A (MPL). dLOS consists of a core oligosaccharide lacking the terminal glucose residue, a glucosamine disaccharide with two phosphate groups, and two N-linked acyl groups. dLOS was similar to MPL in induction of cytokine production in mouse peritoneal macrophages, but was a more potent activator in human monocytes and dendritic cells (DCs). Results of an analysis of allogeneic T cell responses revealed that dLOS induces Th1, Th2, and Th17-type immune responses in a dose-dependent manner. The immunostimulatory activities of dLOS were completely abrogated in TLR4−/− mice, which confirms its TLR4-dependency. These results suggest that in the presence of the core oligosaccharide, O-linked acyl groups of LPS are dispensable for activating the TLR4 signaling pathway. dLOS did not cause any pathological effects or death at 0.25, 0.5, or 1 mg per kg body weight in mice in the acute toxicity tests. This result suggests that dLOS has a low toxicity. dLOS should be considered for further development as a safe and effective adjuvant for human vaccines.
Collapse
MESH Headings
- Acylation
- Adjuvants, Immunologic/chemistry
- Adjuvants, Immunologic/pharmacology
- Animals
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cells, Cultured
- Cytokines/blood
- Cytokines/immunology
- Cytokines/metabolism
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dose-Response Relationship, Drug
- Female
- Flow Cytometry
- Humans
- Immunoblotting
- Lipopolysaccharides/chemistry
- Lipopolysaccharides/immunology
- Lipopolysaccharides/pharmacology
- Macrophages, Peritoneal/drug effects
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Molecular Structure
- Monocytes/drug effects
- Monocytes/immunology
- Monocytes/metabolism
- Toll-Like Receptor 4/deficiency
- Toll-Like Receptor 4/genetics
- Toll-Like Receptor 4/immunology
- Vaccines/immunology
Collapse
Affiliation(s)
- Ji Eun Han
- Department of Bioscience & Biotechnology, College of Bioscience, Sejong University, Seoul, Republic of Korea
| | - Seo Ri Wui
- Department of Bioscience & Biotechnology, College of Bioscience, Sejong University, Seoul, Republic of Korea
| | - Kwang Sung Kim
- Research & Development Center, EyeGene, Seoul, Republic of Korea
| | - Yang Je Cho
- Research & Development Center, EyeGene, Seoul, Republic of Korea
| | - Wan Je Cho
- Yonsei University Gangnam Severance Hospital, Seoul, Republic of Korea
| | - Na Gyong Lee
- Department of Bioscience & Biotechnology, College of Bioscience, Sejong University, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
250
|
Hyaluronan is not a ligand but a regulator of toll-like receptor signaling in mesangial cells: role of extracellular matrix in innate immunity. ISRN NEPHROLOGY 2014; 2014:714081. [PMID: 24967246 PMCID: PMC4045461 DOI: 10.1155/2014/714081] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 10/30/2013] [Indexed: 11/18/2022]
Abstract
Glomerular mesangial cells (MC), like most cell types secrete hyaluronan (HA), which attached to the cell surface via CD44, is the backbone of a hydrophilic gel matrix around these cells. Reduced extracellular matrix thickness and viscosity result from HA cleavage during inflammation. HA fragments were reported to trigger innate immunity via Toll-like receptor-(TLR-) 2 and/or TLR4 in immune cells. We questioned whether HA fragments also regulate the immunostimulatory capacity of smooth muscle cell-like MC. LPS (TLR4-ligand) and PAM3CysSK4 (TLR2-ligand) induced IL-6 secretion in MC; highly purified endotoxin-free HA < 3000 Da up to 50 μg/mL did not. Bovine-testis-hyaluronidase from was used to digest MC-HA into HA fragments of different size directly in the cell culture. Resultant HA fragments did not activate TLR4-deficient MC, while TLR2-deficient MC responded to LPS-contamination of hyaluronidase, not to produced HA fragments. Hyaluronidase increased the stimulatory effect of TLR2-/-3/-5 ligands on their TLR-receptors in TLR4-deficient MC, excluding any effect by LPS-contamination. Supplemented heparin suppressed every stimulatory effect in a dose-dependent manner. We conclude that the glycosaminoglycan HA creates a pericellular jelly barrier, which covers surface receptors like the TLRs. Barrier-thickness and viscosity balanced by HA-synthesis and degradation and the amount of HA-receptors on the cell surface regulate innate immunity via the accessibility of the receptors.
Collapse
|