151
|
Buschor S, Cuenca M, Uster SS, Schären OP, Balmer ML, Terrazos MA, Schürch CM, Hapfelmeier S. Innate immunity restricts Citrobacter rodentium A/E pathogenesis initiation to an early window of opportunity. PLoS Pathog 2017; 13:e1006476. [PMID: 28662171 PMCID: PMC5507559 DOI: 10.1371/journal.ppat.1006476] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/12/2017] [Accepted: 06/16/2017] [Indexed: 11/30/2022] Open
Abstract
Citrobacter rodentium infection is a mouse model for the important human diarrheal infection caused by enteropathogenic E. coli (EPEC). The pathogenesis of both species is very similar and depends on their unique ability to form intimately epithelium-adherent microcolonies, also known as "attachment/effacement" (A/E) lesions. These microcolonies must be dynamic and able to self-renew by continuous re-infection of the rapidly regenerating epithelium. It is unknown whether sustained epithelial A/E lesion pathogenesis is achieved through re-infection by planktonic bacteria from the luminal compartment or local spread of sessile bacteria without a planktonic phase. Focusing on the earliest events as C. rodentium becomes established, we show here that all colonic epithelial A/E microcolonies are clonal bacterial populations, and thus depend on local clonal growth to persist. In wild-type mice, microcolonies are established exclusively within the first 18 hours of infection. These early events shape the ongoing intestinal geography and severity of infection despite the continuous presence of phenotypically virulent luminal bacteria. Mechanistically, induced resistance to A/E lesion de-novo formation is mediated by TLR-MyD88/Trif-dependent signaling and is induced specifically by virulent C. rodentium in a virulence gene-dependent manner. Our data demonstrate that the establishment phase of C. rodentium pathogenesis in vivo is restricted to a very short window of opportunity that determines both disease geography and severity.
Collapse
Affiliation(s)
- Stefanie Buschor
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
- Graduate School GCB, University of Bern, Bern, Switzerland
| | - Miguelangel Cuenca
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
- Graduate School GCB, University of Bern, Bern, Switzerland
| | - Stephanie S. Uster
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Olivier P. Schären
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
- Graduate School GCB, University of Bern, Bern, Switzerland
| | - Maria L. Balmer
- Department of Biomedicine, Immunobiology, University of Basel, Basel, Switzerland
| | - Miguel A. Terrazos
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | | | | |
Collapse
|
152
|
The murine cytomegalovirus M35 protein antagonizes type I IFN induction downstream of pattern recognition receptors by targeting NF-κB mediated transcription. PLoS Pathog 2017; 13:e1006382. [PMID: 28542326 PMCID: PMC5444856 DOI: 10.1371/journal.ppat.1006382] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 04/26/2017] [Indexed: 12/25/2022] Open
Abstract
The type I interferon (IFN) response is imperative for the establishment of the early antiviral immune response. Here we report the identification of the first type I IFN antagonist encoded by murine cytomegalovirus (MCMV) that shuts down signaling following pattern recognition receptor (PRR) sensing. Screening of an MCMV open reading frame (ORF) library identified M35 as a novel and strong negative modulator of IFNβ promoter induction following activation of both RNA and DNA cytoplasmic PRR. Additionally, M35 inhibits the proinflammatory cytokine response downstream of Toll-like receptors (TLR). Using a series of luciferase-based reporters with specific transcription factor binding sites, we determined that M35 targets NF-κB-, but not IRF-mediated, transcription. Expression of M35 upon retroviral transduction of immortalized bone marrow-derived macrophages (iBMDM) led to reduced IFNβ transcription and secretion upon activation of stimulator of IFN genes (STING)-dependent signaling. On the other hand, M35 does not antagonize interferon-stimulated gene (ISG) 56 promoter induction or ISG transcription upon exogenous stimulation of the type I IFN receptor (IFNAR). M35 is present in the viral particle and, upon MCMV infection of fibroblasts, is immediately shuttled to the nucleus where it exerts its immunomodulatory effects. Deletion of M35 from the MCMV genome and hence from the viral particle resulted in elevated type I IFN transcription and secretion in vitro and in vivo. In the absence of M35, lower viral titers are observed during acute infection of the host, and productive infection in the salivary glands was not detected. In conclusion, the M35 protein is released by MCMV immediately upon infection in order to deftly inhibit the antiviral type I IFN response by targeting NF-κB-mediated transcription. The identification of this novel viral protein reinforces the importance of timely countermeasures in the complex relationship between virus and host. The herpesvirus cytomegalovirus can cause severe morbidity in immunosuppressed people and poses a much greater global problem in the context of congenital infections than the Zika virus. To establish infection, cytomegalovirus needs to modulate the antiviral immune response of its host. One of the first lines of defense against viral infections is the type I interferon response which is activated by cellular sensors called pattern recognition receptors. These receptors sense viral entry and rapidly induce the transcription of type I interferons, which are instrumental for the induction of an antiviral state in infected and surrounding cells. We have identified the first viral protein encoded by murine cytomegalovirus, the M35 protein, that counteracts type I interferon transcription downstream of multiple pattern recognition receptors. We found that this viral countermeasure occurs shortly after viral entry into the host cell, as M35 is delivered with the viral particle. M35 then localizes to the nucleus where it modulates NF-κB-mediated transcription. In vivo, murine cytomegalovirus deficient of the M35 protein replicates to lower levels in spleen and liver and cannot establish a productive infection in the salivary glands, which is a key site of viral transmission, highlighting the important role of M35 for the establishment of infection. Our study provides novel insights into the complex interaction between cytomegalovirus and the innate immune response of its host.
Collapse
|
153
|
Gentle IE, McHenry KT, Weber A, Metz A, Kretz O, Porter D, Häcker G. TIR-domain-containing adapter-inducing interferon-β (TRIF) forms filamentous structures, whose pro-apoptotic signalling is terminated by autophagy. FEBS J 2017; 284:1987-2003. [PMID: 28453927 DOI: 10.1111/febs.14091] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 03/14/2017] [Accepted: 04/25/2017] [Indexed: 12/15/2022]
Abstract
The formation of amyloid-like protein structures has recently emerged as a feature in signal transduction, particularly in innate immunity. These structures appear to depend on defined domains for their formation but likely also require dedicated ways to terminate signalling. We, here, define the innate immunity protein/Toll-like receptor adaptor TIR-domain-containing adapter-inducing interferon-β (TRIF) as a novel platform of fibril formation and probe signal initiation through TRIF as well as its termination in Toll-like receptor 3 (TLR3)-stimulated melanoma cells. A main signalling pathway triggered by TLR3 caused apoptosis, which was controlled by inhibitor of apoptosis proteins and was dependent on RIPK1 and independent of TNF. Using correlative electron/fluorescence microscopy, we visualised fibrillar structures formed through both Toll/interleukin-1 receptor and RIP homotypic interacting motif regions of TRIF. We provide evidence that these fibrillary structures are active signalling platforms whose activity is terminated by autophagy. TRIF-signalling enhanced autophagy, and fibrillary structures were partly contained within autophagosomes. Inhibition of autophagy increased levels of pro-apoptotic TRIF complexes, leading to the accumulation of active caspase-8 and enhanced apoptosis while stimulation of autophagy reduced TRIF-dependent death. We conclude that pro-death signals through TRIF are regulated by autophagy and propose that pro-apoptotic signalling through TRIF/RIPK1/caspase-8 occurs in fibrillary platforms.
Collapse
Affiliation(s)
- Ian E Gentle
- Faculty of Medicine, Institute for Medical Microbiology and Hygiene, Medical Center - University of Freiburg, University of Freiburg, Germany
| | - Kevin T McHenry
- Novartis Institutes for Biomedical Research, Inc., Cambridge, MA, USA
| | - Arnim Weber
- Faculty of Medicine, Institute for Medical Microbiology and Hygiene, Medical Center - University of Freiburg, University of Freiburg, Germany
| | - Arlena Metz
- Faculty of Medicine, Institute for Medical Microbiology and Hygiene, Medical Center - University of Freiburg, University of Freiburg, Germany
| | - Oliver Kretz
- Renal Division, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Germany.,Department of Neuroanatomy, University Freiburg, Germany
| | - Dale Porter
- Novartis Institutes for Biomedical Research, Inc., Cambridge, MA, USA
| | - Georg Häcker
- Faculty of Medicine, Institute for Medical Microbiology and Hygiene, Medical Center - University of Freiburg, University of Freiburg, Germany
| |
Collapse
|
154
|
Zhou Y, Wang J, Yang W, Qi X, Lan L, Luo L, Yin Z. Bergapten prevents lipopolysaccharide-induced inflammation in RAW264.7 cells through suppressing JAK/STAT activation and ROS production and increases the survival rate of mice after LPS challenge. Int Immunopharmacol 2017; 48:159-168. [PMID: 28511114 DOI: 10.1016/j.intimp.2017.04.026] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 04/09/2017] [Accepted: 04/24/2017] [Indexed: 01/25/2023]
Abstract
Bergapten (BG) is a cumarine-derivate compound in many medicinal plants. Here, in vitro and in vivo experimental results indicated that BG possesses anti-inflammatory properties, Based on this, we further investigated the precise molecular mechanisms of BG in LPS-stimulated inflammation response. Studies revealed that BG inhibited LPS-stimulated productions of TNF-α, IL-1β, IL-6, PGE2 and NO as well as the expression of iNOS and COX-2, and at the same time, it increased LPS-induced release of IL-10 in a dose-dependent manner in RAW264.7 cells. Mechanistically, BG suppressed the activations of JAK/STAT, but not that of MAPKs and NF-κB. In addition, BG, as an antioxidant, prevented the accumulation of ROS, which further exerted its anti-inflammatory function. In vivo researches revealed that BG decreased LPS-induced mortality in mice. In conclusions, BG may be a potential candidate for inflammation therapy via inhibiting JAK/STAT activation and ROS production in RAW264.7 cells.
Collapse
Affiliation(s)
- Yi Zhou
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, Jiangsu, PR China
| | - Jing Wang
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, Jiangsu, PR China
| | - Weidong Yang
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, Jiangsu, PR China
| | - Xiaowen Qi
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, Jiangsu, PR China
| | - Lei Lan
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, Jiangsu, PR China
| | - Lan Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, Jiangsu, PR China.
| | - Zhimin Yin
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, Jiangsu, PR China.
| |
Collapse
|
155
|
Li X, Zhang P, Jiang X, Du H, Yang C, Zhang Z, Men S, Zhang Z, Jiang W, Wang H. Differences in expression of genes in the MyD88 and TRIF signalling pathways and methylation of TLR4 and TRIF in Tibetan chickens and DaHeng S03 chickens infected with Salmonella enterica serovar enteritidis. Vet Immunol Immunopathol 2017; 189:28-35. [PMID: 28669384 DOI: 10.1016/j.vetimm.2017.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 05/02/2017] [Accepted: 05/08/2017] [Indexed: 12/18/2022]
Abstract
Salmonella enterica serovar (S. enteritidis) is a pathogenic bacterium that can cause symptoms of food poisoning, leading to death of poultry, resulting in serious economic losses. The MyD88 and TRIF signalling pathways play important roles in activating innate and adaptive immunity in chickens infected with S. enteritidis. The objective of the present study was to characterize in vivo mRNA expressions, protein levels and methylation levels of genes in the above two pathways in both Tibetan chickens and DaHeng S03 chickens infected with S. enteritidis. MyD88-dependent and TRIF-dependent signalling pathway were activated by infection, and the MyD88 signalling pathway induced cytokines LITAF and IL-8 played important roles in fighting against the S. enteritidis infection in vivo. The TLR4 methylation might alter expression of genes involved in the MyD88 signalling pathway, and thus different breeds of chickens might show differences in susceptibility to the S. enteritidis. The increased expression of INF β was activated by S. enteritidis, but its expressions were different in levels of mRNA and protein in Tibetan chickens and DaHeng chickens, suggesting its functions on the resistance to S. enteritidis infection in chickens. This study contributes to the understanding of two pathways activated in response to S. enteritidis infection, and gives indications on the mechanisms underlying resistance of Tibetan chickens and DaHeng chickens to S. enteritidis.
Collapse
Affiliation(s)
- Xiaocheng Li
- College of Life Science, Sichuan University, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, "985 Project" Science Innovative Platform for Resource and Environment Protection of Southwestern, Chengdu 610065, China; Sichuan Academy of Animal Sciences, Animal Breeding and Genetics Key Laboratory of Sichuan Province, Chengdu 610066, China
| | - Peng Zhang
- College of Life Science, Sichuan University, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, "985 Project" Science Innovative Platform for Resource and Environment Protection of Southwestern, Chengdu 610065, China
| | - Xiaosong Jiang
- Sichuan Academy of Animal Sciences, Animal Breeding and Genetics Key Laboratory of Sichuan Province, Chengdu 610066, China
| | - Huarui Du
- Sichuan Academy of Animal Sciences, Animal Breeding and Genetics Key Laboratory of Sichuan Province, Chengdu 610066, China
| | - Chaowu Yang
- Sichuan Academy of Animal Sciences, Animal Breeding and Genetics Key Laboratory of Sichuan Province, Chengdu 610066, China
| | - Zengrong Zhang
- Sichuan Academy of Animal Sciences, Animal Breeding and Genetics Key Laboratory of Sichuan Province, Chengdu 610066, China
| | - Shuai Men
- College of Life Science, Sichuan University, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, "985 Project" Science Innovative Platform for Resource and Environment Protection of Southwestern, Chengdu 610065, China
| | - Zhikun Zhang
- College of Life Science, Sichuan University, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, "985 Project" Science Innovative Platform for Resource and Environment Protection of Southwestern, Chengdu 610065, China
| | - Wei Jiang
- College of Life Science, Sichuan University, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, "985 Project" Science Innovative Platform for Resource and Environment Protection of Southwestern, Chengdu 610065, China
| | - Hongning Wang
- College of Life Science, Sichuan University, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, "985 Project" Science Innovative Platform for Resource and Environment Protection of Southwestern, Chengdu 610065, China.
| |
Collapse
|
156
|
Cappelletti M, Presicce P, Lawson MJ, Chaturvedi V, Stankiewicz TE, Vanoni S, Harley IT, McAlees JW, Giles DA, Moreno-Fernandez ME, Rueda CM, Senthamaraikannan P, Sun X, Karns R, Hoebe K, Janssen EM, Karp CL, Hildeman DA, Hogan SP, Kallapur SG, Chougnet CA, Way SS, Divanovic S. Type I interferons regulate susceptibility to inflammation-induced preterm birth. JCI Insight 2017; 2:e91288. [PMID: 28289719 DOI: 10.1172/jci.insight.91288] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Preterm birth (PTB) is a leading worldwide cause of morbidity and mortality in infants. Maternal inflammation induced by microbial infection is a critical predisposing factor for PTB. However, biological processes associated with competency of pathogens, including viruses, to induce PTB or sensitize for secondary bacterial infection-driven PTB are unknown. We show that pathogen/pathogen-associated molecular pattern-driven activation of type I IFN/IFN receptor (IFNAR) was sufficient to prime for systemic and uterine proinflammatory chemokine and cytokine production and induction of PTB. Similarly, treatment with recombinant type I IFNs recapitulated such effects by exacerbating proinflammatory cytokine production and reducing the dose of secondary inflammatory challenge required for induction of PTB. Inflammatory challenge-driven induction of PTB was eliminated by defects in type I IFN, TLR, or IL-6 responsiveness, whereas the sequence of type I IFN sensing by IFNAR on hematopoietic cells was essential for regulation of proinflammatory cytokine production. Importantly, we also show that type I IFN priming effects are conserved from mice to nonhuman primates and humans, and expression of both type I IFNs and proinflammatory cytokines is upregulated in human PTB. Thus, activation of the type I IFN/IFNAR axis in pregnancy primes for inflammation-driven PTB and provides an actionable biomarker and therapeutic target for mitigating PTB risk.
Collapse
Affiliation(s)
| | - Pietro Presicce
- Division of Neonatology/Pulmonary Biology, Cincinnati Children's Hospital Research Foundation
| | - Matthew J Lawson
- Division of Immunobiology.,Molecular, Cellular and Biochemical Pharmacology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | | | - Simone Vanoni
- Division of Allergy and Immunology, Cincinnati Children's Hospital Research Foundation, and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | | | - Daniel A Giles
- Division of Immunobiology.,Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | | | | | | | - Rebekah Karns
- Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Research Foundation, and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | | | | | | | - Simon P Hogan
- Division of Allergy and Immunology, Cincinnati Children's Hospital Research Foundation, and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Suhas G Kallapur
- Division of Neonatology/Pulmonary Biology, Cincinnati Children's Hospital Research Foundation
| | | | | | | |
Collapse
|
157
|
Kumaki Y, Salazar AM, Wandersee MK, Barnard DL. Prophylactic and therapeutic intranasal administration with an immunomodulator, Hiltonol ® (Poly IC:LC), in a lethal SARS-CoV-infected BALB/c mouse model. Antiviral Res 2017; 139:1-12. [PMID: 27956136 PMCID: PMC7113886 DOI: 10.1016/j.antiviral.2016.12.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 12/02/2016] [Accepted: 12/04/2016] [Indexed: 12/25/2022]
Abstract
Hiltonol®, (Poly IC:LC), a potent immunomodulator, is a synthetic, double-stranded polyriboinosinic-polyribocytidylic acid (poly IC) stabilized with Poly-L-lysine and carboxymethyl cellulose (LC). Hiltonol® was tested for efficacy in a lethal SARS-CoV-infected BALB/c mouse model. Hiltonol® at 5, 1, 0.5 or 0.25 mg/kg/day by intranasal (i.n.) route resulted in significant survival benefit when administered at selected times 24 h prior to challenge with a lethal dose of mouse-adapted severe acute respiratory syndrome coronavirus (SARS-CoV). The infected BALB/c mice receiving the Hiltonol® treatments were also significantly effective in protecting mice against weight loss due to infection (p < 0.001). Groups of 20 mice were dosed with Hiltonol® at 2.5 or 0.75 mg/kg by intranasal instillation 7, 14, and 21 days before virus exposure and a second dose was given 24 h later, prophylactic Hiltonol® treatments (2.5 mg/kg/day) were completely protective in preventing death, and in causing significant reduction in lung hemorrhage scores, lung weights and lung virus titers. Hiltonol® was also effective as a therapeutic when give up to 8 h post virus exposure; 100% of the-infected mice were protected against death when Hiltonol® was administered at 5 mg/kg/day 8 h after infection. Our data suggest that Hiltonol® treatment of SARS-CoV infection in mice leads to substantial prophylactic and therapeutic effects and could be used for treatment of other virus disease such as those caused by MERS-CoV a related coronavirus. These properties might be therapeutically advantageous if Hiltonol® is considered for possible clinical use.
Collapse
Affiliation(s)
- Yohichi Kumaki
- Institute for Antiviral Research, Department of Animal, Dairy and Veterinary Science, 5600 Old Main Hill, Logan, Utah State University, Logan, UT 84322, USA.
| | | | - Miles K Wandersee
- Institute for Antiviral Research, Department of Animal, Dairy and Veterinary Science, 5600 Old Main Hill, Logan, Utah State University, Logan, UT 84322, USA
| | - Dale L Barnard
- Institute for Antiviral Research, Department of Animal, Dairy and Veterinary Science, 5600 Old Main Hill, Logan, Utah State University, Logan, UT 84322, USA
| |
Collapse
|
158
|
Abstract
INTRODUCTION The potential roles of toll-like receptors (TLRs) in immunopathogenesis of Ebola virus disease should be unraveled to provoke possible prophylactic or therapeutic implications of TLRs for EVD. Areas covered: The Ebola virus (EBOV) infection virtually paralyses all the main mechanisms responsible for induction of type I interferon (IFN-I) response. To summarize, EBOV infection interferes with: a) the TIR-domain-containing adapter-inducing interferon-β (TRIF) pathway that is mediated by TLR3 and TLR4 signaling; b) the interferon regulatory factor 7 (IRF7) pathway that is stimulated by TLR7 and TLR9; c) the intracellular signaling that is induced by retinoic acid-inducible gene 1 (RIG-I)-like receptors (RLRs); and d) the autocrine/paracrine feedback loop that is mediated by the IFN-stimulated gene factor 3 (ISGF3) complex. Upon infection with EBOV infection, TLR4 plays a key role in production of proinflammatory mediators. Expert opinion: It is theoretically possible that use of TLRs 3, 4, 7, and 9 agonists would be beneficial to improve the IFN-I response, despite their systemic side effects. Also, antagonist of TLR4 can be utilized to prevent production of proinflammatory cytokines. Additionally, it is highly recommended to design future investigations aimed at determining if the utilization of IFN-I would be beneficial for prophylactic/therapeutic programs of Ebola.
Collapse
Affiliation(s)
- Amene Saghazadeh
- a Research Center for Immunodeficiencies, Children's Medical Center , Tehran University of Medical Sciences , Tehran , Iran.,b Systematic Review and Meta-Analysis Expert Group (SRMEG) , Universal Scientific Education and Research Network (USERN) , Tehran , Iran
| | - Nima Rezaei
- a Research Center for Immunodeficiencies, Children's Medical Center , Tehran University of Medical Sciences , Tehran , Iran.,b Systematic Review and Meta-Analysis Expert Group (SRMEG) , Universal Scientific Education and Research Network (USERN) , Tehran , Iran.,c Department of Immunology, School of Medicine , Tehran University of Medical Sciences , Tehran , Iran.,d Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA) , Universal Scientific Education and Research Network (USERN) , Sheffield , UK
| |
Collapse
|
159
|
Cytokine regulation of lung Th17 response to airway immunization using LPS adjuvant. Mucosal Immunol 2017; 10:361-372. [PMID: 27328989 PMCID: PMC5179326 DOI: 10.1038/mi.2016.54] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 05/10/2016] [Indexed: 02/04/2023]
Abstract
Infections caused by bacteria in the airway preferentially induce a Th17 response. However, the mechanisms involved in the regulation of CD4 T-cell responses in the lungs are incompletely understood. Here, we have investigated the mechanisms involved in the regulation of Th17 differentiation in the lungs in response to immunization with lipopolysaccharide (LPS) as an adjuvant. Our data show that both Myd88 and TRIF are necessary for Th17 induction. This distinctive fate determination can be accounted for by the pattern of inflammatory cytokines induced by airway administration of LPS. We identified the production of interleukin (IL)-1β and IL-6 by small macrophages and IL-23 by alveolar dendritic cells (DCs), favoring Th17 responses, and IL-10 repressing interferon (IFN)-γ production. Furthermore, we show that exogenous IL-1β can drastically alter Th1 responses driven by influenza and lymphocytic choriomeningitis virus infection models and induce IL-17 production. Thus, the precision of the lung immune responses to potential threats is orchestrated by the cytokine microenvironment, can be repolarized and targeted therapeutically by altering the cytokine milieu. These results indicate that how the development of Th17 responses in the lung is regulated by the cytokines produced by lung DCs and macrophages in response to intranasal immunization with LPS adjuvant.
Collapse
|
160
|
Indirubin Treatment of Lipopolysaccharide-Induced Mastitis in a Mouse Model and Activity in Mouse Mammary Epithelial Cells. Mediators Inflamm 2017; 2017:3082805. [PMID: 28255203 PMCID: PMC5309412 DOI: 10.1155/2017/3082805] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 12/13/2016] [Accepted: 12/27/2016] [Indexed: 02/07/2023] Open
Abstract
Indirubin is a Chinese medicine extracted from indigo and known to be effective for treating chronic myelogenous leukemia, neoplasia, and inflammatory disease. This study evaluated the in vivo anti-inflammatory activity of indirubin in a lipopolysaccharide- (LPS-) induced mouse mastitis model. The indirubin mechanism and targets were evaluated in vitro in mouse mammary epithelial cells. In the mouse model, indirubin significantly attenuated the severity of inflammatory lesions, edema, inflammatory hyperemia, milk stasis and local tissue necrosis, and neutrophil infiltration. Indirubin significantly decreased myeloperoxidase activity and downregulated the production of tumor necrosis factor-α, interleukin-1β (IL-1β), and IL-6 caused by LPS. In vitro, indirubin inhibited LPS-stimulated expression of proinflammatory cytokines in a dose-dependent manner. It also downregulated LPS-induced toll-like receptor 4 (TLR4) expression and inhibited phosphorylation of LPS-induced nuclear transcription factor-kappa B (NF-κB) P65 protein and inhibitor of kappa B. In addition to its effect on the NF-κB signaling pathway, indirubin suppressed the mitogen-activated protein kinase (MAPK) signaling by inhibiting phosphorylation of extracellular signal-regulated kinase (ERK), P38, and c-jun NH2-terminal kinase (JNK). Indirubin improved LPS-induced mouse mastitis by suppressing TLR4 and downstream NF-κB and MAPK pathway inflammatory signals and might be a potential treatment of mastitis and other inflammatory diseases.
Collapse
|
161
|
Vanoni S, Tsai YT, Waddell A, Waggoner L, Klarquist J, Divanovic S, Hoebe K, Steinbrecher KA, Hogan SP. Myeloid-derived NF-κB negative regulation of PU.1 and c/EBP-β-driven pro-inflammatory cytokine production restrains LPS-induced shock. Innate Immun 2017; 23:175-187. [PMID: 27932520 PMCID: PMC5563821 DOI: 10.1177/1753425916681444] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Sepsis is a life-threatening event predominantly caused by Gram-negative bacteria. Bacterial infection causes a pronounced macrophage (MΦ) and dendritic cell activation that leads to excessive pro-inflammatory cytokine IL-1β, IL-6 and TNF-α production (cytokine storm), resulting in endotoxic shock. Previous experimental studies have revealed that inhibiting NF-κB signaling ameliorates disease symptoms; however, the contribution of myeloid p65 in endotoxic shock remains elusive. In this study, we demonstrate increased mortality in mice lacking p65 in the myeloid lineage (p65Δmye) compared with wild type mice upon ultra-pure LPS challenge. We show that increased susceptibility to LPS-induced shock was associated with elevated serum level of IL-1β and IL-6. Mechanistic analyses revealed that LPS-induced pro-inflammatory cytokine production was ameliorated in p65-deficient bone marrow-derived MΦs; however, p65-deficient 'activated' peritoneal MΦs exhibited elevated IL-1β and IL-6. We show that the elevated pro-inflammatory cytokine secretion was due, in part, to increased accumulation of IL-1β mRNA and protein in activated inflammatory MΦs. The increased IL-1β was linked with heightened binding of PU.1 and CCAAT/enhancer binding protein-β to Il1b and Il6 promoters in activated inflammatory MΦs. Our data provide insight into a role for NF-κB in the negative regulation of pro-inflammatory cytokines in myeloid cells.
Collapse
Affiliation(s)
- Simone Vanoni
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229
| | - Yi Ting Tsai
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229
| | - Amanda Waddell
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229
| | - Lisa Waggoner
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229
| | - Jared Klarquist
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229
| | - Senad Divanovic
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229
| | - Kasper Hoebe
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229
| | - Kris A. Steinbrecher
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229
| | - Simon P. Hogan
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229
| |
Collapse
|
162
|
Ziegler A, Soldner C, Lienenklaus S, Spanier J, Trittel S, Riese P, Kramps T, Weiss S, Heidenreich R, Jasny E, Guzmán CA, Kallen KJ, Fotin-Mleczek M, Kalinke U. A New RNA-Based Adjuvant Enhances Virus-Specific Vaccine Responses by Locally Triggering TLR- and RLH-Dependent Effects. THE JOURNAL OF IMMUNOLOGY 2017; 198:1595-1605. [DOI: 10.4049/jimmunol.1601129] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 12/12/2016] [Indexed: 12/24/2022]
|
163
|
Studzińska M, Jabłońska A, Wiśniewska-Ligier M, Nowakowska D, Gaj Z, Leśnikowski ZJ, Woźniakowska-Gęsicka T, Wilczyński J, Paradowska E. Association of TLR3 L412F Polymorphism with Cytomegalovirus Infection in Children. PLoS One 2017; 12:e0169420. [PMID: 28046022 PMCID: PMC5207783 DOI: 10.1371/journal.pone.0169420] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 12/17/2016] [Indexed: 02/06/2023] Open
Abstract
Intracellular Toll-like receptor 3 (TLR3) recognizes viral double-stranded RNA (dsRNA) and activates antiviral immune responses through the production of type I interferons (IFNs) and inflammatory cytokines. This receptor binds to dsRNA molecules produced during human cytomegalovirus (HCMV) replication. TLR7 senses viral single-stranded RNA (ssRNA) in endosomes, and it can interact with endogenous RNAs. We determined the genotype distribution of single-nucleotide polymorphisms (SNPs) within the TLR3 and TLR7 genes in children with HCMV infection and the relationship between TLR polymorphisms and viral infection. We genotyped 59 children with symptomatic HCMV infection and 78 healthy individuals for SNPs in the TLR3 (rs3775290, c.1377C>T, F459F; rs3775291, c.1234C>T, L412F; rs3775296, c.-7C>A) and TLR7 (rs179008, c.32A>T, Q11L; rs5741880, c.3+1716G>T) genes. SNP genotyping was performed using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) and capillary electrophoresis. The HCMV DNA load was quantified by real-time PCR. We found an increased frequency of the heterozygous genotype TLR3 L412F in children with HCMV infection compared with uninfected cases. In individuals with a mutation present in at least one allele of the L412F SNP, an increased risk of HCMV disease was found, and this result remained highly significant after Bonferroni’s correction for multiple testing (Pc < 0.001). The heterozygous genotype of this SNP was associated with the increased risk of HCMV disease in an adjusted model that included the HCMV DNA copy number in whole blood and urine (P < 0.001 and P = 0.008, respectively). Moreover, those with a heterozygous genotype of rs3775296 showed an increased relative risk of HCMV infection (P = 0.042), but this association did not reach statistical significance after correction for multiple testing. In contrast, the rs3775290 SNP of TLR3 and TLR7 SNPs were not related to viral infection. A moderate linkage disequilibrium (LD) was observed between the SNPs rs3775291 and rs3775296 (r2 = 0.514). We suggest that the L412F polymorphism in the TLR3 gene could be a genetic risk factor for the development of HCMV disease.
Collapse
Affiliation(s)
- Mirosława Studzińska
- Laboratory of Molecular Virology and Biological Chemistry, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | - Agnieszka Jabłońska
- Laboratory of Molecular Virology and Biological Chemistry, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | - Małgorzata Wiśniewska-Ligier
- Department of Pediatrics, Immunology, and Nephrology, Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | - Dorota Nowakowska
- Department of Perinatology and Gynecology, Polish Mother’s Memorial Hospital Research Institute, Lodz, Poland
| | - Zuzanna Gaj
- Department of Perinatology and Gynecology, Polish Mother’s Memorial Hospital Research Institute, Lodz, Poland
| | - Zbigniew J. Leśnikowski
- Laboratory of Molecular Virology and Biological Chemistry, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | | | - Jan Wilczyński
- 2nd Department of Obstetrics and Gynecology, Warsaw Medical University, Warsaw, Poland
| | - Edyta Paradowska
- Laboratory of Molecular Virology and Biological Chemistry, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
- * E-mail:
| |
Collapse
|
164
|
Chen CY, Liu HY, Hsueh YP. TLR3 downregulates expression of schizophrenia gene Disc1 via MYD88 to control neuronal morphology. EMBO Rep 2016; 18:169-183. [PMID: 27979975 PMCID: PMC5210159 DOI: 10.15252/embr.201642586] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 11/08/2016] [Accepted: 11/09/2016] [Indexed: 01/28/2023] Open
Abstract
Viral infection during fetal or neonatal stages increases the risk of developing neuropsychiatric disorders such as schizophrenia and autism spectrum disorders. Although neurons express several key regulators of innate immunity, the role of neuronal innate immunity in psychiatric disorders is still unclear. Using cultured neurons and in vivo mouse brain studies, we show here that Toll‐like receptor 3 (TLR3) acts through myeloid differentiation primary response gene 88 (MYD88) to negatively control Disrupted in schizophrenia 1 (Disc1) expression, resulting in impairment of neuronal development. Cytokines are not involved in TLR3‐mediated inhibition of dendrite outgrowth. Instead, TLR3 signaling suppresses expression of several psychiatric disorder‐related genes, including Disc1. The impaired dendritic arborization caused by TLR3 activation is rescued by MYD88 deficiency or DISC1 overexpression. In addition, TLR3 activation at the neonatal stage increases dendritic spine density, but narrows spine heads at postnatal day 21 (P21), suggesting a long‐lasting effect of TLR3 activation on spinogenesis. Our study reveals a novel mechanism of TLR3 in regulation of dendritic morphology and provides an explanation for how environmental factors influence mental health.
Collapse
Affiliation(s)
- Chiung-Ya Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Hsin-Yu Liu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Yi-Ping Hsueh
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
165
|
Sugiyama KI, Muroi M, Kinoshita M, Hamada O, Minai Y, Sugita-Konishi Y, Kamata Y, Tanamoto KI. NF-κB activation via MyD88-dependent Toll-like receptor signaling is inhibited by trichothecene mycotoxin deoxynivalenol. J Toxicol Sci 2016; 41:273-9. [PMID: 26961612 DOI: 10.2131/jts.41.273] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Macrophages induce the innate immunity by recognizing pathogens through Toll-like receptors (TLRs), which sense pathogen-associated molecular patterns. Myeloid differentiation factor 88 (MyD88), which is an essential adaptor molecule for most TLRs, mediates the induction of inflammatory cytokines through nuclear factor κB (NF-κB). Trichothecene mycotoxin deoxynivalenol (DON) shows immunotoxic effects by interrupting inflammatory mediators produced by activated macrophages. The present study investigates the effect of DON on NF-κB in activated macrophages through MyD88-dependent pathways. DON inhibited NF-κB-dependent reporter activity induced by MyD88-dependent TLR agonists. In addition, lipopolysaccharide-induced phosphorylation of interleukin-1 receptor-associated kinase 1 and inhibitor κBα were attenuated by DON. Furthermore, DON downregulated the expression level of MyD88. These results suggest that DON inhibits NF-κB activation in macrophages stimulated with TLR ligands via MyD88-dependent TLR signals. Therefore exposure to DON may lead to the inhibition of MyD88-dependent pathway of TLR signaling.
Collapse
Affiliation(s)
- Kei-ichi Sugiyama
- Division of Genetics and Mutagenesis, National Institute of Health Sciences
| | | | | | | | | | | | | | | |
Collapse
|
166
|
Rothschild DE, Zhang Y, Diao N, Lee CK, Chen K, Caswell CC, Slade DJ, Helm RF, LeRoith T, Li L, Allen IC. Enhanced Mucosal Defense and Reduced Tumor Burden in Mice with the Compromised Negative Regulator IRAK-M. EBioMedicine 2016; 15:36-47. [PMID: 27939424 PMCID: PMC5233813 DOI: 10.1016/j.ebiom.2016.11.039] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 11/30/2016] [Accepted: 11/30/2016] [Indexed: 12/27/2022] Open
Abstract
Aberrant inflammation is a hallmark of inflammatory bowel disease (IBD) and colorectal cancer. IRAK-M is a critical negative regulator of TLR signaling and overzealous inflammation. Here we utilize data from human studies and Irak-m-/- mice to elucidate the role of IRAK-M in the modulation of gastrointestinal immune system homeostasis. In human patients, IRAK-M expression is up-regulated during IBD and colorectal cancer. Further functional studies in mice revealed that Irak-m-/- animals are protected against colitis and colitis associated tumorigenesis. Mechanistically, our data revealed that the gastrointestinal immune system of Irak-m-/- mice is highly efficient at eliminating microbial translocation following epithelial barrier damage. This attenuation of pathogenesis is associated with expanded areas of gastrointestinal associated lymphoid tissue (GALT), increased neutrophil migration, and enhanced T-cell recruitment. Further evaluation of Irak-m-/- mice revealed a splice variant that robustly activates NF-κB signaling. Together, these data identify IRAK-M as a potential target for future therapeutic intervention.
Collapse
Affiliation(s)
- Daniel E Rothschild
- Department of Biomedical Sciences and Pathobiology, VA-MD College of Veterinary Medicine, Blacksburg, Virginia 24061, United States
| | - Yao Zhang
- Department of Biological Sciences, Virginia Polytechnic and State University, Blacksburg, Virginia 24061, United States
| | - Na Diao
- Department of Biological Sciences, Virginia Polytechnic and State University, Blacksburg, Virginia 24061, United States
| | - Christina K Lee
- Department of Biological Sciences, Virginia Polytechnic and State University, Blacksburg, Virginia 24061, United States
| | - Keqiang Chen
- Department of Biological Sciences, Virginia Polytechnic and State University, Blacksburg, Virginia 24061, United States
| | - Clayton C Caswell
- Department of Biomedical Sciences and Pathobiology, VA-MD College of Veterinary Medicine, Blacksburg, Virginia 24061, United States
| | - Daniel J Slade
- Department of Biochemistry, Virginia Polytechnic and State University, Blacksburg, Virginia 24061, United States
| | - Richard F Helm
- Department of Biochemistry, Virginia Polytechnic and State University, Blacksburg, Virginia 24061, United States
| | - Tanya LeRoith
- Department of Biomedical Sciences and Pathobiology, VA-MD College of Veterinary Medicine, Blacksburg, Virginia 24061, United States
| | - Liwu Li
- Department of Biological Sciences, Virginia Polytechnic and State University, Blacksburg, Virginia 24061, United States.
| | - Irving C Allen
- Department of Biomedical Sciences and Pathobiology, VA-MD College of Veterinary Medicine, Blacksburg, Virginia 24061, United States.
| |
Collapse
|
167
|
Toubai T, Mathewson ND, Magenau J, Reddy P. Danger Signals and Graft-versus-host Disease: Current Understanding and Future Perspectives. Front Immunol 2016; 7:539. [PMID: 27965667 PMCID: PMC5126092 DOI: 10.3389/fimmu.2016.00539] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 11/15/2016] [Indexed: 12/22/2022] Open
Abstract
Graft-versus-host response after allogeneic hematopoietic stem cell transplantation (allo-HCT) represents one of the most intense inflammatory responses observed in humans. Host conditioning facilitates engraftment of donor cells, but the tissue injury caused from it primes the critical first steps in the development of acute graft-versus-host disease (GVHD). Tissue injuries release pro-inflammatory cytokines (such as TNF-α, IL-1β, and IL-6) through widespread stimulation of pattern recognition receptors (PRRs) by the release of danger stimuli, such as damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs). DAMPs and PAMPs function as potent stimulators for host and donor-derived antigen presenting cells (APCs) that in turn activate and amplify the responses of alloreactive donor T cells. Emerging data also point towards a role for suppression of DAMP induced inflammation by the APCs and donor T cells in mitigating GVHD severity. In this review, we summarize the current understanding on the role of danger stimuli, such as the DAMPs and PAMPs, in GVHD.
Collapse
Affiliation(s)
- Tomomi Toubai
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center , Ann Arbor, MI , USA
| | - Nathan D Mathewson
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute , Boston, MA , USA
| | - John Magenau
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center , Ann Arbor, MI , USA
| | - Pavan Reddy
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center , Ann Arbor, MI , USA
| |
Collapse
|
168
|
Westphal A, Cheng W, Yu J, Grassl G, Krautkrämer M, Holst O, Föger N, Lee KH. Lysosomal trafficking regulator Lyst links membrane trafficking to toll-like receptor-mediated inflammatory responses. J Exp Med 2016; 214:227-244. [PMID: 27881733 PMCID: PMC5206490 DOI: 10.1084/jem.20141461] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 04/11/2016] [Accepted: 11/01/2016] [Indexed: 01/28/2023] Open
Abstract
Westphal et al. demonstrate a role of lysosomal trafficking regulator Lyst that couples the regulation of endolysosomal trafficking to inflammatory responses by the control of toll-like receptor–mediated endosomal TRIF signaling pathways. Subcellular compartmentalization of receptor signaling is an emerging principle in innate immunity. However, the functional integration of receptor signaling pathways into membrane trafficking routes and its physiological relevance for immune responses is still largely unclear. In this study, using Lyst-mutant beige mice, we show that lysosomal trafficking regulator Lyst links endolysosomal organization to the selective control of toll-like receptor 3 (TLR3)– and TLR4-mediated proinflammatory responses. Consequently, Lyst-mutant mice showed increased susceptibility to bacterial infection and were largely resistant to endotoxin-induced septic shock. Mechanistic analysis revealed that Lyst specifically controls TLR3- and TLR4-induced endosomal TRIF (TIR domain–containing adapter-inducing interferon β) signaling pathways. Loss of functional Lyst leads to dysregulated phagosomal maturation, resulting in a failure to form an activation-induced Rab7+ endosomal/phagosomal compartment. This specific Rab7+ compartment was further demonstrated to serve as a major site for active TRIF signaling events, thus linking phagosomal maturation to specific TLR signaling pathways. The immunoregulatory role of Lyst on TLR signaling pathways was confirmed in human cells by CRISPR/Cas9-mediated gene inactivation. As mutations in LYST cause human Chédiak-Higashi syndrome, a severe immunodeficiency, our findings also contribute to a better understanding of human disease mechanisms.
Collapse
Affiliation(s)
- Andreas Westphal
- Institute of Clinical Chemistry, Inflammation Research Group, Hannover Medical School, 30625 Hannover, Germany
| | - Weijia Cheng
- Institute of Clinical Chemistry, Inflammation Research Group, Hannover Medical School, 30625 Hannover, Germany
| | - Jinbo Yu
- Institute of Clinical Chemistry, Inflammation Research Group, Hannover Medical School, 30625 Hannover, Germany
| | - Guntram Grassl
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, 30625 Hannover, Germany
| | - Martina Krautkrämer
- Institute of Clinical Chemistry, Inflammation Research Group, Hannover Medical School, 30625 Hannover, Germany
| | - Otto Holst
- Division of Structural Biochemistry, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, 23845 Borstel, Germany
| | - Niko Föger
- Institute of Clinical Chemistry, Inflammation Research Group, Hannover Medical School, 30625 Hannover, Germany
| | - Kyeong-Hee Lee
- Institute of Clinical Chemistry, Inflammation Research Group, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
169
|
Abstract
As it is a hard-wired system for responses to microbes, innate immunity is particularly susceptible to classical genetic analysis. Mutations led the way to the discovery of many of the molecular elements of innate immune sensing and signaling pathways. In turn, the need for a faster way to find the molecular causes of mutation-induced phenotypes triggered a huge transformation in forward genetics. During the 1980s and 1990s, many heritable phenotypes were ascribed to mutations through positional cloning. In mice, this required three steps. First, a genetic mapping step was used to show that a given phenotype emanated from a circumscribed region of the genome. Second, a physical mapping step was undertaken, in which all of the region was cloned and its gene content determined. Finally, a concerted search for the mutation was performed. Such projects usually lasted for several years, but could produce breakthroughs in our understanding of biological processes. Publication of the annotated mouse genome sequence in 2002 made physical mapping unnecessary. More recently we devised a new technology for automated genetic mapping, which eliminated both genetic mapping and the search for mutations among candidate genes. The cause of phenotype can now be determined instantaneously. We have created more than 100,000 coding/splicing mutations. And by screening for defects of innate and adaptive immunity we have discovered many "new" proteins needed for innate immune function.
Collapse
Affiliation(s)
- Bruce Beutler
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8505, United States.
| |
Collapse
|
170
|
Hirai-Yuki A, Hensley L, McGivern DR, González-López O, Das A, Feng H, Sun L, Wilson JE, Hu F, Feng Z, Lovell W, Misumi I, Ting JPY, Montgomery S, Cullen J, Whitmire JK, Lemon SM. MAVS-dependent host species range and pathogenicity of human hepatitis A virus. Science 2016; 353:1541-1545. [PMID: 27633528 PMCID: PMC5068972 DOI: 10.1126/science.aaf8325] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 09/02/2016] [Indexed: 12/24/2022]
Abstract
Hepatotropic viruses are important causes of human disease, but the intrahepatic immune response to hepatitis viruses is poorly understood because of a lack of tractable small- animal models. We describe a murine model of hepatitis A virus (HAV) infection that recapitulates critical features of type A hepatitis in humans. We demonstrate that the capacity of HAV to evade MAVS-mediated type I interferon responses defines its host species range. HAV-induced liver injury was associated with interferon-independent intrinsic hepatocellular apoptosis and hepatic inflammation that unexpectedly resulted from MAVS and IRF3/7 signaling. This murine model thus reveals a previously undefined link between innate immune responses to virus infection and acute liver injury, providing a new paradigm for viral pathogenesis in the liver.
Collapse
Affiliation(s)
- Asuka Hirai-Yuki
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Lucinda Hensley
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA
| | - David R McGivern
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Medicine, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Olga González-López
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Anshuman Das
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Hui Feng
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Lu Sun
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Justin E Wilson
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Genetics, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Fengyu Hu
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Zongdi Feng
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA
| | - William Lovell
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Ichiro Misumi
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Genetics, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Jenny P-Y Ting
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Genetics, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Stephanie Montgomery
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27517, USA
| | - John Cullen
- Department of Population Health and Pathobiology, North Carolina State University College of Veterinary Medicine, Raleigh, NC 27607, USA
| | - Jason K Whitmire
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Genetics, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Stanley M Lemon
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Medicine, University of North Carolina, Chapel Hill, NC 27517, USA.
| |
Collapse
|
171
|
In Vivo Conditions Enable IFNAR-Independent Type I Interferon Production by Peritoneal CD11b+ Cells upon Thogoto Virus Infection. J Virol 2016; 90:9330-7. [PMID: 27512061 DOI: 10.1128/jvi.00744-16] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 07/28/2016] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Type I interferons (IFNs) crucially contribute to host survival upon viral infections. Robust expression of type I IFNs (IFN-α/β) and induction of an antiviral state critically depend on amplification of the IFN signal via the type I IFN receptor (IFNAR). A small amount of type I IFN produced early upon virus infection binds the IFNAR and activates a self-enhancing positive feedback loop, resulting in induction of large, protective amounts of IFN-α. Unexpectedly, we found robust, systemic IFN-α expression upon infection of IFNAR knockout mice with the orthomyxovirus Thogoto virus (THOV). The IFNAR-independent IFN-α production required in vivo conditions and was not achieved during in vitro infection. Using replication-incompetent THOV-derived virus-like particles, we demonstrate that IFNAR-independent type I IFN induction depends on viral polymerase activity but is largely independent of viral replication. To discover the cell type responsible for this effect, we used type I IFN reporter mice and identified CD11b(+) F4/80(+) myeloid cells within the peritoneal cavity of infected animals as the main source of IFNAR-independent type I IFN, corresponding to the particular tropism of THOV for this cell type. IMPORTANCE Type I IFNs are crucial for the survival of a host upon most viral infections, and, moreover, they shape subsequent adaptive immune responses. Production of protective amounts of type I IFN critically depends on the positive feedback amplification via the IFNAR. Unexpectedly, we observed robust IFNAR-independent type I IFN expression upon THOV infection and unraveled molecular mechanisms and determined the tissue and cell type involved. Our data indicate that the host can effectively use alternative pathways to induce type I IFN responses if the classical feedback amplification is not available. Understanding how type I IFN can be produced in large amounts independently of IFNAR-dependent enhancement will identify mechanisms which might contribute to novel therapeutic strategies to fight viral pathogens.
Collapse
|
172
|
Monlish DA, Bhatt ST, Schuettpelz LG. The Role of Toll-Like Receptors in Hematopoietic Malignancies. Front Immunol 2016; 7:390. [PMID: 27733853 PMCID: PMC5039188 DOI: 10.3389/fimmu.2016.00390] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 09/15/2016] [Indexed: 12/02/2022] Open
Abstract
Toll-like receptors (TLRs) are a family of pattern recognition receptors that shape the innate immune system by identifying pathogen-associated molecular patterns and host-derived damage-associated molecular patterns. TLRs are widely expressed on both immune cells and non-immune cells, including hematopoietic stem and progenitor cells, effector immune cell populations, and endothelial cells. In addition to their well-known role in the innate immune response to acute infection or injury, accumulating evidence supports a role for TLRs in the development of hematopoietic and other malignancies. Several hematopoietic disorders, including lymphoproliferative disorders and myelodysplastic syndromes, which possess a high risk of transformation to leukemia, have been linked to aberrant TLR signaling. Furthermore, activation of TLRs leads to the induction of a number of proinflammatory cytokines and chemokines, which can promote tumorigenesis by driving cell proliferation and migration and providing a favorable microenvironment for tumor cells. Beyond hematopoietic malignancies, the upregulation of a number of TLRs has been linked to promoting tumor cell survival, proliferation, and metastasis in a variety of cancers, including those of the colon, breast, and lung. This review focuses on the contribution of TLRs to hematopoietic malignancies, highlighting the known direct and indirect effects of TLR signaling on tumor cells and their microenvironment. In addition, the utility of TLR agonists and antagonists as potential therapeutics in the treatment of hematopoietic malignancies is discussed.
Collapse
Affiliation(s)
- Darlene A Monlish
- Department of Pediatrics, Washington University School of Medicine , St. Louis, MO , USA
| | - Sima T Bhatt
- Department of Pediatrics, Washington University School of Medicine , St. Louis, MO , USA
| | - Laura G Schuettpelz
- Department of Pediatrics, Washington University School of Medicine , St. Louis, MO , USA
| |
Collapse
|
173
|
Wang X, Majumdar T, Kessler P, Ozhegov E, Zhang Y, Chattopadhyay S, Barik S, Sen GC. STING Requires the Adaptor TRIF to Trigger Innate Immune Responses to Microbial Infection. Cell Host Microbe 2016; 20:329-341. [PMID: 27631700 PMCID: PMC5026396 DOI: 10.1016/j.chom.2016.08.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/14/2016] [Accepted: 08/09/2016] [Indexed: 12/19/2022]
Abstract
The intracellular microbial nucleic acid sensors, TLR3 and STING, recognize pathogen molecules and signal to activate the interferon pathway. The TIR-domain containing protein TRIF is the sole adaptor of TLR3. Here, we report an essential role for TRIF in STING signaling: various activators of STING could not induce genes in the absence of TRIF. TRIF and STING interacted directly, through their carboxy-terminal domains, to promote STING dimerization, intermembrane translocation, and signaling. Herpes simplex virus (HSV), which triggers the STING signaling pathway and is controlled by it, replicated more efficiently in the absence of TRIF, and HSV-infected TRIF(-/-) mice displayed pronounced pathology. Our results indicate that defective STING signaling may be responsible for the observed genetic association between TRIF mutations and herpes simplex encephalitis in patients.
Collapse
Affiliation(s)
- Xin Wang
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Tanmay Majumdar
- Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA
| | - Patricia Kessler
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Evgeny Ozhegov
- Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA
| | - Ying Zhang
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Saurabh Chattopadhyay
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Sailen Barik
- Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA
| | - Ganes C Sen
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
174
|
Takeda K. Evolution and integration of innate immune recognition systems: the Toll-like receptors. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519050110011101] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Toll-like receptors (TLRs) recognize specific components of micro-organisms and trigger the activation of innate immunity. TLR-mediated signaling pathways are now rapidly being elucidated. TLR signaling originates from the cytoplasmic Toll/IL-1 receptor (TIR) domain, which is conserved among all TLRs. Furthermore, TIR domain-containing adaptors, such as MyD88, TRIF, TIRAP and TRAM, play essential roles in TLR signaling. MyD88 is essential for inflammatory cytokine production via all TLRs, whereas TRIF is involved in TLR3- and TLR4-mediated MyD88-independent induction of IFN-β. Thus, innate immunity represents a skilful system that senses microbial invasion and initiates appropriate immune responses.
Collapse
Affiliation(s)
- Kiyoshi Takeda
- Department of Molecular Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan, -u.ac.jp
| |
Collapse
|
175
|
Pietras EM, Saha SK, Genhong Cheng. The interferon response to bacterial and viral infections. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519060120040601] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Type I interferons (IFNs) were first described several decades ago as soluble factors that were capable of `interfering' with viral replication when added to infected cells. Type I IFNs have been shown to be induced by recognition of viral DNA and RNA via three distinct pathways: (i) a TRIFdependent pathway in macrophages via TLRs 3 and 4; (ii) a MyD88-dependent pathway in plasmacytoid dendritic cells (pDCs) via TLRs 7/8 and 9; and (iii) an intracellular recognition pathway utilizing the cytoplasmic receptors RIG-I/MDA5. Interestingly, these viral recognition pathways converge on TRAF3, which induces interferon through the activation of IRF3 or IRF7 by the TBK-1 and IKKi complexes. While type I IFN has been traditionally associated with antiviral responses, recent studies have demonstrated that many bacteria also induce type I interferon responses. The mechanisms of type I IFN induction and its role in host defense, however, are largely unclear. Studies with the Gram-positive intracellular bacterium Listeria monocytogenes indicated that it may trigger type I IFN induction through novel TLR-independent intracellular receptors and type I IFN may play a detrimental role to host response against listerial infection. In this article, we summarize some of these findings and discuss the functional differences of type I IFNs in bacterial and viral infections.
Collapse
Affiliation(s)
- Eric M. Pietras
- Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, California, USA
| | - Supriya K. Saha
- Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, California, USA, Medical Scientist Training Program, University of California Los Angeles, Los Angeles, California, USA
| | - Genhong Cheng
- Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, California, USA, , Molecular Biology Institute, University of California Los Angeles, Los Angeles, California, USA, Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
176
|
Medvedev AE, Sabroe I, Hasday JD, Vogel SN. Invited review: Tolerance to microbial TLR ligands: molecular mechanisms and relevance to disease. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519060120030201] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Many host cell types, including endothelial and epithelial cells, neutrophils, monocytes, natural killer cells, dendritic cells and macrophages, initiate the first line of defense against infection by sensing conserved microbial structures through Toll-like receptors (TLRs). Recognition of microbial ligands by TLRs induces their oligomerization and triggers intracellular signaling pathways, leading to production of pro- and anti-inflammatory cytokines. Dysregulation of the fine molecular mechanisms that tightly control TLR signaling may lead to hyperactivation of host cells by microbial products and septic shock. A prior exposure to bacterial products such as lipopolysaccharide (LPS) may result in a transient state of refractoriness to subsequent challenge that has been referred to as `tolerance'. Tolerance has been postulated as a protective mechanism limiting excessive inflammation and preventing septic shock. However, tolerance may compromise the host's ability to counteract subsequent bacterial challenge since many septic patients exhibit an increased incidence of recurrent bacterial infection and suppressed monocyte responsiveness to LPS, closely resembling the tolerant phenotype. Thus, by studying mechanisms of microbial tolerance, we may gain insights into how normal regulatory mechanisms are dysregulated, leading ultimately to microbial hyporesponsivess and life-threatening disease. In this review, we present current theories of the molecular mechanisms that underlie induction and maintenance of `microbial tolerance', and discuss the possible relevance of tolerance to several infectious and non-infectious diseases.
Collapse
Affiliation(s)
- Andrei E. Medvedev
- Department of Microbiology and Immunology, University of Maryland, Baltimore (UMB), Baltimore, Maryland, USA,
| | - Ian Sabroe
- Academic Unit of Respiratory Medicine, Division of Genomic Medicine, University of Sheffield, Royal Hallamshire Hospital, Sheffield, UK
| | - Jeffrey D. Hasday
- Department of Medicine, University of Maryland, Baltimore (UMB), Baltimore, Maryland, USA
| | - Stefanie N. Vogel
- Department of Microbiology and Immunology, University of Maryland, Baltimore (UMB), Baltimore, Maryland, USA
| |
Collapse
|
177
|
Abstract
Toll-like receptors (TLRs) are essential for the recognition of distinct pathogen-associated molecular patterns (PAMPs). Activation of TLRs induces intracellular signaling pathways which lead to the production of pro-inflammatory cytokines, chemokines, and interferon (IFN)-inducible genes. TIR domain containing adaptor molecules in turn determine the signaling specificity of the response. Recent studies demonstrated that serine/threonine kinases IKK-i/TBK1 are critical for the regulation of IFN-β as well as IFN-inducible genes. In response to lipopolysaccharide (LPS), transfection of poly(I:C) and viral infection, embryonic fibroblasts (MEFs) derived from TBK1-deficient ( TBK1— /—) mice show impaired production of IFN-inducible genes, but not pro-inflammatory cytokines. Although IKK-i— /— mice show normal production of these genes, MEFs from IKK-i/ TBK1-doubly deficient mice were completely defective in the induction of IFN-β as well as IFN-inducible genes in response to poly(I:C) stimulation. Activation of IFN-regulatory factor (IRF) 3 in response to LPS and poly(I:C) was abolished in IKK-i/TBK1 doubly deficient cells. Interestingly, intracellular transduction of poly(I:C) initiates activation of IFN response in a TLR3-independent manner. These observations demonstrate that IKK-i/TBK1 signaling is essential for both TLR3-dependent and TLR3-independent viral and dsRNA-induced IFN responses.
Collapse
Affiliation(s)
- Osamu Takeuchi
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan, ERATO, Japan Science and Technology Agency, Osaka, Japan
| | - Hiroaki Hemmi
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Shizuo Akira
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan, ERATO, Japan Science and Technology Agency, Osaka, Japan, -u.ac.jp
| |
Collapse
|
178
|
Hoebe K, Beutler B. LPS, dsRNA and the interferon bridge to adaptive immune responses: Trif, Tram, and other TIR adaptor proteins. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519040100021001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Toll-like receptors (TLRs) expressed on antigen-presenting cells (APCs), form a critical link between innate and the adaptive immune responses. Activation of TLRs by LPS and dsRNA results in up-regulation of co-stimulatory molecules (UCM) essential for the generation of robust T-cell responses. It is now evident that type I interferons (IFNs) play an important role in UCM and in the subsequent maturation of APCs. The recently identified adaptor molecules Trif and Tram, unlike their counterparts MyD88 and MAL/Tirap, induce type I IFN via the TLR4 signaling pathway, whereas Trif appears to be the sole adaptor molecule involved in TLR3 signaling, resulting in subsequent production of type I IFN. Here, we discuss how Trif and type I IFN are involved in the optimization of APC-T cell interaction in response not only to viral but also bacterial stimuli.
Collapse
Affiliation(s)
- Kasper Hoebe
- Department of Immunology, The Scripps Research Institute, La Jolla, California, USA,
| | - Bruce Beutler
- Department of Immunology, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
179
|
Xue S, Liu C, Sun X, Li W, Zhang C, Zhou X, Lu Y, Xiao J, Li C, Xu X, Sun B, Xu G, Wang H. TET3 Inhibits Type I IFN Production Independent of DNA Demethylation. Cell Rep 2016; 16:1096-1105. [PMID: 27425624 DOI: 10.1016/j.celrep.2016.06.068] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 05/11/2016] [Accepted: 06/15/2016] [Indexed: 12/12/2022] Open
Abstract
Type I interferons (IFNs) play both beneficial and harmful roles in antiviral responses. Precise regulation of host type I IFNs is thus needed to prevent immune dysregulation. Here, we find that the DNA demethylase TET3 is a negative regulator of IFN-β in response to poly(I:C) stimulation or viral infection. Deletion of TET3 enhances antiviral responses, with elevated expression of IFN-β and IFN-stimulated genes. The catalytic domain of TET3 was critical for the suppression of IFN-β production, but TET3 enzymatic activity was dispensable. Instead, the catalytic domain of TET3 interacts with HDAC1 and SIN3A, thus enhancing their binding to the Ifnb1 promoter. Our study demonstrates that TET3 negatively regulates type I IFN production independent of DNA demethylation. This not only sheds light on TET3 as a signaling protein in immune cells for gene regulation but also will help to develop strategies to prevent type I IFN-related disease.
Collapse
Affiliation(s)
- Shengjie Xue
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Chang Liu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Xiujie Sun
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Weiyun Li
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Chi Zhang
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Xin Zhou
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Yao Lu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Jun Xiao
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Chunyang Li
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Xiaoyan Xu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Bing Sun
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guoliang Xu
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hongyan Wang
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China.
| |
Collapse
|
180
|
Polycarpou A, Holland MJ, Karageorgiou I, Eddaoudi A, Walker SL, Willcocks S, Lockwood DNJ. Mycobacterium leprae Activates Toll-Like Receptor-4 Signaling and Expression on Macrophages Depending on Previous Bacillus Calmette-Guerin Vaccination. Front Cell Infect Microbiol 2016; 6:72. [PMID: 27458573 PMCID: PMC4937034 DOI: 10.3389/fcimb.2016.00072] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/24/2016] [Indexed: 01/09/2023] Open
Abstract
Toll-like receptor (TLR)-1 and TLR2 have been shown to be receptors for Mycobacterium leprae (M. leprae), yet it is unclear whether M. leprae can signal through alternative TLRs. Other mycobacterial species possess ligands for TLR4 and genetic association studies in human populations suggest that people with TLR4 polymorphisms may be protected against leprosy. Using human embryonic kidney (HEK)-293 cells co-transfected with TLR4, we demonstrate that M. leprae activates TLR4. We used human macrophages to show that M. leprae stimulation of cytokine production is diminished if pre-treated with TLR4 neutralizing antibody. TLR4 protein expression was up-regulated on macrophages derived from non-bacillus Calmette-Guerin (BCG) vaccinated healthy volunteers after incubation with M. leprae, whereas it was down-regulated in macrophages derived from BCG-vaccinated donors. Finally, pre-treatment of macrophages derived from BCG-naive donors with BCG reversed the effect of M. leprae on TLR4 expression. This may be a newly described phenomenon by which BCG vaccination stimulates “non-specific” protection to the human immune system.
Collapse
Affiliation(s)
- Anastasia Polycarpou
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine London, UK
| | - Martin J Holland
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine London, UK
| | - Ioannis Karageorgiou
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine London, UK
| | - Ayad Eddaoudi
- Molecular and Cellular Immunology Unit, Institute of Child Health, University College London London, UK
| | - Stephen L Walker
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine London, UK
| | - Sam Willcocks
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine London, UK
| | - Diana N J Lockwood
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine London, UK
| |
Collapse
|
181
|
Abstract
The innate immune response is a coordinated set of reactions involving cells of myeloid lineage and a network of signaling molecules. Such a response takes place in the CNS during trauma, stroke, spinal cord injury, and neurodegenerative diseases, suggesting that macrophages/microglia are the cells that perpetuate the progressive neuronal damage. However, there is accumulating evidence that these cells and their secreted proinflammatory molecules have more beneficial effects than detrimental consequences for the neuronal elements. Indeed, a timely controlled innate immune response may limit toxicity in swiftly eliminating foreign materials and debris that are known to interfere with recovery and regeneration. Each step of the immune cascade is under the tight control of stimulatory and inhibitory signals. Glucocorticoids (GCs) act as the critical negative feedback on all myeloid cells, including those present within the brain parenchyma. Because too little is like too much, both an inappropriate feedback of GCs on microglia and high circulating GC levels in stressed individuals have been associated with deleterious consequences for the brain. In this review, the authors discuss both sides of the story with a particular emphasis on the neuro-protective role of endogenous GCs during immune challenges and the problems in determining whether GCs can be a good therapy for the treatment of neuropathological conditions.
Collapse
Affiliation(s)
- Isaias Glezer
- Laboratory of Molecular Endocrinology, CHUL Research Center, Department of Anatomy and Physiology, Laval University, Québec, Canada
| | | |
Collapse
|
182
|
Bhattacharya J, Westphalen K. Macrophage-epithelial interactions in pulmonary alveoli. Semin Immunopathol 2016; 38:461-9. [PMID: 27170185 DOI: 10.1007/s00281-016-0569-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 04/29/2016] [Indexed: 12/20/2022]
Abstract
Alveolar macrophages have been investigated for years by approaches involving macrophage extraction from the lung by bronchoalveolar lavage, or by cell removal from lung tissue. Since extracted macrophages are studied outside their natural milieu, there is little understanding of the extent to which alveolar macrophages interact with the epithelium, or with one another to generate the lung's innate immune response to pathogen challenge. Here, we review new evidence of macrophage-epithelial interactions in the lung, and we address the emerging understanding that the alveolar epithelium plays an important role in orchestrating the macrophage-driven immune response.
Collapse
Affiliation(s)
- Jahar Bhattacharya
- Departments of Medicine and Physiology and Cellular Biophysics, Columbia University, New York, NY, USA.
| | - Kristin Westphalen
- Department of Anesthesiology, Ludwig Maximilians University, Munich, Germany.,Comprehensive Pneumology Center (CPC), German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
183
|
Ullah MO, Sweet MJ, Mansell A, Kellie S, Kobe B. TRIF-dependent TLR signaling, its functions in host defense and inflammation, and its potential as a therapeutic target. J Leukoc Biol 2016; 100:27-45. [PMID: 27162325 DOI: 10.1189/jlb.2ri1115-531r] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 04/04/2016] [Indexed: 12/16/2022] Open
Abstract
Toll/IL-1R domain-containing adaptor-inducing IFN-β (TRIF)-dependent signaling is required for TLR-mediated production of type-I IFN and several other proinflammatory mediators. Various pathogens target the signaling molecules and transcriptional regulators acting in the TRIF pathway, thus demonstrating the importance of this pathway in host defense. Indeed, the TRIF pathway contributes to control of both viral and bacterial pathogens through promotion of inflammatory mediators and activation of antimicrobial responses. TRIF signaling also has both protective and pathologic roles in several chronic inflammatory disease conditions, as well as an essential function in wound-repair processes. Here, we review our current understanding of the regulatory mechanisms that control TRIF-dependent TLR signaling, the role of the TRIF pathway in different infectious and noninfectious pathologic states, and the potential for manipulating TRIF-dependent TLR signaling for therapeutic benefit.
Collapse
Affiliation(s)
- M Obayed Ullah
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia; Institute for Molecular Bioscience, Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience, Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia; Institute for Molecular Bioscience, Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, Queensland, Australia; and
| | - Ashley Mansell
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Monash University, Melbourne, Victoria, Australia
| | - Stuart Kellie
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia; Institute for Molecular Bioscience, Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia; Institute for Molecular Bioscience, Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia;
| |
Collapse
|
184
|
Holding the Inflammatory System in Check: TLRs and Their Targeted Therapy in Asthma. Mediators Inflamm 2016; 2016:2180417. [PMID: 27274620 PMCID: PMC4870363 DOI: 10.1155/2016/2180417] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 04/13/2016] [Indexed: 02/06/2023] Open
Abstract
Inflammation is a complex biological response to detrimental stimuli and can be a double-edged sword. Inflammation plays a protective role in removing pathogenic factors, but dysregulated inflammation is associated with several major fatal diseases such as asthma, cancer, and cardiovascular diseases. Asthma is a complex heterogenous disease caused by genetic and environmental factors. TLRs are the primary proteins associated with the innate and adaptive immune responses to these fatal factors and play an important role in recognizing pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs), which initiates the downstream immune response. Due to the complex TLRs cascade and nowadays unsuccessful control in asthma, new studies are focused on TLRs and other potential targets in TLR cascade to minimize airway inflammation.
Collapse
|
185
|
Knockdown of Myeloid Differentiation Factor 88 Attenuates Lipopolysaccharide-Induced Inflammatory Response in Pancreatic Ductal Cells. Pancreas 2016; 45:755-60. [PMID: 26684858 DOI: 10.1097/mpa.0000000000000565] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVES The aim of the study was to explore the potential role of myeloid differentiation factor 88 (MyD88), which acts as an adaptor in the TLR4 signalling pathway, in immune responses of the pancreatic duct during acute pancreatitis. METHODS Primary cultures of pancreatic duct epithelial cells from Wistar rats and cultures of the pancreatic ductal ARIP cell line were treated with lipopolysaccharide (LPS), and expression of toll-like receptor 4 mRNA was determined using real-time PCR, expression of MyD88 protein using Western blot, and levels of inflammatory cytokines using enzyme-linked immunosorbent assay. These experiments were repeated using ARIP cells in which MyD88 expression was stably knocked down. RESULTS Toll-like receptor 4 and MyD88 expression were similar between pancreatic duct epithelial cells and ARIP cells after LPS stimulation. Myeloid differentiation factor 88 knockdown led to significantly lower levels of inflammatory cytokines after LPS induction in ARIP cells. CONCLUSIONS Myeloid differentiation factor 88 knockdown attenuates LPS-induced inflammatory responses in pancreatic ductal cells, suggesting that the MyD88 pathway plays a critical role in their immune defense activity.
Collapse
|
186
|
Webster SJ, Ellis L, O'Brien LM, Tyrrell B, Fitzmaurice TJ, Elder MJ, Clare S, Chee R, Gaston JSH, Goodall JC. IRE1α mediates PKR activation in response to Chlamydia trachomatis infection. Microbes Infect 2016; 18:472-83. [PMID: 27021640 PMCID: PMC4936793 DOI: 10.1016/j.micinf.2016.03.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 02/09/2016] [Accepted: 03/18/2016] [Indexed: 12/20/2022]
Abstract
Protein kinase RNA activated (PKR) is a crucial mediator of anti-viral responses but is reported to be activated by multiple non-viral stimuli. However, mechanisms underlying PKR activation, particularly in response to bacterial infection, remain poorly understood. We have investigated mechanisms of PKR activation in human primary monocyte-derived dendritic cells in response to infection by Chlamydia trachomatis. Infection resulted in potent activation of PKR that was dependent on TLR4 and MyD88 signalling. NADPH oxidase was dispensable for activation of PKR as cells from chronic granulomatous disease (CGD) patients, or mice that lack NADPH oxidase activity, had equivalent or elevated PKR activation. Significantly, stimulation of cells with endoplasmic reticulum (ER) stress-inducing agents resulted in potent activation of PKR that was blocked by an inhibitor of IRE1α RNAse activity. Crucially, infection resulted in robust IRE1α RNAse activity that was dependent on TLR4 signalling and inhibition of IRE1α RNAse activity prevented PKR activation. Finally, we demonstrate that TLR4/IRE1α mediated PKR activation is required for the enhancement of interferon-β production following C. trachomatis infection. Thus, we provide evidence of a novel mechanism of PKR activation requiring ER stress signalling that occurs as a consequence of TLR4 stimulation during bacterial infection and contributes to inflammatory responses.
Collapse
Affiliation(s)
- Steve J Webster
- Rheumatology Research Group, Department of Medicine, University of Cambridge, UK
| | - Lou Ellis
- Rheumatology Research Group, Department of Medicine, University of Cambridge, UK
| | - Louise M O'Brien
- Rheumatology Research Group, Department of Medicine, University of Cambridge, UK
| | - Beatrice Tyrrell
- Rheumatology Research Group, Department of Medicine, University of Cambridge, UK
| | | | - Matthew J Elder
- Rheumatology Research Group, Department of Medicine, University of Cambridge, UK
| | - Simon Clare
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Ronnie Chee
- Department of Immunology, Royal Free Hospital, London, UK
| | - J S Hill Gaston
- Rheumatology Research Group, Department of Medicine, University of Cambridge, UK
| | - Jane C Goodall
- Rheumatology Research Group, Department of Medicine, University of Cambridge, UK.
| |
Collapse
|
187
|
Gomez de Aguero M, Ganal-Vonarburg SC, Fuhrer T, Rupp S, Uchimura Y, Li H, Steinert A, Heikenwalder M, Hapfelmeier S, Sauer U, McCoy KD, Macpherson AJ. The maternal microbiota drives early postnatal innate immune development. Science 2016; 351:1296-302. [DOI: 10.1126/science.aad2571] [Citation(s) in RCA: 696] [Impact Index Per Article: 77.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
188
|
Ferreira TPT, Mariano LL, Ghilosso-Bortolini R, de Arantes ACS, Fernandes AJ, Berni M, Cecchinato V, Uguccioni M, Maj R, Barberis A, Silva PMRE, Martins MA. Potential of PEGylated Toll-Like Receptor 7 Ligands for Controlling Inflammation and Functional Changes in Mouse Models of Asthma and Silicosis. Front Immunol 2016; 7:95. [PMID: 27014274 PMCID: PMC4786742 DOI: 10.3389/fimmu.2016.00095] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 02/26/2016] [Indexed: 01/06/2023] Open
Abstract
Prior investigations show that signaling activation through pattern recognition receptors can directly impact a number of inflammatory lung diseases. While toll-like receptor (TLR) 7 agonists have raised interest for their ability to inhibit allergen-induced pathological changes in experimental asthma conditions, the putative benefit of this treatment is limited by adverse effects. Our aim was to evaluate the therapeutic potential of two PEGylated purine-like compounds, TMX-302 and TMX-306, characterized by TLR7 partial agonistic activity; therefore, the compounds are expected to induce lower local and systemic adverse reactions. In vitro approaches and translation to murine models of obstructive and restrictive lung diseases were explored. In vitro studies with human PBMCs showed that both TMX-302 and TMX-306 marginally affects cytokine production as compared with equivalent concentrations of the TLR7 full agonist, TMX-202. The PEGylated compounds did not induce monocyte-derived DC maturation or B cell proliferation, differently from what observed after stimulation with TMX-202. Impact of PEGylated ligands on lung function and inflammatory changes was studied in animal models of acute lung injury, asthma, and silicosis following Lipopolysaccharide (LPS), allergen (ovalbumin), and silica inhalation, respectively. Subcutaneous injection of TMX-302 prevented LPS- and allergen-induced airway hyper-reactivity (AHR), leukocyte infiltration, and production of pro-inflammatory cytokines in the lung. However, intranasal instillation of TMX-302 led to neutrophil infiltration and failed to prevent allergen-induced AHR, despite inhibiting leukocyte counts in the BAL. Aerosolized TMX-306 given prophylactically, but not therapeutically, inhibited pivotal asthma features. Interventional treatment with intranasal instillation of TMX-306 significantly reduced the pulmonary fibrogranulomatous response and the number of silica particles in lung interstitial space in silicotic mice. These findings highlight the potential of TMX-306, emphasizing its value in drug development for lung diseases, and particularly silicosis.
Collapse
Affiliation(s)
| | - Lívia Lacerda Mariano
- Laboratory of Inflammation, Oswaldo Cruz Institute, FIOCRUZ , Rio de Janeiro , Brazil
| | | | | | | | - Michelle Berni
- Institute for Research in Biomedicine, Universitá della Svizzera Italiana , Bellinzona , Switzerland
| | - Valentina Cecchinato
- Institute for Research in Biomedicine, Universitá della Svizzera Italiana , Bellinzona , Switzerland
| | - Mariagrazia Uguccioni
- Institute for Research in Biomedicine, Universitá della Svizzera Italiana , Bellinzona , Switzerland
| | | | | | | | - Marco Aurélio Martins
- Laboratory of Inflammation, Oswaldo Cruz Institute, FIOCRUZ , Rio de Janeiro , Brazil
| |
Collapse
|
189
|
Barnea ER, Hayrabedyan S, Todorova K, Almogi-Hazan O, Or R, Guingab J, McElhinney J, Fernandez N, Barder T. PreImplantation factor (PIF*) regulates systemic immunity and targets protective regulatory and cytoskeleton proteins. Immunobiology 2016; 221:778-93. [PMID: 26944449 DOI: 10.1016/j.imbio.2016.02.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 02/08/2016] [Accepted: 02/14/2016] [Indexed: 12/29/2022]
Abstract
Secreted by viable embryos, PIF is expressed by the placenta and found in maternal circulation. It promotes implantation and trophoblast invasion, achieving systemic immune homeostasis. Synthetic PIF successfully transposes endogenous PIF features to non-pregnant immune and transplant models. PIF affects innate and activated PBMC cytokines and genes expression. We report that PIF targets similar proteins in CD14+, CD4+ and CD8+ cells instigating integrated immune regulation. PIF-affinity chromatography followed by mass-spectrometry, pathway and heatmap analysis reveals that SET-apoptosis inhibitor, vimentin, myosin-9 and calmodulin are pivotal for immune regulation. PIF acts on macrophages down-stream of LPS (lipopolysaccharide-bacterial antigen) CD14/TLR4/MD2 complex, targeting myosin-9, thymosin-α1 and 14-3-3eta. PIF mainly targets platelet aggregation in CD4+, and skeletal proteins in CD8+ cells. Pathway analysis demonstrates that PIF targets and regulates SET, tubulin, actin-b, and S100 genes expression. PIF targets systemic immunity and has a short circulating half-life. Collectively, PIF targets identified; protective, immune regulatory and cytoskeleton proteins reveal mechanisms involved in the observed efficacy against immune disorders.
Collapse
Affiliation(s)
- Eytan R Barnea
- The Society for the Investigation of Early Pregnancy (SIEP), Cherry Hill, NJ, USA; BioIncept LLC, Cherry Hill, NJ, USA.
| | - Soren Hayrabedyan
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Krassimira Todorova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Osnat Almogi-Hazan
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Reuven Or
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Joy Guingab
- Chemical Biology and Proteomics, Banyan Biomarkers, Alachua, FL, USA
| | - James McElhinney
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, UK
| | - Nelson Fernandez
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, UK
| | - Timothy Barder
- Research & Development, Eprogen, Inc., Downers Grove, IL, USA
| |
Collapse
|
190
|
Barrat FJ, Elkon KB, Fitzgerald KA. Importance of Nucleic Acid Recognition in Inflammation and Autoimmunity. Annu Rev Med 2016; 67:323-36. [DOI: 10.1146/annurev-med-052814-023338] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Franck J. Barrat
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021;
| | - Keith B. Elkon
- Division of Rheumatology, University of Washington, Seattle, Washington 98109
| | - Katherine A. Fitzgerald
- Program in Innate Immunity, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| |
Collapse
|
191
|
Sun J, Li N, Oh KS, Dutta B, Vayttaden SJ, Lin B, Ebert TS, De Nardo D, Davis J, Bagirzadeh R, Lounsbury NW, Pasare C, Latz E, Hornung V, Fraser IDC. Comprehensive RNAi-based screening of human and mouse TLR pathways identifies species-specific preferences in signaling protein use. Sci Signal 2016; 9:ra3. [PMID: 26732763 PMCID: PMC5381726 DOI: 10.1126/scisignal.aab2191] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Toll-like receptors (TLRs) are a major class of pattern recognition receptors, which mediate the responses of innate immune cells to microbial stimuli. To systematically determine the roles of proteins in canonical TLR signaling pathways, we conducted an RNA interference (RNAi)-based screen in human and mouse macrophages. We observed a pattern of conserved signaling module dependencies across species, but found notable species-specific requirements at the level of individual proteins. Among these, we identified unexpected differences in the involvement of members of the interleukin-1 receptor-associated kinase (IRAK) family between the human and mouse TLR pathways. Whereas TLR signaling in mouse macrophages depended primarily on IRAK4 and IRAK2, with little or no role for IRAK1, TLR signaling and proinflammatory cytokine production in human macrophages depended on IRAK1, with knockdown of IRAK4 or IRAK2 having less of an effect. Consistent with species-specific roles for these kinases, IRAK4 orthologs failed to rescue signaling in IRAK4-deficient macrophages from the other species, and only mouse macrophages required the kinase activity of IRAK4 to mediate TLR responses. The identification of a critical role for IRAK1 in TLR signaling in humans could potentially explain the association of IRAK1 with several autoimmune diseases. Furthermore, this study demonstrated how systematic screening can be used to identify important characteristics of innate immune responses across species, which could optimize therapeutic targeting to manipulate human TLR-dependent outputs.
Collapse
Affiliation(s)
- Jing Sun
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ning Li
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kyu-Seon Oh
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bhaskar Dutta
- Bioinformatics Team, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sharat J Vayttaden
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bin Lin
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thomas S Ebert
- Institute of Molecular Medicine, University Hospital, University of Bonn, 53127 Bonn, Germany
| | - Dominic De Nardo
- Institute of Innate Immunity, University Hospital, Biomedical Centre, University of Bonn, 53127 Bonn, Germany. Inflammation Division, Walter and Eliza Hall Institute, Parkville,Victoria 3052, Australia. Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Joie Davis
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rustam Bagirzadeh
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Nicolas W Lounsbury
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chandrashekhar Pasare
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital, Biomedical Centre, University of Bonn, 53127 Bonn, Germany. German Center for Neurodegenerative Diseases (DZNE), 53175 Bonn, Germany. Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Veit Hornung
- Institute of Molecular Medicine, University Hospital, University of Bonn, 53127 Bonn, Germany
| | - Iain D C Fraser
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
192
|
Zong X, Song D, Wang T, Xia X, Hu W, Han F, Wang Y. LFP-20, a porcine lactoferrin peptide, ameliorates LPS-induced inflammation via the MyD88/NF-κB and MyD88/MAPK signaling pathways. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2015; 52:123-131. [PMID: 26003437 DOI: 10.1016/j.dci.2015.05.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/17/2015] [Accepted: 05/18/2015] [Indexed: 06/04/2023]
Abstract
LFP-20 is one of the 20 amino acid anti-microbial peptides identified in the N terminus of porcine lactoferrin. Apart from its extensively studied direct anti-bacterial activity, its potential as an activator of immune-related cellular functions is unknown. Therefore, this study investigated its anti-inflammatory effects in lipopolysaccharide (LPS)-stimulated pig alveolar macrophages in vitro and systemic inflammation in an in vivo mouse model. We found that the inhibitory effects of LFP-20 on production of pro-inflammatory cytokines were independent of its LPS-binding activity. However, they were associated with NF-κB and MAPK-dependent signaling. Furthermore, LFP-20 might directly influence MyD88 levels to block its interaction with NF-κB and MAPK-dependent signaling molecules that might alter LPS-mediated inflammatory responses in activated macrophages. Taken together, our data indicated that LFP-20 prevents the LPS-induced inflammatory response by inhibiting MyD88/NF-κB and MyD88/MAPK signaling pathways, and sheds light on the potential use of LFP-20 in the therapy of LPS-mediated sepsis.
Collapse
Affiliation(s)
- Xin Zong
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Deguang Song
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tenghao Wang
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xi Xia
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wangyang Hu
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Feifei Han
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yizhen Wang
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
193
|
Hotz C, Roetzer LC, Huber T, Sailer A, Oberson A, Treinies M, Heidegger S, Herbst T, Endres S, Bourquin C. TLR and RLR Signaling Are Reprogrammed in Opposite Directions after Detection of Viral Infection. THE JOURNAL OF IMMUNOLOGY 2015; 195:4387-95. [PMID: 26392465 DOI: 10.4049/jimmunol.1500079] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 08/18/2015] [Indexed: 12/25/2022]
Abstract
Innate immune recognition of RNA is key for the initiation of immunity in response to viral infection. Although the factors controlling the detection of viral RNA by innate immune receptors in host cells are increasingly well understood, little is known about the dynamic changes in signaling after the initial triggering of these receptors. In this study, we report that preconditioning with the synthetic dsRNA polyinosinic-polycytidylic acid [poly(I:C)], a mimetic of viral RNA, rapidly reprograms murine APCs by simultaneously augmenting sensitivity of endosomal TLRs and inhibiting activation of RIG-I-like receptors (RLRs) in an IFN-β-dependent manner. These changes in receptor sensitivity were also seen in vivo after treatment of mice with poly(I:C). Mechanistically, the increased sensitivity of the TLR pathway was associated with elevated MAPK and NF-κB activity. The RLR response was inhibited downstream of TANK-binding kinase-1, resulting in decreased IFN regulatory factor 3 phosphorylation. Reprogramming of pattern-recognition receptor signaling also occurred after viral infection, because infection of host cells with Sendai virus or their exposure to supernatant from virus-infected cells induced the same changes in TLR and RLR sensitivity as poly(I:C). Thus, innate recognition of viral infection critically modifies responses to pattern-recognition receptor stimulation. These dynamic adaptations to infection may reinforce antiviral immunity and at the same time serve to limit pathological inflammation.
Collapse
Affiliation(s)
- Christian Hotz
- Department of Medicine, Faculty of Science, University of Fribourg, 1700 Fribourg, Switzerland; Division of Clinical Pharmacology, Center for Integrated Protein Science Munich, Ludwig-Maximilian-University Munich, 80336 Munich, Germany; and
| | - Laurin C Roetzer
- Division of Clinical Pharmacology, Center for Integrated Protein Science Munich, Ludwig-Maximilian-University Munich, 80336 Munich, Germany; and
| | - Thomas Huber
- Division of Clinical Pharmacology, Center for Integrated Protein Science Munich, Ludwig-Maximilian-University Munich, 80336 Munich, Germany; and
| | - Andreas Sailer
- Division of Clinical Pharmacology, Center for Integrated Protein Science Munich, Ludwig-Maximilian-University Munich, 80336 Munich, Germany; and
| | - Anne Oberson
- Department of Medicine, Faculty of Science, University of Fribourg, 1700 Fribourg, Switzerland
| | - Marina Treinies
- Department of Medicine, Faculty of Science, University of Fribourg, 1700 Fribourg, Switzerland
| | - Simon Heidegger
- Division of Clinical Pharmacology, Center for Integrated Protein Science Munich, Ludwig-Maximilian-University Munich, 80336 Munich, Germany; and III Medical Department, Technical University Munich, 81675 Munich, Germany
| | - Tina Herbst
- Department of Medicine, Faculty of Science, University of Fribourg, 1700 Fribourg, Switzerland
| | - Stefan Endres
- Division of Clinical Pharmacology, Center for Integrated Protein Science Munich, Ludwig-Maximilian-University Munich, 80336 Munich, Germany; and
| | - Carole Bourquin
- Department of Medicine, Faculty of Science, University of Fribourg, 1700 Fribourg, Switzerland; Division of Clinical Pharmacology, Center for Integrated Protein Science Munich, Ludwig-Maximilian-University Munich, 80336 Munich, Germany; and
| |
Collapse
|
194
|
Abstract
The Toll/interleukin-1 receptor/resistance protein (TIR) domain is a protein-protein interaction domain consisting of 125-200 residues, widely distributed in animals, plants and bacteria but absent from fungi, archea and viruses. In plants and animals, these domains are found in proteins with functions in innate immune pathways, while in bacteria, some TIR domain-containing proteins interfere with the innate immune pathways in the host. TIR domains function as protein scaffolds, mostly involving self-association and homotypic interactions with other TIR domains. In the last 15 years, the three-dimensional structures of TIR domains from several mammalian, plant and bacterial proteins have been reported. These structures, jointly with functional data including the identification of interacting proteins, have started to provide insight into the molecular basis of the assembly of animal and plant immune signaling complexes, and for host immunosuppression by bacterial pathogens. This review focuses on the current knowledge of the structures of the TIR domains and how the structure relates to function.
Collapse
|
195
|
Németh B, Doczi J, Csete D, Kacso G, Ravasz D, Adams D, Kiss G, Nagy AM, Horvath G, Tretter L, Mócsai A, Csépányi-Kömi R, Iordanov I, Adam-Vizi V, Chinopoulos C. Abolition of mitochondrial substrate-level phosphorylation by itaconic acid produced by LPS-induced Irg1 expression in cells of murine macrophage lineage. FASEB J 2015; 30:286-300. [PMID: 26358042 DOI: 10.1096/fj.15-279398] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 08/31/2015] [Indexed: 01/28/2023]
Abstract
Itaconate is a nonamino organic acid exhibiting antimicrobial effects. It has been recently identified in cells of macrophage lineage as a product of an enzyme encoded by immunoresponsive gene 1 (Irg1), acting on the citric acid cycle intermediate cis-aconitate. In mitochondria, itaconate can be converted by succinate-coenzyme A (CoA) ligase to itaconyl-CoA at the expense of ATP (or GTP), and is also a weak competitive inhibitor of complex II. Here, we investigated specific bioenergetic effects of increased itaconate production mediated by LPS-induced stimulation of Irg1 in murine bone marrow-derived macrophages (BMDM) and RAW-264.7 cells. In rotenone-treated macrophage cells, stimulation by LPS led to impairment in substrate-level phosphorylation (SLP) of in situ mitochondria, deduced by a reversal in the directionality of the adenine nucleotide translocase operation. In RAW-264.7 cells, the LPS-induced impairment in SLP was reversed by short-interfering RNA(siRNA)-but not scrambled siRNA-treatment directed against Irg1. LPS dose-dependently inhibited oxygen consumption rates (61-91%) and elevated glycolysis rates (>21%) in BMDM but not RAW-264.7 cells, studied under various metabolic conditions. In isolated mouse liver mitochondria treated with rotenone, itaconate dose-dependently (0.5-2 mM) reversed the operation of adenine nucleotide translocase, implying impairment in SLP, an effect that was partially mimicked by malonate. However, malonate yielded greater ADP-induced depolarizations (3-19%) than itaconate. We postulate that itaconate abolishes SLP due to 1) a "CoA trap" in the form of itaconyl-CoA that negatively affects the upstream supply of succinyl-CoA from the α-ketoglutarate dehydrogenase complex; 2) depletion of ATP (or GTP), which are required for the thioesterification by succinate-CoA ligase; and 3) inhibition of complex II leading to a buildup of succinate which shifts succinate-CoA ligase equilibrium toward ATP (or GTP) utilization. Our results support the notion that Irg1-expressing cells of macrophage lineage lose the capacity of mitochondrial SLP for producing itaconate during mounting of an immune defense.
Collapse
Affiliation(s)
- Beáta Németh
- *Department of Medical Biochemistry and Department of Physiology, Semmelweis University, Budapest, Hungary; and Lendület Neurobiochemistry Research Group, Lendület Inflammation Physiology Research Group, Laboratory for Neurobiochemistry, and Lendület Ion Channel Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Judit Doczi
- *Department of Medical Biochemistry and Department of Physiology, Semmelweis University, Budapest, Hungary; and Lendület Neurobiochemistry Research Group, Lendület Inflammation Physiology Research Group, Laboratory for Neurobiochemistry, and Lendület Ion Channel Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Dániel Csete
- *Department of Medical Biochemistry and Department of Physiology, Semmelweis University, Budapest, Hungary; and Lendület Neurobiochemistry Research Group, Lendület Inflammation Physiology Research Group, Laboratory for Neurobiochemistry, and Lendület Ion Channel Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Gergely Kacso
- *Department of Medical Biochemistry and Department of Physiology, Semmelweis University, Budapest, Hungary; and Lendület Neurobiochemistry Research Group, Lendület Inflammation Physiology Research Group, Laboratory for Neurobiochemistry, and Lendület Ion Channel Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Dora Ravasz
- *Department of Medical Biochemistry and Department of Physiology, Semmelweis University, Budapest, Hungary; and Lendület Neurobiochemistry Research Group, Lendület Inflammation Physiology Research Group, Laboratory for Neurobiochemistry, and Lendület Ion Channel Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Daniel Adams
- *Department of Medical Biochemistry and Department of Physiology, Semmelweis University, Budapest, Hungary; and Lendület Neurobiochemistry Research Group, Lendület Inflammation Physiology Research Group, Laboratory for Neurobiochemistry, and Lendület Ion Channel Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Gergely Kiss
- *Department of Medical Biochemistry and Department of Physiology, Semmelweis University, Budapest, Hungary; and Lendület Neurobiochemistry Research Group, Lendület Inflammation Physiology Research Group, Laboratory for Neurobiochemistry, and Lendület Ion Channel Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Adam M Nagy
- *Department of Medical Biochemistry and Department of Physiology, Semmelweis University, Budapest, Hungary; and Lendület Neurobiochemistry Research Group, Lendület Inflammation Physiology Research Group, Laboratory for Neurobiochemistry, and Lendület Ion Channel Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Gergo Horvath
- *Department of Medical Biochemistry and Department of Physiology, Semmelweis University, Budapest, Hungary; and Lendület Neurobiochemistry Research Group, Lendület Inflammation Physiology Research Group, Laboratory for Neurobiochemistry, and Lendület Ion Channel Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Laszlo Tretter
- *Department of Medical Biochemistry and Department of Physiology, Semmelweis University, Budapest, Hungary; and Lendület Neurobiochemistry Research Group, Lendület Inflammation Physiology Research Group, Laboratory for Neurobiochemistry, and Lendület Ion Channel Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Attila Mócsai
- *Department of Medical Biochemistry and Department of Physiology, Semmelweis University, Budapest, Hungary; and Lendület Neurobiochemistry Research Group, Lendület Inflammation Physiology Research Group, Laboratory for Neurobiochemistry, and Lendület Ion Channel Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Roland Csépányi-Kömi
- *Department of Medical Biochemistry and Department of Physiology, Semmelweis University, Budapest, Hungary; and Lendület Neurobiochemistry Research Group, Lendület Inflammation Physiology Research Group, Laboratory for Neurobiochemistry, and Lendület Ion Channel Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Iordan Iordanov
- *Department of Medical Biochemistry and Department of Physiology, Semmelweis University, Budapest, Hungary; and Lendület Neurobiochemistry Research Group, Lendület Inflammation Physiology Research Group, Laboratory for Neurobiochemistry, and Lendület Ion Channel Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Vera Adam-Vizi
- *Department of Medical Biochemistry and Department of Physiology, Semmelweis University, Budapest, Hungary; and Lendület Neurobiochemistry Research Group, Lendület Inflammation Physiology Research Group, Laboratory for Neurobiochemistry, and Lendület Ion Channel Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Christos Chinopoulos
- *Department of Medical Biochemistry and Department of Physiology, Semmelweis University, Budapest, Hungary; and Lendület Neurobiochemistry Research Group, Lendület Inflammation Physiology Research Group, Laboratory for Neurobiochemistry, and Lendület Ion Channel Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| |
Collapse
|
196
|
TIR Domain-Containing Adapter-Inducing Beta Interferon (TRIF) Mediates Immunological Memory against Bacterial Pathogens. Infect Immun 2015; 83:4404-15. [PMID: 26351279 DOI: 10.1128/iai.00674-15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 08/26/2015] [Indexed: 01/08/2023] Open
Abstract
Induction of adaptive immunity leads to the establishment of immunological memory; however, how innate immunity regulates memory T cell function remains obscure. Here we show a previously undefined mechanism in which innate and adaptive immunity are linked by TIR domain-containing adapter-inducing beta interferon (TRIF) during establishment and reactivation of memory T cells against Gram-negative enteropathogens. Absence of TRIF in macrophages (Mϕs) but not dendritic cells led to a predominant generation of CD4(+) central memory T cells that express IL-17 during enteric bacterial infection in mice. TRIF-dependent type I interferon (IFN) signaling in T cells was essential to Th1 lineage differentiation and reactivation of memory T cells. TRIF activated memory T cells to facilitate local neutrophil influx and enhance bacterial elimination. These results highlight the importance of TRIF as a mediator of the innate and adaptive immune interactions in achieving the protective properties of memory immunity against Gram-negative bacteria and suggest TRIF as a potential therapeutic target.
Collapse
|
197
|
Iribarren K, Bloy N, Buqué A, Cremer I, Eggermont A, Fridman WH, Fucikova J, Galon J, Špíšek R, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Immunostimulation with Toll-like receptor agonists in cancer therapy. Oncoimmunology 2015; 5:e1088631. [PMID: 27141345 DOI: 10.1080/2162402x.2015.1088631] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 08/25/2015] [Indexed: 12/19/2022] Open
Abstract
Accumulating preclinical evidence indicates that Toll-like receptor (TLR) agonists efficiently boost tumor-targeting immune responses (re)initiated by most, if not all, paradigms of anticancer immunotherapy. Moreover, TLR agonists have been successfully employed to ameliorate the efficacy of various chemotherapeutics and targeted anticancer agents, at least in rodent tumor models. So far, only three TLR agonists have been approved by regulatory agencies for use in cancer patients. Moreover, over the past decade, the interest of scientists and clinicians in these immunostimulatory agents has been fluctuating. Here, we summarize recent advances in the preclinical and clinical development of TLR agonists for cancer therapy.
Collapse
Affiliation(s)
- Kristina Iribarren
- INSERM, U1138, Paris, France; Equipe 13, Center de Recherche des Cordeliers, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Norma Bloy
- INSERM, U1138, Paris, France; Gustave Roussy Cancer Campus, Villejuif, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France
| | - Aitziber Buqué
- INSERM, U1138, Paris, France; Gustave Roussy Cancer Campus, Villejuif, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France
| | - Isabelle Cremer
- INSERM, U1138, Paris, France; Equipe 13, Center de Recherche des Cordeliers, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France
| | | | - Wolf Hervé Fridman
- INSERM, U1138, Paris, France; Equipe 13, Center de Recherche des Cordeliers, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Jitka Fucikova
- Sotio, Prague, Czech Republic; Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Jérôme Galon
- INSERM, U1138, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Laboratory of Integrative Cancer Immunology, Center de Recherche des Cordeliers, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
| | - Radek Špíšek
- Sotio, Prague, Czech Republic; Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Villejuif, France; INSERM, U1015, CICBT507, Villejuif, France
| | - Guido Kroemer
- INSERM, U1138, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center 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, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France; Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- INSERM, U1138, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Gustave Roussy Cancer Campus, Villejuif, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
198
|
An T, Li S, Pan W, Tien P, Zhong B, Shu HB, Wu S. DYRK2 Negatively Regulates Type I Interferon Induction by Promoting TBK1 Degradation via Ser527 Phosphorylation. PLoS Pathog 2015; 11:e1005179. [PMID: 26407194 PMCID: PMC4583546 DOI: 10.1371/journal.ppat.1005179] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 09/01/2015] [Indexed: 12/04/2022] Open
Abstract
Viral infection activates the transcription factors NF-κB and IRF3, which contribute to the induction of type I interferons (IFNs) and cellular antiviral responses. Protein kinases play a critical role in various signaling pathways by phosphorylating their substrates. Here, we identified dual-specificity tyrosine-(Y)-phosphorylation-regulated kinase 2 (DYRK2) as a negative regulator of virus-triggered type I IFN induction. DYRK2 inhibited the virus-triggered induction of type I IFNs and promoted the K48-linked ubiquitination and degradation of TANK-binding kinase 1 (TBK1) in a kinase-activity-dependent manner. We further found that DYRK2 phosphorylated Ser527 of TBK1, which is essential for the recruitment of NLRP4 and for the E3 ubiquitin ligase DTX4 to degrade TBK1. These findings suggest that DYRK2 negatively regulates virus-triggered signaling by targeting TBK1 for phosphorylation and priming it for degradation, and these data provide new insights into the molecular mechanisms that dictate the cellular antiviral response.
Collapse
Affiliation(s)
- Tai An
- The College of Life Sciences, State Key Laboratory of Virology, Modern Virology Research Center, Wuhan University, Wuhan, China
- The College of Basic Medical Science, Shaanxi University of Chinese Medicine, Xi’an, China
| | - Shu Li
- The College of Life Sciences, State Key Laboratory of Virology, Modern Virology Research Center, Wuhan University, Wuhan, China
| | - Wei Pan
- The College of Life Sciences, State Key Laboratory of Virology, Modern Virology Research Center, Wuhan University, Wuhan, China
| | - Po Tien
- The College of Life Sciences, State Key Laboratory of Virology, Modern Virology Research Center, Wuhan University, Wuhan, China
| | - Bo Zhong
- The College of Life Sciences, State Key Laboratory of Virology, Modern Virology Research Center, Wuhan University, Wuhan, China
| | - Hong-Bing Shu
- The College of Life Sciences, State Key Laboratory of Virology, Modern Virology Research Center, Wuhan University, Wuhan, China
| | - Shuwen Wu
- The College of Life Sciences, State Key Laboratory of Virology, Modern Virology Research Center, Wuhan University, Wuhan, China
| |
Collapse
|
199
|
Singh MV, Cicha MZ, Meyerholz DK, Chapleau MW, Abboud FM. Dual Activation of TRIF and MyD88 Adaptor Proteins by Angiotensin II Evokes Opposing Effects on Pressure, Cardiac Hypertrophy, and Inflammatory Gene Expression. Hypertension 2015. [PMID: 26195481 DOI: 10.1161/hypertensionaha.115.06011] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hypertension is recognized as an immune disorder whereby immune cells play a defining role in the genesis and progression of the disease. The innate immune system and its component toll-like receptors are key determinants of the immunologic outcome through their proinflammatory response. Toll-like receptor-activated signaling pathways use several adaptor proteins of which adaptor proteins myeloid differentiation protein 88 (MyD88) and toll-interleukin receptor domain-containing adaptor protein-inducing interferon-β (TRIF) define 2 major inflammatory pathways. In this study, we compared the contributions of MyD88 and TRIF adaptor proteins to angiotensin II (Ang II)-induced hypertension and cardiac hypertrophy in mice. Deletion of MyD88 did not prevent cardiac hypertrophy and the pressor response to Ang II tended to increase. Moreover, the increase in inflammatory gene expression (Tnfa, Nox4, and Agtr1a) was significantly greater in the heart and kidney of MyD88-deficient mice when compared with wild-type mice. Thus, pathways involving MyD88 may actually restrain the inflammatory responses. However, in mice with nonfunctional TRIF (Trif(mut) mice), Ang II-induced hypertension and cardiac hypertrophy were abrogated, and proinflammatory gene expression in heart and kidneys was unchanged or decreased. Our results indicate that Ang II induces activation of a proinflammatory innate immune response, causing hypertension and cardiac hypertrophy. These effects require functional adaptor protein TRIF-mediated pathways. However, the common MyD88-dependent signaling pathway, which is also activated simultaneously by Ang II, paradoxically exerts a negative regulatory influence on these responses.
Collapse
Affiliation(s)
- Madhu V Singh
- From the Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine (M.V.S., M.W.C., F.M.A.), Department of Molecular Physiology and Biophysics, Carver College of Medicine (M.W.C., F.M.A.), and Department of Pathology (D.K.M.), University of Iowa, Iowa City; and Department of Veterans Affairs Medical Center, Iowa City, IA (M.Z.C., M.W.C.).
| | - Michael Z Cicha
- From the Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine (M.V.S., M.W.C., F.M.A.), Department of Molecular Physiology and Biophysics, Carver College of Medicine (M.W.C., F.M.A.), and Department of Pathology (D.K.M.), University of Iowa, Iowa City; and Department of Veterans Affairs Medical Center, Iowa City, IA (M.Z.C., M.W.C.)
| | - David K Meyerholz
- From the Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine (M.V.S., M.W.C., F.M.A.), Department of Molecular Physiology and Biophysics, Carver College of Medicine (M.W.C., F.M.A.), and Department of Pathology (D.K.M.), University of Iowa, Iowa City; and Department of Veterans Affairs Medical Center, Iowa City, IA (M.Z.C., M.W.C.)
| | - Mark W Chapleau
- From the Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine (M.V.S., M.W.C., F.M.A.), Department of Molecular Physiology and Biophysics, Carver College of Medicine (M.W.C., F.M.A.), and Department of Pathology (D.K.M.), University of Iowa, Iowa City; and Department of Veterans Affairs Medical Center, Iowa City, IA (M.Z.C., M.W.C.)
| | - François M Abboud
- From the Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine (M.V.S., M.W.C., F.M.A.), Department of Molecular Physiology and Biophysics, Carver College of Medicine (M.W.C., F.M.A.), and Department of Pathology (D.K.M.), University of Iowa, Iowa City; and Department of Veterans Affairs Medical Center, Iowa City, IA (M.Z.C., M.W.C.).
| |
Collapse
|
200
|
Nair S, Arora S, Lim JY, Lee LH, Lim LH. The regulation of TNFα production after heat and endotoxin stimulation is dependent on Annexin-A1 and HSP70. Cell Stress Chaperones 2015; 20:583-93. [PMID: 25753354 PMCID: PMC4463914 DOI: 10.1007/s12192-015-0580-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 02/15/2015] [Accepted: 02/16/2015] [Indexed: 10/23/2022] Open
Abstract
Febrile temperatures can induce stress responses which protect cells from damage and can reduce inflammation during infections and sepsis. However, the mechanisms behind the protective functions of heat in response to the bacterial endotoxin LPS are unclear. We have recently shown that Annexin-1 (ANXA1)-deficient macrophages exhibited higher TNFα levels after LPS stimulation. Moreover, we have previously reported that ANXA1 can function as a stress protein. Therefore in this study, we determined if ANXA1 is involved in the protective effects of heat on cytokine levels in macrophages after heat and LPS. Exposure of macrophages to 42 °C for 1 h prior to LPS results in an inhibition of TNFα production, which was not evident in ANXA1(-/-) macrophages. We show that this regulation involves primarily MYD88-independent pathways. ANXA1 regulates TNFα mRNA stability after heat and LPS, and this is dependent on endogenous ANXA1 expression and not exogenously secreted factors. Further mechanistic studies revealed the possible involvement of the heat shock protein HSP70 and JNK in the heat and inflammatory stress response regulated by ANXA1. This study shows that ANXA1, an immunomodulatory protein, is critical in the heat stress response induced after heat and endotoxin stimulation.
Collapse
Affiliation(s)
- Sunitha Nair
- />Department of Physiology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 28 Medical Drive, Singapore, 117456 Singapore
- />NUS Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456 Singapore
| | - Suruchi Arora
- />Department of Physiology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 28 Medical Drive, Singapore, 117456 Singapore
- />NUS Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456 Singapore
| | - Jyue Yuan Lim
- />Singapore Immunology Network (SigN), 8A Biomedical Grove, Immunos, Singapore, 138648 Singapore
| | - Lay Hoon Lee
- />Department of Physiology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 28 Medical Drive, Singapore, 117456 Singapore
- />NUS Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456 Singapore
| | - Lina H.K. Lim
- />Department of Physiology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 28 Medical Drive, Singapore, 117456 Singapore
- />NUS Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456 Singapore
| |
Collapse
|