51
|
Boyle JP, Parkhouse R, Monie TP. Insights into the molecular basis of the NOD2 signalling pathway. Open Biol 2015; 4:rsob.140178. [PMID: 25520185 PMCID: PMC4281710 DOI: 10.1098/rsob.140178] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The cytosolic pattern recognition receptor NOD2 is activated by the peptidoglycan fragment muramyl dipeptide to generate a proinflammatory immune response. Downstream effects include the secretion of cytokines such as interleukin 8, the upregulation of pro-interleukin 1β, the induction of autophagy, the production of antimicrobial peptides and defensins, and contributions to the maintenance of the composition of the intestinal microbiota. Polymorphisms in NOD2 are the cause of the inflammatory disorder Blau syndrome and act as susceptibility factors for the inflammatory bowel condition Crohn's disease. The complexity of NOD2 signalling is highlighted by the observation that over 30 cellular proteins interact with NOD2 directly and influence or regulate its functional activity. Previously, the majority of reviews on NOD2 function have focused upon the role of NOD2 in inflammatory disease or in its interaction with and response to microbes. However, the functionality of NOD2 is underpinned by its biochemical interactions. Consequently, in this review, we have taken the opportunity to address the more ‘basic’ elements of NOD2 signalling. In particular, we have focused upon the core interactions of NOD2 with protein factors that influence and modulate the signal transduction pathways involved in NOD2 signalling. Further, where information exists, such as in relation to the role of RIP2, we have drawn comparison with the closely related, but functionally discrete, pattern recognition receptor NOD1. Overall, we provide a comprehensive resource targeted at understanding the complexities of NOD2 signalling.
Collapse
Affiliation(s)
- Joseph P Boyle
- Department of Biochemistry, University of Cambridge, Cambridge, UK Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | | | - Tom P Monie
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK MRC Human Nutrition Research, Elsie Widdowson Laboratory, 120 Fulbourn Road, Cambridge, UK
| |
Collapse
|
52
|
Bolshakova A, Magnusson KE, Pinaev G, Petukhova O. Functional compartmentalisation of NF-kB-associated proteins in A431 cells. Cell Biol Int 2015; 37:387-96. [PMID: 23408724 DOI: 10.1002/cbin.10053] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 01/10/2013] [Indexed: 12/30/2022]
Abstract
NF-kB proteins belong to a family of ubiquitous transcription factors involved in a number of cellular responses. While the pathways of NF-kB activation and input into the regulation of gene activity have been comprehensively investigated, its cytoplasmic functions are poorly understood. In this study we addressed effects of the compartmentalisation of NF-kB proteins RelA/p65 and p50 in relation to the inhibitor IkB-a, using fibronectin (FN) and epidermal growth factor (EGF) for environmental stimulation of epidermoid carcinoma A431 cells. We thus assessed the presence of NF-kB family proteins in the cytosol, membrane, nuclear and cytoskeletal fractions with a special attention to the cytoskeletal fraction to define whether NFkB was active or not. Sub-cellular fractionation demonstrated that the proportion of RelA/p65 differed in diverse sub-cellular fractions, and that the cytoskeleton harboured about 7% thereof. Neither the nuclear nor the cytoskeleton fraction did contain IkB-a. The cytoskeleton binding of RelA/p65 and p50 was further confirmed by co-localisation and electron microscopy data. During 30-min EGF stimulation similar dynamics were found for RelA/p65 and IkB-a in the cytosol, RelA/p65 and p50 in the nucleus and p50 and IkB-a in the membrane. Furthermore, EGF stimulation for 30 min resulted in a threefold accumulation of RelA/p65 in cytoskeletal fraction. Our results suggest that nuclear-, membrane- and cytoskeleton-associated NF-kB are dynamic and comprise active pools, whereas the cytoplasmic is more constant and likely non-active due to the presence of IkB-a. Moreover, we discovered the existence of a dynamic, IkB-a-free pool of RelA/p65 associated with cytoskeletal fraction, what argues for a special regulatory role of the cytoskeleton in NF-kB stimulation.
Collapse
Affiliation(s)
- Anastasia Bolshakova
- Department of Cell Cultures, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, St. Petersburg 194064, Russian Federation
| | | | | | | |
Collapse
|
53
|
Rhodes DM, Smith SA, Holcombe M, Qwarnstrom EE. Computational Modelling of NF-κB Activation by IL-1RI and Its Co-Receptor TILRR, Predicts a Role for Cytoskeletal Sequestration of IκBα in Inflammatory Signalling. PLoS One 2015; 10:e0129888. [PMID: 26110282 PMCID: PMC4482363 DOI: 10.1371/journal.pone.0129888] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 05/14/2015] [Indexed: 11/19/2022] Open
Abstract
The transcription factor NF-κB (nuclear factor kappa B) is activated by Toll-like receptors and controlled by mechanotransduction and changes in the cytoskeleton. In this study we combine 3-D predictive protein modelling and in vitro experiments with in silico simulations to determine the role of the cytoskeleton in regulation of NF-κB. Simulations used a comprehensive agent-based model of the NF-κB pathway, which includes the type 1 IL-1 receptor (IL-1R1) complex and signalling intermediates, as well as cytoskeletal components. Agent based modelling relies on in silico reproductions of systems through the interactions of its components, and provides a reliable tool in investigations of biological processes, which require spatial considerations and involve complex formation and translocation of regulatory components. We show that our model faithfully reproduces the multiple steps comprising the NF-κB pathway, and provides a framework from which we can explore novel aspects of the system. The analysis, using 3-D predictive protein modelling and in vitro assays, demonstrated that the NF-κB inhibitor, IκBα is sequestered to the actin/spectrin complex within the cytoskeleton of the resting cell, and released during IL-1 stimulation, through a process controlled by the IL-1RI co-receptor TILRR (Toll-like and IL-1 receptor regulator). In silico simulations using the agent-based model predict that the cytoskeletal pool of IκBα is released to adjust signal amplification in relation to input levels. The results suggest that the process provides a mechanism for signal calibration and enables efficient, activation-sensitive regulation of NF-κB and inflammatory responses.
Collapse
Affiliation(s)
- David M. Rhodes
- Department of Cardiovascular Science, Medical School, University of Sheffield, United Kingdom
- Department of Computer Science, University of Sheffield, Sheffield, United Kingdom
| | - Sarah A. Smith
- Department of Cardiovascular Science, Medical School, University of Sheffield, United Kingdom
| | - Mike Holcombe
- Department of Computer Science, University of Sheffield, Sheffield, United Kingdom
| | - Eva E. Qwarnstrom
- Department of Cardiovascular Science, Medical School, University of Sheffield, United Kingdom
- * E-mail:
| |
Collapse
|
54
|
Tir Triggers Expression of CXCL1 in Enterocytes and Neutrophil Recruitment during Citrobacter rodentium Infection. Infect Immun 2015; 83:3342-54. [PMID: 26077760 DOI: 10.1128/iai.00291-15] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/09/2015] [Indexed: 12/28/2022] Open
Abstract
The hallmarks of enteropathogenic Escherichia coli (EPEC) infection are formation of attaching and effacing (A/E) lesions on mucosal surfaces and actin-rich pedestals on cultured cells, both of which are dependent on the type III secretion system effector Tir. Following translocation into cultured cells and clustering by intimin, Tir Y474 is phosphorylated, leading to recruitment of Nck, activation of N-WASP, and actin polymerization via the Arp2/3 complex. A secondary, weak, actin polymerization pathway is triggered via an NPY motif (Y454). Importantly, Y454 and Y474 play no role in A/E lesion formation on mucosal surfaces following infection with the EPEC-like mouse pathogen Citrobacter rodentium. In this study, we investigated the roles of Tir segments located upstream of Y451 and downstream of Y471 in C. rodentium colonization and A/E lesion formation. We also tested the role that Tir residues Y451 and Y471 play in host immune responses to C. rodentium infection. We found that deletion of amino acids 382 to 462 or 478 to 547 had no impact on the ability of Tir to mediate A/E lesion formation, although deletion of amino acids 478 to 547 affected Tir translocation. Examination of enterocytes isolated from infected mice revealed that a C. rodentium strain expressing Tir_Y451A/Y471A recruited significantly fewer neutrophils to the colon and triggered less colonic hyperplasia on day 14 postinfection than the wild-type strain. Consistently, enterocytes isolated from mice infected with C. rodentium expressing Tir_Y451A/Y471A expressed significantly less CXCL1. These result show that Tir-induced actin remodeling plays a direct role in modulation of immune responses to C. rodentium infection.
Collapse
|
55
|
Zheng K, Kitazato K, Wang Y, He Z. Pathogenic microbes manipulate cofilin activity to subvert actin cytoskeleton. Crit Rev Microbiol 2015; 42:677-95. [PMID: 25853495 DOI: 10.3109/1040841x.2015.1010139] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Actin-depolymerizing factor (ADF)/cofilin proteins are key players in controlling the temporal and spatial extent of actin dynamics, which is crucial for mediating host-pathogen interactions. Pathogenic microbes have evolved molecular mechanisms to manipulate cofilin activity to subvert the actin cytoskeletal system in host cells, promoting their internalization into the target cells, modifying the replication niche and facilitating their intracellular and intercellular dissemination. The study of how these pathogens exploit cofilin pathways is crucial for understanding infectious disease and providing potential targets for drug therapies.
Collapse
Affiliation(s)
- Kai Zheng
- a Department of Pharmacy, School of Medicine , Shenzhen University , Shenzhen , Guangdong , People's Republic of China .,c Guangzhou Jinan Biomedicine Research and Development Center, National Engineering Research Center of Genetic Medicine, Jinan University , Guangzhou , China
| | - Kaio Kitazato
- b Division of Molecular Pharmacology of Infectious Agents, Department of Molecular Microbiology and Immunology , Nagasaki University , Nagasaki , Japan , and
| | - Yifei Wang
- c Guangzhou Jinan Biomedicine Research and Development Center, National Engineering Research Center of Genetic Medicine, Jinan University , Guangzhou , China
| | - Zhendan He
- a Department of Pharmacy, School of Medicine , Shenzhen University , Shenzhen , Guangdong , People's Republic of China
| |
Collapse
|
56
|
Bielig H, Lautz K, Braun PR, Menning M, Machuy N, Brügmann C, Barisic S, Eisler SA, Andree M, Zurek B, Kashkar H, Sansonetti PJ, Hausser A, Meyer TF, Kufer TA. The cofilin phosphatase slingshot homolog 1 (SSH1) links NOD1 signaling to actin remodeling. PLoS Pathog 2014; 10:e1004351. [PMID: 25187968 PMCID: PMC4154870 DOI: 10.1371/journal.ppat.1004351] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 07/15/2014] [Indexed: 01/01/2023] Open
Abstract
NOD1 is an intracellular pathogen recognition receptor that contributes to anti-bacterial innate immune responses, adaptive immunity and tissue homeostasis. NOD1-induced signaling relies on actin remodeling, however, the details of the connection of NOD1 and the actin cytoskeleton remained elusive. Here, we identified in a druggable-genome wide siRNA screen the cofilin phosphatase SSH1 as a specific and essential component of the NOD1 pathway. We show that depletion of SSH1 impaired pathogen induced NOD1 signaling evident from diminished NF-κB activation and cytokine release. Chemical inhibition of actin polymerization using cytochalasin D rescued the loss of SSH1. We further demonstrate that NOD1 directly interacted with SSH1 at F-actin rich sites. Finally, we show that enhanced cofilin activity is intimately linked to NOD1 signaling. Our data thus provide evidence that NOD1 requires the SSH1/cofilin network for signaling and to detect bacterial induced changes in actin dynamics leading to NF-κB activation and innate immune responses.
Collapse
Affiliation(s)
- Harald Bielig
- Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
| | - Katja Lautz
- Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
| | - Peter R. Braun
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
- Steinbeis-Innovationszentrum Center for Systems Biomedicine, Falkensee, Germany
| | - Maureen Menning
- Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
| | - Nikolaus Machuy
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Christine Brügmann
- Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
| | - Sandra Barisic
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Stephan A. Eisler
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Maria Andree
- Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
| | - Birte Zurek
- Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
| | - Hamid Kashkar
- Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
| | - Philippe J. Sansonetti
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Paris, France
- INSERM U786, Institut Pasteur, Paris, France
- Microbiologie et Maladies Infectieuses, Collège de France, Paris, France
| | - Angelika Hausser
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Thomas F. Meyer
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Thomas A. Kufer
- Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
- University of Hohenheim, Institute of Nutritional Medicine, Stuttgart, Germany
- * E-mail:
| |
Collapse
|
57
|
Rho protein GTPases and their interactions with NFκB: crossroads of inflammation and matrix biology. Biosci Rep 2014; 34:BSR20140021. [PMID: 24877606 PMCID: PMC4069681 DOI: 10.1042/bsr20140021] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The RhoGTPases, with RhoA, Cdc42 and Rac being major members, are a group of key ubiquitous proteins present in all eukaryotic organisms that subserve such important functions as cell migration, adhesion and differentiation. The NFκB (nuclear factor κB) is a family of constitutive and inducible transcription factors that through their diverse target genes, play a major role in processes such as cytokine expression, stress regulation, cell division and transformation. Research over the past decade has uncovered new molecular links between the RhoGTPases and the NFκB pathway, with the RhoGTPases playing a positive or negative regulatory role on NFκB activation depending on the context. The RhoA–NFκB interaction has been shown to be important in cytokine-activated NFκB processes, such as those induced by TNFα (tumour necrosis factor α). On the other hand, Rac is important for activating the NFκB response downstream of integrin activation, such as after phagocytosis. Specific residues of Rac1 are important for triggering NFκB activation, and mutations do obliterate this response. Other upstream triggers of the RhoGTPase–NFκB interactions include the suppressive p120 catenin, with implications for skin inflammation. The networks described here are not only important areas for further research, but are also significant for discovery of targets for translational medicine.
Collapse
|
58
|
Jakopin Ž. Nucleotide-binding oligomerization domain (NOD) inhibitors: a rational approach toward inhibition of NOD signaling pathway. J Med Chem 2014; 57:6897-918. [PMID: 24707857 DOI: 10.1021/jm401841p] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dysregulation of nucleotide-binding oligomerization domains 1 and 2 (NOD1 and NOD2) has been implicated in the pathology of various inflammatory disorders, rendering them and their downstream signaling proteins potential therapeutic targets. Selective inhibition of NOD1 and NOD2 signaling could be advantageous in treating many acute and chronic diseases; therefore, harnessing the full potential of NOD inhibitors is a key topic in medicinal chemistry. Although they are among the best studied NOD-like receptors (NLRs), the therapeutic potential of pharmacological modulation of NOD1 and NOD2 is largely unexplored. This review is focused on the scientific progress in the field of NOD inhibitors over the past decade, including the recently reported selective inhibitors of NOD1 and NOD2. In addition, the potential approaches to inhibition of NOD signaling as well as the advantages and disadvantages linked with inhibition of NOD signaling are discussed. Finally, the potential directions for drug discovery are also discussed.
Collapse
Affiliation(s)
- Žiga Jakopin
- Faculty of Pharmacy, University of Ljubljana , Aškerčeva 7, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
59
|
Kawano Y, Kaneko-Kawano T, Shimamoto K. Rho family GTPase-dependent immunity in plants and animals. FRONTIERS IN PLANT SCIENCE 2014; 5:522. [PMID: 25352853 PMCID: PMC4196510 DOI: 10.3389/fpls.2014.00522] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 09/16/2014] [Indexed: 05/21/2023]
Abstract
In plants, sophisticated forms of immune systems have developed to cope with a variety of pathogens. Accumulating evidence indicates that Rac (also known as Rop), a member of the Rho family of small GTPases, is a key regulator of immunity in plants and animals. Like other small GTPases, Rac/Rop GTPases function as a molecular switch downstream of immune receptors by cycling between GDP-bound inactive and GTP-bound active forms in cells. Rac/Rop GTPases trigger various immune responses, thereby resulting in enhanced disease resistance to pathogens. In this review, we highlight recent studies that have contributed to our current understanding of the Rac/Rop family GTPases and the upstream and downstream proteins involved in plant immunity. We also compare the features of effector-triggered immunity between plants and animals, and discuss the in vivo monitoring of Rac/Rop activation.
Collapse
Affiliation(s)
- Yoji Kawano
- Laboratory of Plant Molecular Genetics, Nara Institute of Science and TechnologyIkoma, Japan
- *Correspondence: Yoji Kawano, Laboratory of Plant Molecular Genetics, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0101, Japan e-mail:
| | | | - Ko Shimamoto
- Laboratory of Plant Molecular Genetics, Nara Institute of Science and TechnologyIkoma, Japan
| |
Collapse
|
60
|
Lipinski S, Rosenstiel P. Debug Your Bugs - How NLRs Shape Intestinal Host-Microbe Interactions. Front Immunol 2013; 4:479. [PMID: 24409180 PMCID: PMC3873519 DOI: 10.3389/fimmu.2013.00479] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 12/09/2013] [Indexed: 12/14/2022] Open
Abstract
The host's ability to discriminate friend and foe and to establish a precise homeostasis with its associated microbiota is crucial for its survival and fitness. Among the mediators of intestinal host-microbe interactions, NOD-like receptor (NLR) proteins take center stage. They are present in the epithelial lining and innate immune cells that constantly monitor microbial activities at the intestinal barrier. Dysfunctional NLRs predispose to intestinal inflammation as well as sensitization to extra-intestinal immune-mediated diseases and are linked to the alteration of microbial communities. Here, we review advances in our understanding of their reciprocal relationship in the regulation of intestinal homeostasis and implications for intestinal health.
Collapse
Affiliation(s)
- Simone Lipinski
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| |
Collapse
|
61
|
Philpott DJ, Sorbara MT, Robertson SJ, Croitoru K, Girardin SE. NOD proteins: regulators of inflammation in health and disease. Nat Rev Immunol 2013; 14:9-23. [PMID: 24336102 DOI: 10.1038/nri3565] [Citation(s) in RCA: 482] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Entry of bacteria into host cells is an important virulence mechanism. Through peptidoglycan recognition, the nucleotide-binding oligomerization domain (NOD) proteins NOD1 and NOD2 enable detection of intracellular bacteria and promote their clearance through initiation of a pro-inflammatory transcriptional programme and other host defence pathways, including autophagy. Recent findings have expanded the scope of the cellular compartments monitored by NOD1 and NOD2 and have elucidated the signalling pathways that are triggered downstream of NOD activation. In vivo, NOD1 and NOD2 have complex roles, both during bacterial infection and at homeostasis. The association of alleles that encode constitutively active or constitutively inactive forms of NOD2 with different diseases highlights this complexity and indicates that a balanced level of NOD signalling is crucial for the maintenance of immune homeostasis.
Collapse
Affiliation(s)
- Dana J Philpott
- 1] Department of Immunology, University of Toronto, Toronto M5S 1A8, Canada. [2]
| | - Matthew T Sorbara
- 1] Department of Immunology, University of Toronto, Toronto M5S 1A8, Canada. [2]
| | | | - Kenneth Croitoru
- Institute of Medical Science, Department of Medicine, University of Toronto, Toronto M5S 1A8, Canada
| | - Stephen E Girardin
- 1] Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada. [2]
| |
Collapse
|
62
|
Keestra AM, Bäumler AJ. Detection of enteric pathogens by the nodosome. Trends Immunol 2013; 35:123-30. [PMID: 24268520 DOI: 10.1016/j.it.2013.10.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 10/23/2013] [Accepted: 10/24/2013] [Indexed: 01/31/2023]
Abstract
Nucleotide-binding oligomerization domain protein (NOD)1 and NOD2 participate in signaling pathways that detect pathogen-induced processes, such as the presence of peptidoglycan fragments in the host cell cytosol, as danger signals. Recent work suggests that peptidoglycan fragments activate NOD1 indirectly, through activation of the small Rho GTPase Ras-related C3 botulinum toxin substrate 1 (RAC1). Excessive activation of small Rho GTPases by virulence factors of enteric pathogens also triggers the NOD1 signaling pathway. Many enteric pathogens use virulence factors that alter the activation state of small Rho GTPases, thereby manipulating the host cell cytoskeleton of intestinal epithelial cells to promote bacterial attachment or entry. These data suggest that the NOD1 signaling pathway in intestinal epithelial cells provides an important sentinel function for detecting 'breaking and entering' by enteric pathogens.
Collapse
Affiliation(s)
- A Marijke Keestra
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis CA 95616, USA
| | - Andreas J Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis CA 95616, USA.
| |
Collapse
|
63
|
Chaput C, Sander LE, Suttorp N, Opitz B. NOD-Like Receptors in Lung Diseases. Front Immunol 2013; 4:393. [PMID: 24312100 PMCID: PMC3836004 DOI: 10.3389/fimmu.2013.00393] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 11/07/2013] [Indexed: 12/15/2022] Open
Abstract
The lung is a particularly vulnerable organ at the interface of the body and the exterior environment. It is constantly exposed to microbes and particles by inhalation. The innate immune system needs to react promptly and adequately to potential dangers posed by these microbes and particles, while at the same time avoiding extensive tissue damage. Nucleotide-binding oligomerization domain-like receptors (NLRs) represent a group of key sensors for microbes and damage in the lung. As such they are important players in various infectious as well as acute and chronic sterile inflammatory diseases, such as pneumonia, chronic obstructive pulmonary disease (COPD), acute lung injury/acute respiratory distress syndrome, pneumoconiosis, and asthma. Activation of most known NLRs leads to the production and release of pro-inflammatory cytokines, and/or to the induction of cell death. We will review NLR functions in the lung during infection and sterile inflammation.
Collapse
Affiliation(s)
- Catherine Chaput
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin , Berlin , Germany
| | | | | | | |
Collapse
|
64
|
Biasi F, Leonarduzzi G, Oteiza PI, Poli G. Inflammatory bowel disease: mechanisms, redox considerations, and therapeutic targets. Antioxid Redox Signal 2013; 19:1711-47. [PMID: 23305298 PMCID: PMC3809610 DOI: 10.1089/ars.2012.4530] [Citation(s) in RCA: 215] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Oxidative stress is thought to play a key role in the development of intestinal damage in inflammatory bowel disease (IBD), because of its primary involvement in intestinal cells' aberrant immune and inflammatory responses to dietary antigens and to the commensal bacteria. During the active disease phase, activated leukocytes generate not only a wide spectrum of pro-inflammatory cytokines, but also excess oxidative reactions, which markedly alter the redox equilibrium within the gut mucosa, and maintain inflammation by inducing redox-sensitive signaling pathways and transcription factors. Moreover, several inflammatory molecules generate further oxidation products, leading to a self-sustaining and auto-amplifying vicious circle, which eventually impairs the gut barrier. The current treatment of IBD consists of long-term conventional anti-inflammatory therapy and often leads to drug refractoriness or intolerance, limiting patients' quality of life. Immune modulators or anti-tumor necrosis factor α antibodies have recently been used, but all carry the risk of significant side effects and a poor treatment response. Recent developments in molecular medicine point to the possibility of treating the oxidative stress associated with IBD, by designing a proper supplementation of specific lipids to induce local production of anti-inflammatory derivatives, as well as by developing biological therapies that target selective molecules (i.e., nuclear factor-κB, NADPH oxidase, prohibitins, or inflammasomes) involved in redox signaling. The clinical significance of oxidative stress in IBD is now becoming clear, and may soon lead to important new therapeutic options to lessen intestinal damage in this disease.
Collapse
Affiliation(s)
- Fiorella Biasi
- 1 Department of Clinical and Biological Sciences, University of Turin , San Luigi Gonzaga Hospital, Orbassano, Italy
| | | | | | | |
Collapse
|
65
|
Nabatov AA, Hatzis P, Rouschop KMA, van Diest P, Vooijs M. Hypoxia inducible NOD2 interacts with 3-O-sulfogalactoceramide and regulates vesicular homeostasis. Biochim Biophys Acta Gen Subj 2013; 1830:5277-86. [PMID: 23880069 DOI: 10.1016/j.bbagen.2013.07.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 07/12/2013] [Accepted: 07/15/2013] [Indexed: 01/06/2023]
Abstract
BACKGROUND Oxygen sensing in mammalian cells is a conserved signaling pathway regulated by hypoxia inducible factor type 1 (HIF-1). Inadequate oxygen supply (hypoxia) is common to many pathological disorders where autophagy plays an import role. The aim of this study was the identification and characterization of novel HIF-1 target genes that promote autophagy during hypoxia. METHODS Whole genome Chromatin Immune Precipitation from hypoxic HeLa cells was used to identify novel HIF-1 target genes. Hypoxia induced expression and transcription regulation was studied in wild type and HIF-deficient cells. siRNA silencing of candidate genes was used to establish their role during autophagy. Recombinant protein was used for screening immobilized glycosylated lipids to identify potential ligands. RESULTS We identified the Nucleotide Oligomerization Domain 2 (NOD2/CARD15) as a novel HIF-1 target and 3-O-sulfo-galactoceramide (sulfatide) and Mycobacterium sp. specific sulfolipid-1 as the first NOD2 ligands that both compete for binding to NOD2. Loss of NOD2 function impaired autophagy upstream of the autophagy inhibitor chloroquine by reducing the number of acidic vesicles. Inhibition of sulfatide synthesis elicited defects in autophagy similar to the NOD2 loss of function but did not influence NOD2-mediated NF-kB signaling. CONCLUSIONS Our findings suggest that the interaction of NOD2 with sulfatide may mediate the balance between autophagy and inflammation in hypoxic cells. GENERAL SIGNIFICANCE These findings may lead to a better understanding of complex inflammatory pathologies like Crohn's disease and tuberculosis where both NOD2 and hypoxia are implicated.
Collapse
Affiliation(s)
- Alexey A Nabatov
- Maastricht Radiation Oncology, MAASTRO/GROW Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
| | | | | | | | | |
Collapse
|
66
|
Chen Y, Wang C, Liu Y, Tang L, Zheng M, Xu C, Song J, Meng X. miR-122 targets NOD2 to decrease intestinal epithelial cell injury in Crohn's disease. Biochem Biophys Res Commun 2013; 438:133-9. [PMID: 23872065 DOI: 10.1016/j.bbrc.2013.07.040] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 07/05/2013] [Indexed: 12/11/2022]
Abstract
Crohn's disease (CD) is one of the two major types of inflammatory bowel disease (IBD) thought to be caused by genetic and environmental factors. Recently, miR-122 was found to be deregulated in association with CD progression. However, the underlying molecular mechanisms remain unclear. In the present study, the gene nucleotide-binding oligomerization domain 2 (NOD2/CARD15), which is strongly associated with susceptibility to CD, was identified as a functional target of miR-122. MiR-122 inhibited LPS-induced apoptosis by suppressing NOD2 in HT-29 cells. NOD2 interaction with LPS initiates signal transduction mechanisms resulting in the activation of nuclear factor κB (NF-κB) and the stimulation of downstream pro-inflammatory events. The activation of NF-κB was inhibited in LPS-stimulated HT-29 cells pretreated with miR-122 precursor or NOD2 shRNA. The expression of the pro-inflammatory cytokines TNF-α and IFN-γ was significantly decreased, whereas therelease of the anti-inflammatory cytokines IL-4 and IL-10 was increased in LPS-stimulated HT-29 cells pretreated with miR-122 precursor, NOD2 shRNA or the NF-κB inhibitor QNZ. Taken together, these results indicate that miR-122 and its target gene NOD2 may play an important role in the injury of intestinal epithelial cells induced by LPS.
Collapse
Affiliation(s)
- Yu Chen
- Department of Pediatrics, Jiangwan Hospital of Shanghai, Shanghai 200434, China
| | | | | | | | | | | | | | | |
Collapse
|
67
|
Karlsen TA, Brinchmann JE. Liposome delivery of microRNA-145 to mesenchymal stem cells leads to immunological off-target effects mediated by RIG-I. Mol Ther 2013; 21:1169-81. [PMID: 23568258 PMCID: PMC3677300 DOI: 10.1038/mt.2013.55] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 02/27/2013] [Indexed: 12/17/2022] Open
Abstract
Synthetic microRNAs regulate gene expression when transfected into cells, and may be used in strategies for molecular therapy both in vitro and in vivo. Liposomal transfection reagents are frequently used as delivery vehicles in both settings. Here, we report on the immunological off-target effects observed following liposome transfection of synthetic microRNA-145 into human mesenchymal stem cells and human articular chondrocytes (hAC). The immune response was independent on endosome delivery and toll-like receptors (TLRs) but was mediated by retinoic acid inducible-gene 1 (RIG-I). Upregulation of immune genes required liposomal delivery, as no immune response was observed after electroporation of smiR-145 directly in to the cytosol, suggesting a new role of RIG-I. Immune response was seen both with blunt ended and 2-nucleotide 3' overhang versions of synthetic miR-145, and occurred in the absence of a 5'ppp cap. Mutations in a centrally placed poly (UUUU) sequence reduced, but did not abolish the immune response. Interestingly, exposure to liposomes alone led to upregulation of several immune genes, including RIG-I mRNA. However, this process was not mediated by RIG-I. This insight is important for researchers to avoid unexpected results from gene transfer experiments in vitro and unwanted immune responses following the use of lipid-based transfection reagents in vivo.
Collapse
Affiliation(s)
- Tommy A Karlsen
- Institute of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway
- The Norwegian Center for Stem Cell Research, University of Oslo, Oslo, Norway
| | - Jan E Brinchmann
- Institute of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway
- The Norwegian Center for Stem Cell Research, University of Oslo, Oslo, Norway
| |
Collapse
|
68
|
Damm A, Lautz K, Kufer TA. Roles of NLRP10 in innate and adaptive immunity. Microbes Infect 2013; 15:516-23. [PMID: 23562614 DOI: 10.1016/j.micinf.2013.03.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2013] [Accepted: 03/22/2013] [Indexed: 12/19/2022]
|
69
|
Menning M, Kufer TA. A role for the Ankyrin repeat containing protein Ankrd17 in Nod1- and Nod2-mediated inflammatory responses. FEBS Lett 2013; 587:2137-42. [DOI: 10.1016/j.febslet.2013.05.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 04/17/2013] [Accepted: 05/01/2013] [Indexed: 10/26/2022]
|
70
|
Guo W, Zhang D, Wang L, Zhang Y, Liu W. Disruption of asymmetric dimethylarginine-induced RelA/P65 association with actin in endothelial cells. Acta Biochim Biophys Sin (Shanghai) 2013; 45:229-35. [PMID: 23296075 DOI: 10.1093/abbs/gms120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Asymmetric dimethylarginine (ADMA) activates nuclear factor (NF)-κB in endothelial cells, while actin-stabilizing or -destabilizing drugs prevent ADMA-induced activation of NF-κB. Here we investigated how actin-targeting drugs regulated ADMA-induced NF-κB activation in endothelial cells. Human umbilical vein endothelial cells were treated with ADMA for 24 h in the absence and presence of cytochalasin D or jasplakinolide. Expression levels of proteins and genes were measured by immunoblotting and reverse-transcription polymerase chain reaction, respectively. Chromatin immunoprecipitation was used to detect the binding of NF-κB to the vascular cell adhesion molecule 1 (VCAM-1) promoter. The association of actin with RelA/P65 was detected by immunoprecipitation. It was demonstrated that ADMA induced IκBα degradation, increased nuclear RelA/P65 translocation, and promoted the binding of NF-κB to the VCAM-1 promoter. Consequently, this increased the expression of VCAM-1. In parallel studies, actin-stabilizing and -destabilizing drugs decreased ADMA-induced RelA/P65 nuclear translocation, interfered with NF-κB binding to the VCAM-1 promoter and prevented the expression of VCAM-1. This was independent of total RelA/P65 levels and ADMA-induced IκBα degradation. Most importantly, the association of RelA/P65 with actin was increased after stimulation with ADMA, and impaired after treatment with actin-targeting drugs. In brief, actin-stabilizing or -destabilizing drugs interfere with the ADMA-induced association of RelA/P65 with actin, and consequently disrupt NF-κB activation.
Collapse
Affiliation(s)
- Weikang Guo
- Department of Nephrology, Affiliated Beijing Friendship Hospital, Faculty of Kidney Diseases, Capital Medical University, Beijing 100050, China
| | | | | | | | | |
Collapse
|
71
|
Park S, Ha SD, Coleman M, Meshkibaf S, Kim SO. p62/SQSTM1 enhances NOD2-mediated signaling and cytokine production through stabilizing NOD2 oligomerization. PLoS One 2013; 8:e57138. [PMID: 23437331 PMCID: PMC3577775 DOI: 10.1371/journal.pone.0057138] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 01/18/2013] [Indexed: 02/04/2023] Open
Abstract
NOD2 is a cytosolic pattern-recognition receptor that senses muramyl dipeptide of peptidoglycan that constitutes the bacterial cell wall, and plays an important role in maintaining immunological homeostasis in the intestine. To date, multiple molecules have shown to be involved in regulating NOD2 signaling cascades. p62 (sequestosome-1; SQSTM1) is a multifaceted scaffolding protein involved in trafficking molecules to autophagy, and regulating signal cascades activated by Toll-like receptors, inflammasomes and several cytokine receptors. Here, we show that p62 positively regulates NOD2-induced NF-κB activation and p38 MAPK, and subsequent production of cytokines IL-1β and TNF-α. p62 associated with the nucleotide binding domain of NOD2 through a bi-directional interaction mediated by either TRAF6-binding or ubiquitin-associated domains. NOD2 formed a large complex with p62 in an electron-dense area of the cytoplasm, which increased its signaling cascade likely through preventing its degradation. This study for the first time demonstrates a novel role of p62 in enhancing NOD2 signaling effects.
Collapse
Affiliation(s)
- Sangwook Park
- Department of Microbiology and Immunology and Centre for Human Immunology, Siebens-Drake Research Institute, University of Western Ontario, London, Ontario, Canada
| | | | | | | | | |
Collapse
|
72
|
Zurek B, Schoultz I, Neerincx A, Napolitano LM, Birkner K, Bennek E, Sellge G, Lerm M, Meroni G, Söderholm JD, Kufer TA. TRIM27 negatively regulates NOD2 by ubiquitination and proteasomal degradation. PLoS One 2012; 7:e41255. [PMID: 22829933 PMCID: PMC3400628 DOI: 10.1371/journal.pone.0041255] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 06/19/2012] [Indexed: 01/07/2023] Open
Abstract
NOD2, the nucleotide-binding domain and leucine-rich repeat containing gene family (NLR) member 2 is involved in mediating antimicrobial responses. Dysfunctional NOD2 activity can lead to severe inflammatory disorders, but the regulation of NOD2 is still poorly understood. Recently, proteins of the tripartite motif (TRIM) protein family have emerged as regulators of innate immune responses by acting as E3 ubiquitin ligases. We identified TRIM27 as a new specific binding partner for NOD2. We show that NOD2 physically interacts with TRIM27 via the nucleotide-binding domain, and that NOD2 activation enhances this interaction. Dependent on functional TRIM27, ectopically expressed NOD2 is ubiquitinated with K48-linked ubiquitin chains followed by proteasomal degradation. Accordingly, TRIM27 affects NOD2-mediated pro-inflammatory responses. NOD2 mutations are linked to susceptibility to Crohn's disease. We found that TRIM27 expression is increased in Crohn's disease patients, underscoring a physiological role of TRIM27 in regulating NOD2 signaling. In HeLa cells, TRIM27 is partially localized in the nucleus. We revealed that ectopically expressed NOD2 can shuttle to the nucleus in a Walker A dependent manner, suggesting that NOD2 and TRIM27 might functionally cooperate in the nucleus. We conclude that TRIM27 negatively regulates NOD2-mediated signaling by degradation of NOD2 and suggest that TRIM27 could be a new target for therapeutic intervention in NOD2-associated diseases.
Collapse
Affiliation(s)
- Birte Zurek
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
| | - Ida Schoultz
- Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden, and Department of Surgery, Linköping, Sweden
| | - Andreas Neerincx
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
| | - Luisa M. Napolitano
- Cluster in Biomedicine (CBM), AREA Science Park, Trieste, Italy
- Telethon Institute of Genetics and Medicine, Naples, Italy
| | - Katharina Birkner
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
| | - Eveline Bennek
- Department of Medicine III, University Hospital Aachen, Aachen, Germany
| | - Gernot Sellge
- Department of Medicine III, University Hospital Aachen, Aachen, Germany
| | - Maria Lerm
- Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden, and Department of Surgery, Linköping, Sweden
| | - Germana Meroni
- Cluster in Biomedicine (CBM), AREA Science Park, Trieste, Italy
| | - Johan D. Söderholm
- Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden, and Department of Surgery, Linköping, Sweden
| | - Thomas A. Kufer
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
- * E-mail:
| |
Collapse
|
73
|
Lough D, Abdo J, Guerra-Castro JF, Matsumoto C, Kaufman S, Shetty K, Kwon YK, Zasloff M, Fishbein TM. Abnormal CX3CR1⁺ lamina propria myeloid cells from intestinal transplant recipients with NOD2 mutations. Am J Transplant 2012; 12:992-1003. [PMID: 22233287 DOI: 10.1111/j.1600-6143.2011.03897.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Although progress has been made in intestinal transplantation, chronic inflammation remains a challenge. We have reported that the risk of immunological graft loss is almost 100-fold greater in recipients who carry any of the prevalent NOD2 polymorphisms associated with Crohn's disease, and have shown that the normal levels of a key antimicrobial peptide produced by the Paneth cells of the allograft, fall as the graft becomes repopulated by hematopoietic cells of the NOD2 mutant recipient. These studies are extended in this report. Within several months following engraftment into a NOD2 mutant recipient the allograft loses its capacity to prevent adherence of lumenal microbes. Despite the significantly increased expression of CX3CL1, a stress protein produced by the injured enterocyte, NOD2 mutant CX3CR1(+) myeloid cells within the lamina propria fail to exhibit the characteristic morphological phenotype, and fail to express key genes required expressed by NOD2 wild-type cells, including Wnt 5a. We propose that the CX3CR1(+) myeloid cell within the lamina propria supports normal Paneth cell function through expression of Wnt 5a, and that this function is impaired in the setting of intestinal transplantation into a NOD2 mutant recipient. The therapeutic value of Wnt 5a administration in this setting is proposed.
Collapse
Affiliation(s)
- D Lough
- Department of Surgery, Transplant Institute, Georgetown University Medical Center, Washington, DC, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Bozym RA, Delorme-Axford E, Harris K, Morosky S, Ikizler M, Dermody TS, Sarkar SN, Coyne CB. Focal adhesion kinase is a component of antiviral RIG-I-like receptor signaling. Cell Host Microbe 2012; 11:153-66. [PMID: 22341464 PMCID: PMC3995454 DOI: 10.1016/j.chom.2012.01.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 11/30/2011] [Accepted: 01/02/2012] [Indexed: 01/06/2023]
Abstract
Viruses modulate the actin cytoskeleton at almost every step of their cellular journey from entry to egress. Cellular sensing of these cytoskeletal changes may function in the recognition of viral infection. Here we show that focal adhesion kinase (FAK), a focal adhesion localized tyrosine kinase that transmits signals between the extracellular matrix and the cytoplasm, serves as a RIG-I-like receptor antiviral signaling component by directing mitochondrial antiviral signaling adaptor (MAVS) activation. Cells deficient in FAK are highly susceptible to RNA virus infection and attenuated in antiviral signaling. We show that FAK interacts with MAVS at the mitochondrial membrane in a virus infection-dependent manner and potentiates MAVS-mediated signaling via a kinase-independent mechanism. A cysteine protease encoded by enteroviruses cleaves FAK to suppress its role in innate immune signaling. These findings suggest that FAK serves as a link between cytoskeletal perturbations that occur during virus infection and activation of innate immune signaling.
Collapse
Affiliation(s)
- Rebecca A. Bozym
- Department of Microbiology and Molecular Genetics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Elizabeth Delorme-Axford
- Department of Microbiology and Molecular Genetics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Katharine Harris
- Department of Microbiology and Molecular Genetics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Setanie Morosky
- Department of Microbiology and Molecular Genetics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Mine Ikizler
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
- Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Terence S. Dermody
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
- Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Saumendra N. Sarkar
- Department of Microbiology and Molecular Genetics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA
| | - Carolyn B. Coyne
- Department of Microbiology and Molecular Genetics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| |
Collapse
|
75
|
Eitel J, Meixenberger K, van Laak C, Orlovski C, Hocke A, Schmeck B, Hippenstiel S, N'Guessan PD, Suttorp N, Opitz B. Rac1 regulates the NLRP3 inflammasome which mediates IL-1beta production in Chlamydophila pneumoniae infected human mononuclear cells. PLoS One 2012; 7:e30379. [PMID: 22276187 PMCID: PMC3262829 DOI: 10.1371/journal.pone.0030379] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Accepted: 12/19/2011] [Indexed: 01/19/2023] Open
Abstract
Chlamydophila pneumoniae causes acute respiratory tract infections and has been associated with development of asthma and atherosclerosis. The production of IL-1β, a key mediator of acute and chronic inflammation, is regulated on a transcriptional level and additionally on a posttranslational level by inflammasomes. In the present study we show that C. pneumoniae-infected human mononuclear cells produce IL-1β protein depending on an inflammasome consisting of NLRP3, the adapter protein ASC and caspase-1. We further found that the small GTPase Rac1 is activated in C. pneumoniae-infected cells. Importantly, studies with specific inhibitors as well as siRNA show that Rac1 regulates inflammasome activation in C. pneumoniae-infected cells. In conclusion, C. pneumoniae infection of mononuclear cells stimulates IL-1β production dependent on a NLRP3 inflammasome-mediated processing of proIL-1β which is controlled by Rac1.
Collapse
Affiliation(s)
- Julia Eitel
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Karolin Meixenberger
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia van Laak
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Christine Orlovski
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Hocke
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Bernd Schmeck
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Hippenstiel
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Philippe Dje N'Guessan
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Norbert Suttorp
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Bastian Opitz
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
76
|
Abstract
Initial recognition of bacteria by the innate immune system is thought to occur primarily by germline-encoded pattern recognition receptors (PRRs). These receptors are present in multiple compartments of host cells and are thus capable of surveying both the intracellular and extracellular milieu for bacteria. It has generally been presumed that the cellular location of these receptors dictates what type of bacteria they respond to: extracellular bacteria being recognized by cell surface receptors, such as certain Toll-like receptors, and bacteria that are capable of breaching the plasma membrane and entering the cytoplasm, being sensed by cytoplasmic receptors, including the Nod-like receptors (NLRs). Increasingly, it is becoming apparent that this is a false dichotomy and that extracellular bacteria can be sensed by cytoplasmic PRRs and this is crucial for controlling the levels of these bacteria. In this review, we discuss the role of two NLRs, Nod1 and Nod2, in the recognition of and response to extracellular bacteria.
Collapse
Affiliation(s)
- Thomas B Clarke
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6076, USA
| | | |
Collapse
|
77
|
Abstract
Two-signal models have a rich history in immunology. In the classic two-signal model of T-cell activation, signal one consists of engagement of the T-cell receptor by antigen/major histocompatibility complex, whereas signal two arises from costimulatory ligands on antigen-presenting cells. A requirement for two independent signals helps to ensure that T-cell responses are initiated only in response to bona fide infectious threats. Our studies have led us to conclude that initiation of innate immune responses to pathogens also often requires two signals: signal one is initiated by a microbe-derived ligand, such as lipopolysaccharide (LPS) or flagellin, whereas signal two conveys additional contextual information that often accompanies infectious microbes. Although signal one alone is sufficient to initiate many innate responses, certain responses-particularly ones with the potential for self-damage-require two signals for activation. Many of our studies have employed the intracellular bacterial pathogen Legionella pneumophila, which has been established as a valuable model for understanding innate immune responses. In this review, we discuss how the innate immune system integrates multiple signals to generate an effective response to L. pneumophila and other bacterial pathogens.
Collapse
Affiliation(s)
- Mary F Fontana
- Division of Immunology & Pathogenesis, Department of Molecular & Cell Biology, University of California, Berkeley, CA 94720, USA
| | | |
Collapse
|
78
|
Sorbara MT, Philpott DJ. Peptidoglycan: a critical activator of the mammalian immune system during infection and homeostasis. Immunol Rev 2011; 243:40-60. [PMID: 21884166 DOI: 10.1111/j.1600-065x.2011.01047.x] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Peptidoglycan is a conserved structural component of the bacterial cell wall with molecular motifs unique to bacteria. The mammalian immune system takes advantage of these properties and has evolved to recognize this microbial associated molecular pattern. Mammals have four secreted peptidoglycan recognition proteins, PGLYRP-1-4, as well as two intracellular sensors of peptidoglycan, Nod1 and Nod2. Recognition of peptidoglycan is important in initiating and shaping the immune response under both homeostatic and infection conditions. During infection, peptidoglycan recognition drives both cell-autonomous and whole-organism defense responses. Here, we examine recent advances in the understanding of how peptidoglycan recognition shapes mammalian immune responses in these diverse contexts.
Collapse
Affiliation(s)
- Matthew T Sorbara
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
79
|
Dahiya Y, Pandey RK, Sodhi A. Nod2 downregulates TLR2/1 mediated IL1β gene expression in mouse peritoneal macrophages. PLoS One 2011; 6:e27828. [PMID: 22114704 PMCID: PMC3219683 DOI: 10.1371/journal.pone.0027828] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Accepted: 10/26/2011] [Indexed: 12/22/2022] Open
Abstract
Nod2 is a cytosolic pattern recognition receptor. It has been implicated in many inflammatory conditions. Its signaling has been suggested to modulate TLR responses in a variety of ways, yet little is known about the mechanistic details of the process. We show in this study that Nod2 knockdown mouse peritoneal macrophages secrete more IL1β than normal macrophages when stimulated with peptidoglycan (PGN). Muramyl dipeptide (MDP, a Nod2 ligand) + PGN co-stimulated macrophages have lower expression of IL1β than PGN (TLR2/1 ligand) stimulated macrophages. MDP co-stimulation have similar effects on Pam3CSK4 (synthetic TLR2/1 ligand) mediated IL1β expression suggesting that MDP mediated down regulating effects are receptor dependent and ligand independent. MDP mediated down regulation was specific for TLR2/1 signaling as MDP does not affect LPS (TLR4 ligand) or zymosan A (TLR2/6 ligand) mediated IL1β expression. Mechanistically, MDP exerts its down regulating effects by lowering PGN/Pam3CSK4 mediated nuclear cRel levels. Lower nuclear cRel level were observed to be because of enhanced transporting back rather than reduced nuclear translocation of cRel in MDP + PGN stimulated macrophages. These results demonstrate that Nod2 and TLR2/1 signaling pathways are independent and do not interact at the level of MAPK or NF-κB activation.
Collapse
Affiliation(s)
- Yogesh Dahiya
- School of Biotechnology, Faculty of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Rajeev Kumar Pandey
- School of Biotechnology, Faculty of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ajit Sodhi
- School of Biotechnology, Faculty of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
- * E-mail:
| |
Collapse
|
80
|
Morosky SA, Zhu J, Mukherjee A, Sarkar SN, Coyne CB. Retinoic acid-induced gene-I (RIG-I) associates with nucleotide-binding oligomerization domain-2 (NOD2) to negatively regulate inflammatory signaling. J Biol Chem 2011; 286:28574-83. [PMID: 21690088 PMCID: PMC3151099 DOI: 10.1074/jbc.m111.227942] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Revised: 05/17/2011] [Indexed: 11/06/2022] Open
Abstract
Cytoplasmic caspase recruiting domain (CARD)-containing molecules often function in the induction of potent antimicrobial responses in order to protect mammalian cells from invading pathogens. Retinoic acid-induced gene-I (RIG-I) and nucleotide binding oligomerization domain 2 (NOD2) serve as key factors in the detection of viral and bacterial pathogens, and in the subsequent initiation of innate immune signals to combat infection. RIG-I and NOD2 share striking similarities in their cellular localization, both localize to membrane ruffles in non-polarized epithelial cells and both exhibit a close association with the junctional complex of polarized epithelia. Here we show that RIG-I and NOD2 not only colocalize to cellular ruffles and cell-cell junctions, but that they also form a direct interaction that is mediated by the CARDs of RIG-I and multiple regions of NOD2. Moreover, we show that RIG-I negatively regulates ligand-induced nuclear factor-κB (NF-κB) signaling mediated by NOD2, and that NOD2 negatively regulates type I interferon induction by RIG-I. We also show that the three main Crohn disease-associated mutants of NOD2 (1007fs, R702W, G908R) form an interaction with RIG-I and negatively regulate its signaling to a greater extent than wild-type NOD2. Our results show that in addition to their role in innate immune recognition, RIG-I and NOD2 form a direct interaction at actin-enriched sites within cells and suggest that this interaction may impact RIG-I- and NOD2-dependent innate immune signaling.
Collapse
Affiliation(s)
| | - Jianzhong Zhu
- From the Department of Microbiology and Molecular Genetics
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| | | | - Saumendra N. Sarkar
- From the Department of Microbiology and Molecular Genetics
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| | | |
Collapse
|
81
|
Kometani K, Yamada T, Sasaki Y, Yokosuka T, Saito T, Rajewsky K, Ishiai M, Hikida M, Kurosaki T. CIN85 drives B cell responses by linking BCR signals to the canonical NF-kappaB pathway. ACTA ACUST UNITED AC 2011; 208:1447-57. [PMID: 21708930 PMCID: PMC3135365 DOI: 10.1084/jem.20102665] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
CIN85 transduces B cell receptor signals to IKK-β, and its expression in B cells is essential for T cell–independent type II antibody responses in mice. CIN85, an adaptor protein which binds the C-terminal domain of tyrosine phosphorylated Cbl and Cbl-b, has been thought to be involved in the internalization and subsequent degradation of receptors. However, its physiological function remains unclear. To determine its role in B cells, we used Mb1-cre to generate mice with a B cell–specific deletion of CIN85. These mice had impaired T cell–independent type II antibody responses in vivo and diminished IKK-β activation and cellular responses to B cell receptor (BCR) cross-linking in vitro. Introduction of a constitutively active IKK-β construct corrected the defective antibody responses as well as cellular responses in the mutant mice. Together, our results suggest that CIN85 links the BCR to IKK-β activation, thereby contributing to T cell–independent immune responses.
Collapse
Affiliation(s)
- Kohei Kometani
- Laboratory for Lymphocyte Differentiation, RIKEN Research Center for Allergy and Immunology, Turumi-ku, Kanagawa 230-0045, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Staquet MJ, Carrouel F, Keller JF, Baudouin C, Msika P, Bleicher F, Kufer T, Farges JC. Pattern-recognition Receptors in Pulp Defense. Adv Dent Res 2011; 23:296-301. [DOI: 10.1177/0022034511405390] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Initial sensing of infection is mediated by germline-encoded pattern-recognition receptors (PRRs), the activation of which leads to the expression of inflammatory mediators responsible for the elimination of pathogens and infected cells. PRRs act as immune sensors that provide immediate cell responses to pathogen invasion or tissue injury. Here, we review the expression of PRRs in human dental pulp cells, namely, receptors from the Toll-like (TLR) and Nod-like NLR families, by which cells recognize bacteria. Particular attention is given to odontoblasts, which are the first cells encountered by pathogens and represent, in the tooth, the first line of defense for the host. Understanding cellular and molecular mechanisms associated with the recognition of bacterial pathogens by odontoblasts is critical for the development of therapeutic strategies that aim at preventing excessive pulp inflammation and related deleterious effects.
Collapse
Affiliation(s)
- M.-J. Staquet
- Odontoblastes et Régénération des Tissus Dentaires, Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, INRA, Ecole Normale Supérieure de Lyon, 46 allée d’Italie, F-69364 Lyon Cedex 07, France
| | - F. Carrouel
- Odontoblastes et Régénération des Tissus Dentaires, Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, INRA, Ecole Normale Supérieure de Lyon, 46 allée d’Italie, F-69364 Lyon Cedex 07, France
| | - J.-F. Keller
- Odontoblastes et Régénération des Tissus Dentaires, Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, INRA, Ecole Normale Supérieure de Lyon, 46 allée d’Italie, F-69364 Lyon Cedex 07, France
- Hospices Civils de Lyon, Service de Consultations et de Traitements Dentaires, Lyon, France
| | - C. Baudouin
- Laboratoires Expanscience, Département Innovation, Recherche et Développement, Epernon, France
| | - P. Msika
- Laboratoires Expanscience, Département Innovation, Recherche et Développement, Epernon, France
| | - F. Bleicher
- Odontoblastes et Régénération des Tissus Dentaires, Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, INRA, Ecole Normale Supérieure de Lyon, 46 allée d’Italie, F-69364 Lyon Cedex 07, France
| | - T.A. Kufer
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
| | - J.-C. Farges
- Odontoblastes et Régénération des Tissus Dentaires, Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, INRA, Ecole Normale Supérieure de Lyon, 46 allée d’Italie, F-69364 Lyon Cedex 07, France
- Hospices Civils de Lyon, Service de Consultations et de Traitements Dentaires, Lyon, France
| |
Collapse
|
83
|
Brooks MN, Rajaram MVS, Azad AK, Amer AO, Valdivia-Arenas MA, Park JH, Núñez G, Schlesinger LS. NOD2 controls the nature of the inflammatory response and subsequent fate of Mycobacterium tuberculosis and M. bovis BCG in human macrophages. Cell Microbiol 2011; 13:402-18. [PMID: 21040358 PMCID: PMC3259431 DOI: 10.1111/j.1462-5822.2010.01544.x] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mycobacterium tuberculosis (M.tb), which causes tuberculosis, is a host-adapted intracellular pathogen of macrophages. Intracellular pattern recognition receptors in macrophages such as nucleotide-binding oligomerization domain (NOD) proteins regulate pro-inflammatory cytokine production. NOD2-mediated signalling pathways in response to M.tb have been studied primarily in mouse models and cell lines but not in primary human macrophages. Thus we sought to determine the role of NOD2 in regulating cytokine production and growth of virulent M.tb and attenuated Mycobacterium bovis BCG (BCG) in human macrophages. We examined NOD2 expression during monocyte differentiation and observed a marked increase in NOD2 transcript and protein following 2-3 days in culture. Pre-treatment of human monocyte-derived and alveolar macrophages with the NOD2 ligand muramyl dipeptide enhanced production of TNF-α and IL-1β in response to M.tb and BCG in a RIP2-dependent fashion. The NOD2-mediated cytokine response was significantly reduced following knock-down of NOD2 expression by using small interfering RNA (siRNA) in human macrophages. Finally, NOD2 controlled the growth of both M.tb and BCG in human macrophages, whereas controlling only BCG growth in murine macrophages. Together, our results provide evidence that NOD2 is an important intracellular receptor in regulating the host response to M.tb and BCG infection in human macrophages.
Collapse
Affiliation(s)
- Michelle N. Brooks
- Center for Microbial Interface Biology, The Ohio State University, 460 West 12th Avenue, Biomedical Research Tower, Room 1004, Columbus, OH 43210, USA
- Department of Microbiology, The Ohio State University, 460 West 12th Avenue, Biomedical Research Tower, Room 1004, Columbus, OH 43210, USA
| | - Murugesan V. S. Rajaram
- Center for Microbial Interface Biology, The Ohio State University, 460 West 12th Avenue, Biomedical Research Tower, Room 1004, Columbus, OH 43210, USA
| | - Abul K. Azad
- Center for Microbial Interface Biology, The Ohio State University, 460 West 12th Avenue, Biomedical Research Tower, Room 1004, Columbus, OH 43210, USA
- Department of Internal Medicine, Division of Infectious Diseases, The Ohio State University, 460 West 12th Avenue, Biomedical Research Tower, Room 1004, Columbus, OH 43210, USA
| | - Amal O. Amer
- Center for Microbial Interface Biology, The Ohio State University, 460 West 12th Avenue, Biomedical Research Tower, Room 1004, Columbus, OH 43210, USA
- The Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University, 460 West 12th Avenue, Biomedical Research Tower, Room 1004, Columbus, OH 43210, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, 460 West 12th Avenue, Biomedical Research Tower, Room 1004, Columbus, OH 43210, USA
| | - Martin A. Valdivia-Arenas
- Center for Microbial Interface Biology, The Ohio State University, 460 West 12th Avenue, Biomedical Research Tower, Room 1004, Columbus, OH 43210, USA
- The Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University, 460 West 12th Avenue, Biomedical Research Tower, Room 1004, Columbus, OH 43210, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, 460 West 12th Avenue, Biomedical Research Tower, Room 1004, Columbus, OH 43210, USA
| | - Jong-Hwan Park
- Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, 4219 CCGC 0938, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Gabriel Núñez
- Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, 4219 CCGC 0938, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Larry S. Schlesinger
- Center for Microbial Interface Biology, The Ohio State University, 460 West 12th Avenue, Biomedical Research Tower, Room 1004, Columbus, OH 43210, USA
- Department of Microbiology, The Ohio State University, 460 West 12th Avenue, Biomedical Research Tower, Room 1004, Columbus, OH 43210, USA
- Department of Internal Medicine, Division of Infectious Diseases, The Ohio State University, 460 West 12th Avenue, Biomedical Research Tower, Room 1004, Columbus, OH 43210, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, 460 West 12th Avenue, Biomedical Research Tower, Room 1004, Columbus, OH 43210, USA
| |
Collapse
|
84
|
Zurek B, Proell M, Wagner RN, Schwarzenbacher R, Kufer TA. Mutational analysis of human NOD1 and NOD2 NACHT domains reveals different modes of activation. Innate Immun 2011; 18:100-11. [PMID: 21310790 DOI: 10.1177/1753425910394002] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Nucleotide-binding oligomerization domain-containing protein (NOD)1 and NOD2 are intracellular pattern recognition receptors (PRRs) of the nucleotide-binding domain and leucine-rich repeat containing (NLR) gene family involved in innate immune responses. Their centrally located NACHT domain displays ATPase activity and is necessary for activation and oligomerization leading to inflammatory signaling responses. Mutations affecting key residues of the ATPase domain of NOD2 are linked to severe auto-inflammatory diseases, such as Blau syndrome and early-onset sarcoidosis. By mutational dissection of the ATPase domain function, we show that the NLR-specific extended Walker B box (DGhDE) can functionally replace the canonical Walker B sequence (DDhWD) found in other ATPases. A requirement for an intact Walker A box and the magnesium-co-ordinating aspartate of the classical Walker B box suggest that an initial ATP hydrolysis step is necessary for activation of both NOD1 and NOD2. In contrast, a Blau-syndrome associated mutation located in the extended Walker B box of NOD2 that results in higher autoactivation and ligand-induced signaling does not affect NOD1 function. Moreover, mutation of a conserved histidine in the NACHT domain also has contrasting effects on NOD1 and NOD2 mediated NF-κB activation. We conclude that these two NLRs employ different modes of activation and propose distinct models for activation of NOD1 and NOD2.
Collapse
Affiliation(s)
- Birte Zurek
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
| | | | | | | | | |
Collapse
|
85
|
Meshcheriakova EA, Andronova TM, Ivanov VT. [A protein interaction network and cell signaling pathways activated by muramyl peptides]. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2010; 36:581-95. [PMID: 21063445 DOI: 10.1134/s1068162010050018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Review is devoted to studying the interaction muramyl peptides with protein components of immune system cells. Systems analysis of published results may be useful to select not only the strategy to further explore the function of this class of glycopeptides, but their use in clinical practice.
Collapse
|
86
|
Yang C, McCoy K, Davis JL, Schmidt-Supprian M, Sasaki Y, Faccio R, Novack DV. NIK stabilization in osteoclasts results in osteoporosis and enhanced inflammatory osteolysis. PLoS One 2010; 5:e15383. [PMID: 21151480 PMCID: PMC2975662 DOI: 10.1371/journal.pone.0015383] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Accepted: 08/31/2010] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Maintenance of healthy bone requires the balanced activities of osteoclasts (OCs), which resorb bone, and osteoblasts, which build bone. Disproportionate action of OCs is responsible for the bone loss associated with postmenopausal osteoporosis and rheumatoid arthritis. NF-κB inducing kinase (NIK) controls activation of the alternative NF-κB pathway, a critical pathway for OC differentiation. Under basal conditions, TRAF3-mediated NIK degradation prevents downstream signaling, and disruption of the NIK:TRAF3 interaction stabilizes NIK leading to constitutive activation of the alternative NF-κB pathway. METHODOLOGY/PRINCIPAL FINDINGS Using transgenic mice with OC-lineage expression of NIK lacking its TRAF3 binding domain (NT3), we now find that alternative NF-κB activation enhances not only OC differentiation but also OC function. Activating NT3 with either lysozyme M Cre or cathepsinK Cre causes high turnover osteoporosis with increased activity of OCs and osteoblasts. In vitro, NT3-expressing precursors form OCs more quickly and at lower doses of RANKL. When cultured on bone, they exhibit larger actin rings and increased resorptive activity. OC-specific NT3 transgenic mice also have an exaggerated osteolytic response to the serum transfer model of arthritis. CONCLUSIONS Constitutive activation of NIK drives enhanced osteoclastogenesis and bone resorption, both in basal conditions and in response to inflammatory stimuli.
Collapse
Affiliation(s)
- Chang Yang
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Kathleen McCoy
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Jennifer L. Davis
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | | | | | - Roberta Faccio
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Deborah Veis Novack
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Pathology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
87
|
Regueiro V, Moranta D, Frank CG, Larrarte E, Margareto J, March C, Garmendia J, Bengoechea JA. Klebsiella pneumoniae subverts the activation of inflammatory responses in a NOD1-dependent manner. Cell Microbiol 2010; 13:135-53. [PMID: 20846183 DOI: 10.1111/j.1462-5822.2010.01526.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Klebsiella pneumoniae is an important cause of community-acquired and nosocomial pneumonia. Subversion of inflammation is essential for pathogen survival during infection. Evidence indicates that K. pneumoniae infections are characterized by lacking an early inflammatory response although the molecular bases are currently unknown. Here we unveil a novel strategy employed by a pathogen to counteract the activation of inflammatory responses. K. pneumoniae attenuates pro-inflammatory mediators-induced IL-8 secretion. Klebsiella antagonizes the activation of NF-κB via the deubiquitinase CYLD and blocks the phosphorylation of mitogen-activated protein kinases (MAPKs) via the MAPK phosphatase MKP-1. Our studies demonstrate that K. pneumoniae has evolved the capacity to manipulate host systems dedicated to control the immune balance. To exert this anti-inflammatory effect, Klebsiella engages NOD1. In NOD1 knock-down cells, Klebsiella neither induces the expression of CYLD and MKP-1 nor blocks the activation of NF-κB and MAPKs. Klebsiella inhibits Rac1 activation; and inhibition of Rac1 activity triggers a NOD1-mediated CYLD and MKP-1 expression which in turn attenuates IL-1β-induced IL-8 secretion. A capsule (CPS) mutant does not attenuate the inflammatory response. However, purified CPS neither reduces IL-1β-induced IL-8 secretion nor induces the expression of CYLD and MKP-1 thereby indicating that CPS is necessary but not sufficient to attenuate inflammation.
Collapse
Affiliation(s)
- Verónica Regueiro
- Program Infection and Immunity, Fundació Caubet-CIMERA Illes Balears, Bunyola, Spain
| | | | | | | | | | | | | | | |
Collapse
|
88
|
Lecat A, Piette J, Legrand-Poels S. The protein Nod2: an innate receptor more complex than previously assumed. Biochem Pharmacol 2010; 80:2021-31. [PMID: 20643110 DOI: 10.1016/j.bcp.2010.07.016] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Revised: 07/02/2010] [Accepted: 07/08/2010] [Indexed: 02/08/2023]
Abstract
For almost 10 years, Nod2 has been known as a cytosolic innate receptor able to sense peptidoglycan from Gram-positive and -negative bacteria and to trigger RIP2- and NF-κB-mediated pro-inflammatory and antibacterial response. Mutations in the gene encoding Nod2 in humans have been associated with Crohn's disease (CD). Mechanisms by which Nod2 variants can lead to CD development are still under investigation. The most admitted hypothesis suggests that the impaired function of Nod2 variants in intestinal epithelial and phagocytic cells results in deficiencies in epithelial-barrier function which subsequently lead to increased bacterial invasion and inflammation at intestinal sites. Very recent results have just reinforced this hypothesis by demonstrating that Nod2 wild-type (unlike Nod2 variants) could mediate autophagy, allowing an efficient bacterial clearance and adaptative immune response. Other recent data have attributed new roles to Nod2. Indeed, Nod2 has been shown to activate antiviral innate immune responses involving IRF3-dependent IFN-β production after viral ssRNA recognition through a RIP2-independent mechanism requiring the mitochondrial adaptor protein MAVS. Recently, Nod2 has been also shown to be exquisitely tuned to detect mycobacterial infections and mount a protective immunity against these pathogens.
Collapse
Affiliation(s)
- Aurore Lecat
- Laboratory of Virology and Immunology, GIGA-Research, GIGA B34, University of Liège, 4000 Liège, Belgium
| | | | | |
Collapse
|
89
|
Kawano Y, Akamatsu A, Hayashi K, Housen Y, Okuda J, Yao A, Nakashima A, Takahashi H, Yoshida H, Wong HL, Kawasaki T, Shimamoto K. Activation of a Rac GTPase by the NLR Family Disease Resistance Protein Pit Plays a Critical Role in Rice Innate Immunity. Cell Host Microbe 2010; 7:362-75. [DOI: 10.1016/j.chom.2010.04.010] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Revised: 02/01/2010] [Accepted: 04/22/2010] [Indexed: 12/21/2022]
|
90
|
Losick VP, Haenssler E, Moy MY, Isberg RR. LnaB: a Legionella pneumophila activator of NF-kappaB. Cell Microbiol 2010; 12:1083-97. [PMID: 20148897 PMCID: PMC2947841 DOI: 10.1111/j.1462-5822.2010.01452.x] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Legionella pneumophila possesses a large arsenal of type IV translocated substrates. Over 100 such proteins have been identified, but the functions of most are unknown. Previous studies have demonstrated that L. pneumophila activates NF-kappaB, a master transcriptional regulator of the mammalian innate immune response. Activation of NF-kappaB is dependent on the Legionella Icm/Dot type IV protein translocation system, consistent with the possibility that translocated bacterial proteins contribute to this response. To test this hypothesis, an expression library of 159 known and putative translocated substrates was created to evaluate whether ectopic production of a single L. pneumophila protein could activate NF-kappaB in mammalian cells. Expression of two of these proteins, LnaB (Legionella NF-kappaB activator B) and LegK1, resulted in approximately 150-fold induction of NF-kappaB activity in HEK293T cells, levels similar to the strong induction that occurs with ectopic expression of the known activator Nod1. LnaB is a substrate of the Icm/Dot system, and in the absence of this protein, a partial reduction of NF-kappaB activation in host cells occurs after challenge by post-exponential phase bacteria. These data indicate that LnaB is an Icm/Dot substrate that contributes to NF-kappaB activation during L. pneumophila infection in host cells.
Collapse
Affiliation(s)
- Vicki P Losick
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | | | | | | |
Collapse
|
91
|
Src-mediated regulation of inflammatory responses by actin polymerization. Biochem Pharmacol 2010; 79:431-43. [DOI: 10.1016/j.bcp.2009.09.016] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Revised: 09/11/2009] [Accepted: 09/14/2009] [Indexed: 11/19/2022]
|
92
|
Lipinski S, Till A, Sina C, Arlt A, Grasberger H, Schreiber S, Rosenstiel P. DUOX2-derived reactive oxygen species are effectors of NOD2-mediated antibacterial responses. J Cell Sci 2009; 122:3522-30. [PMID: 19759286 DOI: 10.1242/jcs.050690] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Generation of microbicidal reactive oxygen species (ROS) is a pivotal protective component of the innate immune system in many eukaryotes. NOD (nucleotide oligomerisation domain containing protein)-like receptors (NLRs) have been implicated as phylogenetically ancient sensors of intracellular pathogens or endogenous danger signals. NOD2 recognizes the bacterial cell wall component muramyldipeptide leading to NFkappaB and MAPK activation via induced proximity signalling through the serine-threonine kinase RIP2. In addition to the subsequent induction of cytokines and antimicrobial peptides, NOD2 has been shown also to exert a direct antibacterial effect. Using a fluorescence-based ROS detection assay we demonstrate controlled ROS generation as an integral component of NOD2-induced signalling in epithelial cells. We demonstrate that the NAD(P)H oxidase family member DUOX2 is involved in NOD2-dependent ROS production. Coimmunoprecipitation and fluorescence microscopy were used to show that DUOX2 interacts and colocalizes with NOD2 at the plasma membrane. Moreover, simultaneous overexpression of NOD2 and DUOX2 was found to result in cooperative protection against bacterial cytoinvasion using the Listeria monocytogenes infection model. RNAi-based studies revealed that DUOX2 is required for the direct bactericidal properties of NOD2. Our results demonstrate a new role of ROS as effector molecules of protective cellular signalling in response to a defined danger signal carried out by a mammalian intracellular NLR system.
Collapse
Affiliation(s)
- Simone Lipinski
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, 24105 Kiel, Germany
| | | | | | | | | | | | | |
Collapse
|
93
|
Auerbuch V, Golenbock DT, Isberg RR. Innate immune recognition of Yersinia pseudotuberculosis type III secretion. PLoS Pathog 2009; 5:e1000686. [PMID: 19997504 PMCID: PMC2779593 DOI: 10.1371/journal.ppat.1000686] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Accepted: 11/09/2009] [Indexed: 01/08/2023] Open
Abstract
Specialized protein translocation systems are used by many bacterial pathogens to deliver effector proteins into host cells that interfere with normal cellular functions. How the host immune system recognizes and responds to this intrusive event is not understood. To address these questions, we determined the mammalian cellular response to the virulence-associated type III secretion system (T3SS) of the human pathogen Yersinia pseudotuberculosis. We found that macrophages devoid of Toll-like receptor (TLR) signaling regulate expression of 266 genes following recognition of the Y. pseudotuberculosis T3SS. This analysis revealed two temporally distinct responses that could be separated into activation of NFkappaB- and type I IFN-regulated genes. Extracellular bacteria were capable of triggering these signaling events, as inhibition of bacterial uptake had no effect on the ensuing innate immune response. The cytosolic peptidoglycan sensors Nod1 and Nod2 and the inflammasome component caspase-1 were not involved in NFkappaB activation following recognition of the Y. pseudotuberculosis T3SS. However, caspase-1 was required for secretion of the inflammatory cytokine IL-1beta in response to T3SS-positive Y. pseudotuberculosis. In order to characterize the bacterial requirements for induction of this novel TLR-, Nod1/2-, and caspase-1-independent response, we used Y. pseudotuberculosis strains lacking specific components of the T3SS. Formation of a functional T3SS pore was required, as bacteria expressing a secretion needle, but lacking the pore-forming proteins YopB or YopD, did not trigger these signaling events. However, nonspecific membrane disruption could not recapitulate the NFkappaB signaling triggered by Y. pseudotuberculosis expressing a functional T3SS pore. Although host cell recognition of the T3SS did not require known translocated substrates, the ensuing response could be modulated by effectors such as YopJ and YopT, as YopT amplified the response, while YopJ dampened it. Collectively, these data suggest that combined recognition of the T3SS pore and YopBD-mediated delivery of immune activating ligands into the host cytosol informs the host cell of pathogenic challenge. This leads to a unique, multifactorial response distinct from the canonical immune response to a bacterium lacking a T3SS.
Collapse
Affiliation(s)
- Victoria Auerbuch
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| | - Douglas T. Golenbock
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Ralph R. Isberg
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
94
|
Keller JF, Carrouel F, Staquet MJ, Kufer TA, Baudouin C, Msika P, Bleicher F, Farges JC. Expression of NOD2 is increased in inflamed human dental pulps and lipoteichoic acid-stimulated odontoblast-like cells. Innate Immun 2009; 17:29-34. [DOI: 10.1177/1753425909348527] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Human odontoblasts trigger immune response s to oral bacteria that invade dental tissues during the caries process. To date, their ability to regulate the expression of the nucleotide-binding domain leucine-rich repeat containing receptor NOD2 when challenged by Gram-positive bacteria is unknown. In this study, we investigated NOD2 expression in healthy and inflamed human dental pulps challenged by bacteria, and in cultured odontoblast-like cells stimulated with lipoteichoic acid (LTA), a Toll-like receptor (TLR) 2 agonist which is specific for Gram-positive bacteria. We found that NOD2 gene expression was significantly up-regulated in pulps with acute inflammation compared to healthy ones. In vitro, LTA augmented NOD2 gene expression and protein level in odontoblast-like cells. The increase was more pronounced in odontoblast-like cells compared to dental pulp fibroblasts. Blocking experiments in odontoblast-like cells with anti-TLR2 antibody strongly reduced the NOD2 gene expression increase, whereas stimulation with the synthetic TLR2 ligand Pam2CSK4 confirmed NOD2 gene up-regulation following TLR2 engagement. These data suggest that NOD2 up-regulation is part of the odontoblast immune response to Gram-positive bacteria and might be important in protecting human dental pulp from the deleterious effects of cariogenic pathogens.
Collapse
Affiliation(s)
- Jean-François Keller
- Université de Lyon, Université Lyon 1, Institut de Génomique Fonctionnelle de Lyon, UMR5242, CNRS, INRA, Ecole Normale Supérieure de Lyon, Groupe Odontoblastes et Régénération des Tissus Dentaires, Hospices Civils de Lyon, Service de Consultations et de Traitements Dentaires, Lyon, France
| | - Florence Carrouel
- Université de Lyon, Université Lyon 1, Institut de Génomique Fonctionnelle de Lyon, UMR5242, CNRS, INRA, Ecole Normale Supérieure de Lyon, Groupe Odontoblastes et Régénération des Tissus Dentaires
| | - Marie-Jeanne Staquet
- Université de Lyon, Université Lyon 1, Institut de Génomique Fonctionnelle de Lyon, UMR5242, CNRS, INRA, Ecole Normale Supérieure de Lyon, Groupe Odontoblastes et Régénération des Tissus Dentaires, Institut Fédératif Biosciences Gerland Lyon Sud, Faculté d'Odontologie, Lyon, France,
| | - Thomas A. Kufer
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
| | - Caroline Baudouin
- Laboratoires Expanscience, Département Innovation, Recherche et Développement, Epernon, France
| | - Philippe Msika
- Laboratoires Expanscience, Département Innovation, Recherche et Développement, Epernon, France
| | - Françoise Bleicher
- Université de Lyon, Université Lyon 1, Institut de Génomique Fonctionnelle de Lyon, UMR5242, CNRS, INRA, Ecole Normale Supérieure de Lyon, Groupe Odontoblastes et Régénération des Tissus Dentaires
| | - Jean-Christophe Farges
- Université de Lyon, Université Lyon 1, Institut de Génomique Fonctionnelle de Lyon, UMR5242, CNRS, INRA, Ecole Normale Supérieure de Lyon, Groupe Odontoblastes et Régénération des Tissus Dentaires, Hospices Civils de Lyon, Service de Consultations et de Traitements Dentaires, Lyon, France
| |
Collapse
|
95
|
Schmuckli-Maurer J, Kinnaird J, Pillai S, Hermann P, McKellar S, Weir W, Dobbelaere D, Shiels B. Modulation of NF-kappaB activation in Theileria annulata-infected cloned cell lines is associated with detection of parasite-dependent IKK signalosomes and disruption of the actin cytoskeleton. Cell Microbiol 2009; 12:158-73. [PMID: 19804486 DOI: 10.1111/j.1462-5822.2009.01386.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Apicomplexan parasites within the genus Theileria have the ability to induce continuous proliferation and prevent apoptosis of the infected bovine leukocyte. Protection against apoptosis involves constitutive activation of the bovine transcription factor NF-kappaB in a parasite-dependent manner. Activation of NF-kappaB is thought to involve recruitment of IKK signalosomes at the surface of the macroschizont stage of the parasite, and it has been postulated that additional host proteins with adaptor or scaffolding function may be involved in signalosome formation. In this study two clonal cell lines were identified that show marked differences in the level of activated NF-kappaB. Further characterization of these lines demonstrated that elevated levels of activated NF-kappaB correlated with increased resistance to cell death and detection of parasite-associated IKK signalosomes, supporting results of our previous studies. Evidence was also provided for the existence of host- and parasite-dependent NF-kappaB activation pathways that are influenced by the architecture of the actin cytoskeleton. Despite this influence, it appears that the primary event required for formation of the parasite-dependent IKK signalosome is likely to be an interaction between a signalosome component and a parasite-encoded surface ligand.
Collapse
Affiliation(s)
- Jacqueline Schmuckli-Maurer
- Division of Molecular Pathobiology, Department of Clinical Research and VPH, Vetsuisse Faculty Bern, Bern, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
96
|
Vance RE, Isberg RR, Portnoy DA. Patterns of pathogenesis: discrimination of pathogenic and nonpathogenic microbes by the innate immune system. Cell Host Microbe 2009; 6:10-21. [PMID: 19616762 PMCID: PMC2777727 DOI: 10.1016/j.chom.2009.06.007] [Citation(s) in RCA: 401] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2009] [Revised: 06/19/2009] [Accepted: 06/19/2009] [Indexed: 01/01/2023]
Abstract
The dominant conceptual framework for understanding innate immunity has been that host cells respond to evolutionarily conserved molecular features of pathogens called pathogen-associated molecular patterns (PAMPs). Here, we propose that PAMPs should be understood in the context of how they are naturally presented by pathogens. This can be experimentally challenging, since pathogens, almost by definition, bypass host defense. Nevertheless, in this review, we explore the idea that the immune system responds to PAMPs in the context of additional signals that derive from common "patterns of pathogenesis" employed by pathogens to infect, multiply within, and spread among their hosts.
Collapse
Affiliation(s)
- Russell E Vance
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | | |
Collapse
|
97
|
Mukherjee A, Morosky SA, Shen L, Weber CR, Turner JR, Kim KS, Wang T, Coyne CB. Retinoic acid-induced gene-1 (RIG-I) associates with the actin cytoskeleton via caspase activation and recruitment domain-dependent interactions. J Biol Chem 2009; 284:6486-94. [PMID: 19122199 PMCID: PMC5405323 DOI: 10.1074/jbc.m807547200] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Revised: 12/11/2008] [Indexed: 12/21/2022] Open
Abstract
The actin cytoskeleton serves as a barrier that protects mammalian cells from environmental pathogens such as bacteria, fungi, and viruses. Several components of antimicrobial signaling pathways have been shown to associate directly with the actin cytoskeleton, indicating that the cytoskeleton may also serve as a platform for immune-associated molecules. Here we report that retinoic acid-induced gene-I (RIG-I), an important viral RNA recognition molecule, is associated with the actin cytoskeleton and localizes predominantly to actin-enriched membrane ruffles in non-polarized epithelial cells. Subcellular localization and fractionation experiments revealed that the association between RIG-I and the actin cytoskeleton was mediated by its N-terminal caspase activation and recruitment domains (CARDs), which were necessary and sufficient to induce cytoskeletal association. We also show that RIG-I plays a role in cellular migration, as ectopic expression of RIG-I enhanced cellular migration in a wound healing assay and depletion of endogenous RIG-I significantly reduced wound healing. We further show that in both cultured intestinal epithelial cells (IEC) and human colon and small intestine biopsies, RIG-I is enriched at apico-lateral cell junctions and colocalizes with markers of the tight junction. Depolymerization of the actin cytoskeleton in polarized IEC led to the rapid relocalization of RIG-I and to the induction of type I interferon signaling. These data provide evidence that RIG-I is associated with the actin cytoskeleton in non-polarized epithelial cells and with the junctional complex in polarized IECs and human intestine and colon biopsies and may point to a physiological role for RIG-I in the regulation of cellular migration.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Carolyn B. Coyne
- To whom correspondence should be addressed: S313 Biomedical Science Tower, 3500 Terrace St., Pittsburgh, PA 15261. Tel.: 412-383-5149; Fax: 412-383-6517; E-mail:
| |
Collapse
|
98
|
Critical role of NOD2 in regulating the immune response to Staphylococcus aureus. Infect Immun 2009; 77:1376-82. [PMID: 19139201 DOI: 10.1128/iai.00940-08] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
NOD2 (the nucleotide-binding oligomerization domain containing protein 2) is known to be involved in host recognition of bacteria, although its role in the host response to Staphylococcus aureus infection is unknown. NOD2-deficient (Nod2(-/-)) mice and wild-type (WT) littermate controls were injected intraperitoneally with S. aureus suspension (10(7) bacteria/g of body weight), and their survival was monitored. Cultured bone marrow-derived neutrophils were harvested from Nod2(-/-) and WT mice and tested for cytokine production and phagocytosis. Compared to WT mice, Nod2(-/-) mice were significantly more susceptible to S. aureus infection (median survival of 1.5 days versus >5 days; P = 0.003) and had a significantly higher bacterial tissue burden. Cultured bone marrow-derived neutrophils from Nod2(-/-) and WT mice had similar levels of peritoneal neutrophil recruitment and intracellular killing, but bone marrow-derived neutrophils from Nod2(-/-) mice had significantly reduced ability to internalize fluorescein-labeled S. aureus. Nod2(-/-) mice had significantly higher levels of Th1-derived cytokines in serum (tumor necrosis factor alpha, gamma interferon, and interleukin-2 [IL-2]) compared to WT mice, whereas the levels of Th2-derived cytokines (IL-1beta, IL-4, IL-6, and IL-10) were similar in Nod2(-/-) and WT mice. Thus, mice deficient in NOD2 are more susceptible to S. aureus. Increased susceptibility is due in part to defective neutrophil phagocytosis, elevated serum levels of Th1 cytokines, and a higher bacterial tissue burden.
Collapse
|
99
|
Fukazawa A, Alonso C, Kurachi K, Gupta S, Lesser CF, McCormick BA, Reinecker HC. GEF-H1 mediated control of NOD1 dependent NF-kappaB activation by Shigella effectors. PLoS Pathog 2008; 4:e1000228. [PMID: 19043560 PMCID: PMC2583055 DOI: 10.1371/journal.ppat.1000228] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2008] [Accepted: 11/04/2008] [Indexed: 02/07/2023] Open
Abstract
Shigella flexneri has evolved the ability to modify host cell function with intracellular active effectors to overcome the intestinal barrier. The detection of these microbial effectors and the initiation of innate immune responses are critical for rapid mucosal defense activation. The guanine nucleotide exchange factor H1 (GEF-H1) mediates RhoA activation required for cell invasion by the enteroinvasive pathogen Shigella flexneri. Surprisingly, GEF-H1 is requisite for NF-κB activation in response to Shigella infection. GEF-H1 interacts with NOD1 and is required for RIP2 dependent NF-κB activation by H-Ala-D-γGlu-DAP (γTriDAP). GEF-H1 is essential for NF-κB activation by the Shigella effectors IpgB2 and OspB, which were found to signal in a NOD1 and RhoA Kinase (ROCK) dependent manner. Our results demonstrate that GEF-H1 is a critical component of cellular defenses forming an intracellular sensing system with NOD1 for the detection of microbial effectors during cell invasion by pathogens. Shigella is a bacterium that causes food poisoning and serious intestinal infections with diarrheal illness. Pathogens like Shigella utilize intracellular active effectors to overcome the intestinal barrier and invade the host. We demonstrate that intestinal epithelial cells can sense the disturbance of the tight junctional seal, which normally prevents access of microbes to the circulation. A signaling molecule, which is required for cell invasion by Shigella, also activates messengers that activate immune defenses. This pathway of intestinal pathogen detection is activated by Shigella products, which are injected into host cells by the pathogen and depends on intracellular microbial recognition receptors. The detection of altered cellular function by bacterial effectors may be important for the ability to rapidly respond to barrier disruption in the intestine with the attraction and activation of immune cells to defend against the intruders.
Collapse
Affiliation(s)
- Atsuko Fukazawa
- Department of Medicine, Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Carmen Alonso
- Department of Medicine, Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Kiyotaka Kurachi
- Department of Medicine, Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Sonal Gupta
- Department of Medicine, Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Cammie F. Lesser
- Department of Microbiology and Molecular Genetics and Department of Medicine, Division of Infectious Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Beth Ann McCormick
- Department of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Hans-Christian Reinecker
- Department of Medicine, Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
100
|
Actin cytoskeleton differentially modulates NF-κB-mediated IL-8 expression in myelomonocytic cells. Biochem Pharmacol 2008; 76:1214-28. [DOI: 10.1016/j.bcp.2008.08.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Revised: 08/12/2008] [Accepted: 08/13/2008] [Indexed: 12/11/2022]
|