1
|
Matsuda Y, Yamauchi H, Hara H. Activation of inflammasomes and mechanisms for intracellular recognition of Listeria monocytogenes. Microbiol Immunol 2023; 67:429-437. [PMID: 37461376 DOI: 10.1111/1348-0421.13091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 07/03/2023] [Indexed: 10/06/2023]
Abstract
The high mortality rate associated with Listeria monocytogenes can be attributed to its ability to invade the body systemically and to activate inflammasomes. Both of these processes are facilitated by expressing a major virulence factor known as listeriolysin O, a 56 kDa pore-forming protein encoded by the hly gene. Listeriolysin O plays a crucial role in the pathogenesis of the bacterium by facilitating the escape of the pathogen from the phagosome into the cytosol. This process is essential for the successful establishment of infection. In addition, listeriolysin O is known as an immunomodulator that activates host signal transduction. In addition to listeriolysin O, Listeria expresses a variety of bacterial ligands, such as lipoteichoic acid, nucleotide, and flagellin, that are recognized by host intracellular pattern-recognition receptors including Nod-like receptors, AIM2-like receptors, and RIG-I-like receptors. This review introduces intracellular recognition of Listeria monocytogenes since recent studies have revealed that the activation of inflammasome exacerbates Gram-positive bacteria infection.
Collapse
Affiliation(s)
- Yasuyuki Matsuda
- Department of Infectious Diseases, Division of Microbiology and Immunochemistry, Asahikawa Medical University, Asahikawa, Japan
| | - Hajime Yamauchi
- Department of Infectious Diseases, Division of Microbiology and Immunochemistry, Asahikawa Medical University, Asahikawa, Japan
| | - Hideki Hara
- Department of Infectious Diseases, Division of Microbiology and Immunochemistry, Asahikawa Medical University, Asahikawa, Japan
| |
Collapse
|
2
|
Danielli S, Ma Z, Pantazi E, Kumar A, Demarco B, Fischer FA, Paudel U, Weissenrieder J, Lee RJ, Joyce S, Foskett JK, Bezbradica JS. The ion channel CALHM6 controls bacterial infection-induced cellular cross-talk at the immunological synapse. EMBO J 2023; 42:e111450. [PMID: 36861806 PMCID: PMC10068325 DOI: 10.15252/embj.2022111450] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 03/03/2023] Open
Abstract
Membrane ion channels of the calcium homeostasis modulator (CALHM) family promote cell-cell crosstalk at neuronal synapses via ATP release, where ATP acts as a neurotransmitter. CALHM6, the only CALHM highly expressed in immune cells, has been linked to the induction of natural killer (NK) cell anti-tumour activity. However, its mechanism of action and broader functions in the immune system remain unclear. Here, we generated Calhm6-/- mice and report that CALHM6 is important for the regulation of the early innate control of Listeria monocytogenes infection in vivo. We find that CALHM6 is upregulated in macrophages by pathogen-derived signals and that it relocates from the intracellular compartment to the macrophage-NK cell synapse, facilitating ATP release and controlling the kinetics of NK cell activation. Anti-inflammatory cytokines terminate CALHM6 expression. CALHM6 forms an ion channel when expressed in the plasma membrane of Xenopus oocytes, where channel opening is controlled by a conserved acidic residue, E119. In mammalian cells, CALHM6 is localised to intracellular compartments. Our results contribute to the understanding of neurotransmitter-like signal exchange between immune cells that fine-tunes the timing of innate immune responses.
Collapse
Affiliation(s)
- Sara Danielli
- The Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Zhongming Ma
- Department of Physiology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Eirini Pantazi
- The Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Amrendra Kumar
- Department of Veterans AffairsTennessee Valley Healthcare SystemNashvilleTNUSA
- Department of Pathology, Microbiology, & ImmunologyVanderbilt University Medical CenterNashvilleTNUSA
| | - Benjamin Demarco
- The Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Fabian A Fischer
- The Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Usha Paudel
- Department of Physiology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Jillian Weissenrieder
- Department of Physiology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Robert J Lee
- Department of Physiology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
- Department of Otorhinolaryngology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Sebastian Joyce
- Department of Veterans AffairsTennessee Valley Healthcare SystemNashvilleTNUSA
- Department of Pathology, Microbiology, & ImmunologyVanderbilt University Medical CenterNashvilleTNUSA
| | - J Kevin Foskett
- Department of Physiology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
- Department of Cell and Developmental Biology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | | |
Collapse
|
3
|
Deletion of pbpC Enhances Bacterial Pathogenicity on Tomato by Affecting Biofilm Formation, Exopolysaccharides Production, and Exoenzyme Activities in Clavibacter michiganensis. Int J Mol Sci 2023; 24:ijms24065324. [PMID: 36982399 PMCID: PMC10049144 DOI: 10.3390/ijms24065324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 02/14/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
Penicillin-binding proteins (PBPs) are considered essential for bacterial peptidoglycan biosynthesis and cell wall assembly. Clavibacter michiganensis is a representative Gram-positive bacterial species that causes bacterial canker in tomato. pbpC plays a significant role in maintaining cell morphological characteristics and stress responses in C. michiganensis. The current study demonstrated that the deletion of pbpC commonly enhances bacterial pathogenicity in C. michiganensis and revealed the mechanisms through which this occurs. The expression of interrelated virulence genes, including celA, xysA, xysB, and pelA, were significantly upregulated in △pbpC mutants. Compared with those in wild-type strains, exoenzyme activities, the formation of biofilm, and the production of exopolysaccharides (EPS) were significantly increased in △pbpC mutants. It is noteworthy that EPS were responsible for the enhancement in bacterial pathogenicity, with the degree of necrotic tomato stem cankers intensifying with the injection of a gradient of EPS from C. michiganensis. These findings highlight new insights into the role of pbpC affecting bacterial pathogenicity, with an emphasis on EPS, advancing the current understanding of phytopathogenic infection strategies for Gram-positive bacteria.
Collapse
|
4
|
Waldman MM, Rahkola JT, Sigler AL, Chung JW, Willett BAS, Kedl RM, Friedman RS, Jacobelli J. Ena/VASP Protein-Mediated Actin Polymerization Contributes to Naïve CD8 + T Cell Activation and Expansion by Promoting T Cell-APC Interactions In Vivo. Front Immunol 2022; 13:856977. [PMID: 35757762 PMCID: PMC9222560 DOI: 10.3389/fimmu.2022.856977] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
Naïve T cell activation in secondary lymphoid organs such as lymph nodes (LNs) occurs upon recognition of cognate antigen presented by antigen presenting cells (APCs). T cell activation requires cytoskeleton rearrangement and sustained interactions with APCs. Enabled/vasodilator-stimulated phosphoprotein (Ena/VASP) proteins are a family of cytoskeletal effector proteins responsible for actin polymerization and are frequently found at the leading edge of motile cells. Ena/VASP proteins have been implicated in motility and adhesion in various cell types, but their role in primary T cell interstitial motility and activation has not been explored. Our goal was to determine the contribution of Ena/VASP proteins to T cell–APC interactions, T cell activation, and T cell expansion in vivo. Our results showed that naïve T cells from Ena/VASP-deficient mice have a significant reduction in antigen-specific T cell accumulation following Listeria monocytogenes infection. The kinetics of T cell expansion impairment were further confirmed in Ena/VASP-deficient T cells stimulated via dendritic cell immunization. To investigate the cause of this T cell expansion defect, we analyzed T cell–APC interactions in vivo by two-photon microscopy and observed fewer Ena/VASP-deficient naïve T cells interacting with APCs in LNs during priming. We also determined that Ena/VASP-deficient T cells formed conjugates with significantly less actin polymerization at the T cell–APC synapse, and that these conjugates were less stable than their WT counterparts. Finally, we found that Ena/VASP-deficient T cells have less LFA-1 polarized to the T cell–APC synapse. Thus, we conclude that Ena/VASP proteins contribute to T cell actin remodeling during T cell–APC interactions, which promotes the initiation of stable T cell conjugates during APC scanning. Therefore, Ena/VASP proteins are required for efficient activation and expansion of T cells in vivo.
Collapse
Affiliation(s)
- Monique M Waldman
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Barbara Davis Research Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jeremy T Rahkola
- Rocky Mountain Regional Veterans Affairs (VA) Medical Center, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Ashton L Sigler
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Barbara Davis Research Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jeffrey W Chung
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Barbara Davis Research Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Benjamin A S Willett
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Ross M Kedl
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Rachel S Friedman
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Barbara Davis Research Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jordan Jacobelli
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Barbara Davis Research Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, United States
| |
Collapse
|
5
|
Martinez-Espinosa I, Serrato JA, Ortiz-Quintero B. Role of IL-10-Producing Natural Killer Cells in the Regulatory Mechanisms of Inflammation during Systemic Infection. Biomolecules 2021; 12:biom12010004. [PMID: 35053151 PMCID: PMC8773486 DOI: 10.3390/biom12010004] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/08/2021] [Accepted: 12/11/2021] [Indexed: 12/29/2022] Open
Abstract
Natural killer (NK) cells have the dual ability to produce pro-inflammatory (IFNγ) and anti-inflammatory (IL-10) cytokines during systemic infection, which points to their crucial role both as inflammatory effectors for infection clearance and as regulators to counterbalance inflammation to limit immune-mediated damage to the host. In particular, immunosuppressive IL-10 secretion by NK cells has been described to occur in systemic, but not local, infections as a recent immunoregulatory mechanism of inflammation that may be detrimental or beneficial, depending on the timing of release, type of disease, or the infection model. Understanding the factors that drive the production of IL-10 by NK cells and their impact during dualistic inflammatory states, such as sepsis and other non-controlled inflammatory diseases, is relevant for achieving effective therapeutic advancements. In this review, the evidence regarding the immunoregulatory role of IL-10-producing NK cells in systemic infection is summarized and discussed in detail, and the potential molecular mechanisms that drive IL-10 production by NK cells are considered.
Collapse
|
6
|
Bortell N, Aguilera ER, Lenz LL. Pulmonary insults exacerbate susceptibility to oral Listeria monocytogenes infection through the production of IL-10 by NK cells. PLoS Pathog 2021; 17:e1009531. [PMID: 33878120 PMCID: PMC8087096 DOI: 10.1371/journal.ppat.1009531] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 04/30/2021] [Accepted: 04/05/2021] [Indexed: 12/18/2022] Open
Abstract
Most individuals who consume foods contaminated with the bacterial pathogen Listeria monocytogenes (Lm) develop mild symptoms, while others are susceptible to life-threatening systemic infections (listeriosis). Although it is known that the risk of severe disease is increased in certain human populations, including the elderly, it remains unclear why others who consume contaminated food develop listeriosis. Here, we used a murine model to discover that pulmonary coinfections can impair the host's ability to adequately control and eradicate systemic Lm that cross from the intestines to the bloodstream. We found that the resistance of mice to oral Lm infection was dramatically reduced by coinfection with Streptococcus pneumoniae (Spn), a bacterium that colonizes the respiratory tract and can also cause severe infections in the elderly. Exposure to Spn or microbial products, including a recombinant Lm protein (L1S) and lipopolysaccharide (LPS), rendered otherwise resistant hosts susceptible to severe systemic Lm infection. In addition, we show that this increase in susceptibility was dependent on an increase in the production of interleukin-10 (IL-10) from Ncr1+ cells, including natural killer (NK) cells. Lastly, the ability of Ncr1+ cell derived IL-10 to increase disease susceptibility correlated with a dampening of both myeloid cell accumulation and myeloid cell phagocytic capacity in infected tissues. These data suggest that efforts to minimize inflammation in response to an insult at the respiratory mucosa render the host more susceptible to infections by Lm and possibly other pathogens that access the oral mucosa.
Collapse
Affiliation(s)
- Nikki Bortell
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Elizabeth R. Aguilera
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Laurel L. Lenz
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| |
Collapse
|
7
|
Gerbec ZJ, Hashemi E, Nanbakhsh A, Holzhauer S, Yang C, Mei A, Tsaih SW, Lemke A, Flister MJ, Riese MJ, Thakar MS, Malarkannan S. Conditional Deletion of PGC-1α Results in Energetic and Functional Defects in NK Cells. iScience 2020; 23:101454. [PMID: 32858341 PMCID: PMC7474003 DOI: 10.1016/j.isci.2020.101454] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 12/30/2019] [Accepted: 08/10/2020] [Indexed: 01/07/2023] Open
Abstract
During an immune response, natural killer (NK) cells activate specific metabolic pathways to meet the increased energetic and biosynthetic demands associated with effector functions. Here, we found in vivo activation of NK cells during Listeria monocytogenes infection-augmented transcription of genes encoding mitochondria-associated proteins in a manner dependent on the transcriptional coactivator PGC-1α. Using an Ncr1Cre-based conditional knockout mouse, we found that PGC-1α was crucial for optimal NK cell effector functions and bioenergetics, as the deletion of PGC-1α was associated with decreased cytotoxic potential and cytokine production along with altered ADP/ATP ratios. Lack of PGC-1α also significantly impaired the ability of NK cells to control B16F10 tumor growth in vivo, and subsequent gene expression analysis showed that PGC-1α mediates transcription required to maintain mitochondrial activity within the tumor microenvironment. Together, these data suggest that PGC-1α-dependent transcription of specific target genes is required for optimal NK cell function during the response to infection or tumor growth.
Collapse
Affiliation(s)
- Zachary J. Gerbec
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Elaheh Hashemi
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Arash Nanbakhsh
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
| | - Sandra Holzhauer
- Laboratory of Lymphocyte Signaling, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
| | - Chao Yang
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ao Mei
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Shirng-Wern Tsaih
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Angela Lemke
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Michael J. Flister
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Matthew J. Riese
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Laboratory of Lymphocyte Signaling, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Monica S. Thakar
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Subramaniam Malarkannan
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
8
|
Opportunities for broadening the application of cell wall lytic enzymes. Appl Microbiol Biotechnol 2020; 104:9019-9040. [DOI: 10.1007/s00253-020-10862-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/14/2020] [Accepted: 08/26/2020] [Indexed: 01/21/2023]
|
9
|
Eberlein J, Davenport B, Nguyen TT, Victorino F, Jhun K, van der Heide V, Kuleshov M, Ma'ayan A, Kedl R, Homann D. Chemokine Signatures of Pathogen-Specific T Cells I: Effector T Cells. THE JOURNAL OF IMMUNOLOGY 2020; 205:2169-2187. [PMID: 32948687 DOI: 10.4049/jimmunol.2000253] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 08/07/2020] [Indexed: 12/16/2022]
Abstract
The choreography of complex immune responses, including the priming, differentiation, and modulation of specific effector T cell populations generated in the immediate wake of an acute pathogen challenge, is in part controlled by chemokines, a large family of mostly secreted molecules involved in chemotaxis and other patho/physiological processes. T cells are both responsive to various chemokine cues and a relevant source for certain chemokines themselves; yet, the actual range, regulation, and role of effector T cell-derived chemokines remains incompletely understood. In this study, using different in vivo mouse models of viral and bacterial infection as well as protective vaccination, we have defined the entire spectrum of chemokines produced by pathogen-specific CD8+ and CD4+T effector cells and delineated several unique properties pertaining to the temporospatial organization of chemokine expression patterns, synthesis and secretion kinetics, and cooperative regulation. Collectively, our results position the "T cell chemokine response" as a notably prominent, largely invariant, yet distinctive force at the forefront of pathogen-specific effector T cell activities and establish novel practical and conceptual approaches that may serve as a foundation for future investigations into the role of T cell-produced chemokines in infectious and other diseases.
Collapse
Affiliation(s)
- Jens Eberlein
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Bennett Davenport
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Tom T Nguyen
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Francisco Victorino
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Kevin Jhun
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Verena van der Heide
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Maxim Kuleshov
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029; and.,Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Avi Ma'ayan
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029; and.,Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ross Kedl
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Dirk Homann
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045; .,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
10
|
Davenport B, Eberlein J, Nguyen TT, Victorino F, van der Heide V, Kuleshov M, Ma'ayan A, Kedl R, Homann D. Chemokine Signatures of Pathogen-Specific T Cells II: Memory T Cells in Acute and Chronic Infection. THE JOURNAL OF IMMUNOLOGY 2020; 205:2188-2206. [PMID: 32948682 DOI: 10.4049/jimmunol.2000254] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 08/07/2020] [Indexed: 12/20/2022]
Abstract
Pathogen-specific memory T cells (TM) contribute to enhanced immune protection under conditions of reinfection, and their effective recruitment into a recall response relies, in part, on cues imparted by chemokines that coordinate their spatiotemporal positioning. An integrated perspective, however, needs to consider TM as a potentially relevant chemokine source themselves. In this study, we employed a comprehensive transcriptional/translational profiling strategy to delineate the identities, expression patterns, and dynamic regulation of chemokines produced by murine pathogen-specific TM CD8+TM, and to a lesser extent CD4+TM, are a prodigious source for six select chemokines (CCL1/3/4/5, CCL9/10, and XCL1) that collectively constitute a prominent and largely invariant signature across acute and chronic infections. Notably, constitutive CCL5 expression by CD8+TM serves as a unique functional imprint of prior antigenic experience; induced CCL1 production identifies highly polyfunctional CD8+ and CD4+TM subsets; long-term CD8+TM maintenance is associated with a pronounced increase of XCL1 production capacity; chemokines dominate the earliest stages of the CD8+TM recall response because of expeditious synthesis/secretion kinetics (CCL3/4/5) and low activation thresholds (CCL1/3/4/5/XCL1); and TM chemokine profiles modulated by persisting viral Ags exhibit both discrete functional deficits and a notable surplus. Nevertheless, recall responses and partial virus control in chronic infection appear little affected by the absence of major TM chemokines. Although specific contributions of TM-derived chemokines to enhanced immune protection therefore remain to be elucidated in other experimental scenarios, the ready visualization of TM chemokine-expression patterns permits a detailed stratification of TM functionalities that may be correlated with differentiation status, protective capacities, and potential fates.
Collapse
Affiliation(s)
- Bennett Davenport
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jens Eberlein
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Tom T Nguyen
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Francisco Victorino
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Verena van der Heide
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Maxim Kuleshov
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029; and.,Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Avi Ma'ayan
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029; and.,Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ross Kedl
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Dirk Homann
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045; .,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
11
|
Böning MAL, Trittel S, Riese P, van Ham M, Heyner M, Voss M, Parzmair GP, Klawonn F, Jeron A, Guzman CA, Jänsch L, Schraven B, Reinhold A, Bruder D. ADAP Promotes Degranulation and Migration of NK Cells Primed During in vivo Listeria monocytogenes Infection in Mice. Front Immunol 2020; 10:3144. [PMID: 32038647 PMCID: PMC6987423 DOI: 10.3389/fimmu.2019.03144] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 12/27/2019] [Indexed: 12/18/2022] Open
Abstract
The adhesion and degranulation-promoting adaptor protein (ADAP) serves as a multifunctional scaffold and is involved in the formation of immune signaling complexes. To date only limited and moreover conflicting data exist regarding the role of ADAP in NK cells. To extend existing knowledge we investigated ADAP-dependency of NK cells in the context of in vivo infection with the intracellular pathogen Listeria monocytogenes (Lm). Ex vivo analysis of infection-primed NK cells revealed impaired cytotoxic capacity in NK cells lacking ADAP as indicated by reduced CD107a surface expression and inefficient perforin production. However, ADAP-deficiency had no global effect on NK cell morphology or intracellular distribution of CD107a-containing vesicles. Proteomic definition of ADAPko and wild type NK cells did not uncover obvious differences in protein composition during the steady state and moreover, similar early response patterns were induced in NK cells upon infection independent of the genotype. In line with protein network analyses that suggested an altered migration phenotype in naïve ADAPko NK cells, in vitro migration assays uncovered significantly reduced migration of both naïve as well as infection-primed ADAPko NK cells compared to wild type NK cells. Notably, this migration defect was associated with a significantly reduced expression of the integrin CD11a on the surface of splenic ADAP-deficient NK cells 1 day post-Lm infection. We propose that ADAP-dependent alterations in integrin expression might account at least in part for the fact that during in vivo infection significantly lower numbers of ADAPko NK cells accumulate in the spleen i.e., the site of infection. In conclusion, we show here that during systemic Lm infection in mice ADAP is essential for efficient cytotoxic capacity and migration of NK cells.
Collapse
Affiliation(s)
- Martha A L Böning
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Stephanie Trittel
- Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Peggy Riese
- Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Marco van Ham
- Cellular Proteome Research, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Maxi Heyner
- Cellular Proteome Research, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Martin Voss
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Gerald P Parzmair
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Frank Klawonn
- Cellular Proteome Research, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Andreas Jeron
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Carlos A Guzman
- Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Lothar Jänsch
- Cellular Proteome Research, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Annegret Reinhold
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Dunja Bruder
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
12
|
Clark SE, Schmidt RL, McDermott DS, Lenz LL. A Batf3/Nlrp3/IL-18 Axis Promotes Natural Killer Cell IL-10 Production during Listeria monocytogenes Infection. Cell Rep 2019; 23:2582-2594. [PMID: 29847790 PMCID: PMC6170157 DOI: 10.1016/j.celrep.2018.04.106] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/06/2018] [Accepted: 04/25/2018] [Indexed: 11/27/2022] Open
Abstract
The bacterial pathogen Listeria monocytogenes (Lm) capitalizes on natural killer (NK) cell production of regulatory interleukin (IL)-10 to establish severe systemic infections. Here, we identify regulators of this IL-10 secretion. We show that IL-18 signals to NK cells license their ability to produce IL-10. IL-18 acts independent of IL-12 and STAT4, which co-stimulate IFNγ secretion. Dendritic cell (DC) expression of Nlrp3 is required for IL-18 release in response to the Lm p60 virulence protein. Therefore, mice lacking Nlrp3, Il18, or Il18R fail to accumulate serum IL-10 and are highly resistant to systemic Lm infection. We further show that cells expressing or dependent on Batf3 are required for IL-18-inducing IL-10 production observed in infected mice. These findings explain how Il18 and Batf3 promote susceptibility to bacterial infection and demonstrate the ability of Lm to exploit NLRP3 for the promotion of regulatory NK cell activity.
Collapse
Affiliation(s)
- Sarah E Clark
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Rebecca L Schmidt
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206, USA
| | - Daniel S McDermott
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Laurel L Lenz
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA; Department of Biomedical Research, National Jewish Health, Denver, CO 80206, USA.
| |
Collapse
|
13
|
Clark SE, Burrack KS, Jameson SC, Hamilton SE, Lenz LL. NK Cell IL-10 Production Requires IL-15 and IL-10 Driven STAT3 Activation. Front Immunol 2019; 10:2087. [PMID: 31552035 PMCID: PMC6736993 DOI: 10.3389/fimmu.2019.02087] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 08/19/2019] [Indexed: 01/22/2023] Open
Abstract
Natural killer (NK) cells can produce IFNγ or IL-10 to regulate inflammation and immune responses but the factors driving NK cell IL-10 secretion are poorly-defined. Here, we identified NK cell-intrinsic STAT3 activation as vital for IL-10 production during both systemic Listeria monocytogenes (Lm) infection and following IL-15 cytokine/receptor complex (IL15C) treatment for experimental cerebral malaria (ECM). In both contexts, conditional Stat3 deficiency in NK cells abrogated production of IL-10. Initial NK cell STAT3 phosphorylation was driven by IL-15. During Lm infection, this required capture or presentation of IL-15 by NK cell IL-15Rα. Persistent STAT3 activation was required to drive measurable IL-10 secretion and required NK cell expression of IL-10Rα. Survival-promoting effects of IL-15C treatment in ECM were dependent on NK cell Stat3 while NK cell-intrinsic deficiency for Stat3, Il15ra, or Il10ra abrogated NK cell IL-10 production and increased resistance against Lm. NK cell Stat3 deficiency did not impact production of IFNγ, indicating the STAT3 activation initiated by IL-15 and amplified by IL-10 selectively drives the production of anti-inflammatory IL-10 by responding NK cells.
Collapse
Affiliation(s)
- Sarah E Clark
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Kristina S Burrack
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Stephen C Jameson
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Sara E Hamilton
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Laurel L Lenz
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
14
|
D'Orazio SEF. Innate and Adaptive Immune Responses during Listeria monocytogenes Infection. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0065-2019. [PMID: 31124430 PMCID: PMC11086964 DOI: 10.1128/microbiolspec.gpp3-0065-2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Indexed: 12/15/2022] Open
Abstract
It could be argued that we understand the immune response to infection with Listeria monocytogenes better than the immunity elicited by any other bacteria. L. monocytogenes are Gram-positive bacteria that are genetically tractable and easy to cultivate in vitro, and the mouse model of intravenous (i.v.) inoculation is highly reproducible. For these reasons, immunologists frequently use the mouse model of systemic listeriosis to dissect the mechanisms used by mammalian hosts to recognize and respond to infection. This article provides an overview of what we have learned over the past few decades and is divided into three sections: "Innate Immunity" describes how the host initially detects the presence of L. monocytogenes and characterizes the soluble and cellular responses that occur during the first few days postinfection; "Adaptive Immunity" discusses the exquisitely specific T cell response that mediates complete clearance of infection and immunological memory; "Use of Attenuated Listeria as a Vaccine Vector" highlights the ways that investigators have exploited our extensive knowledge of anti-Listeria immunity to develop cancer therapeutics.
Collapse
Affiliation(s)
- Sarah E F D'Orazio
- University of Kentucky, Microbiology, Immunology & Molecular Genetics, Lexington, KY 40536-0298
| |
Collapse
|
15
|
Ortiz AL, Lenz LL. A Listeria-derived polypeptide promotes in vivo activation of NK cells for antitumor therapy. Immunohorizons 2017; 1:53-62. [PMID: 29658011 DOI: 10.4049/immunohorizons.1700013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Immunotherapies have shown promise in treatment of cancer, but more potent and targeted therapies are needed. Natural killer (NK) cells are lymphocytes with innate ability to recognize and lyse tumor cells. When activated, they also produce type II interferon (IFNγ) to orchestrate the activity of other immune cells. Strategies to elicit NK cell activation in vivo have potential usefulness in anti-tumor immunotherapies. Here, we report on a strategy to stimulate NK cell activation and anti-tumor activity in mice with established B16.F10 murine melanomas. We and others previously observed that NK cells are rapidly activated during infection by pathogens such as the bacterium Listeria monocytogenes (Lm). A secreted Lm virulence protein, p60, and a fragment of p60 termed L1S were previously shown to stimulate innate immune responses and promote NK cell activation. We purified recombinant L1S and characterized its activity in cell culture studies. Recombinant L1S protein was also observed to promote accumulation and robust NK cell activation in the lungs when given via intratracheal instillation to control and tumor-bearing mice. Importantly, therapeutic administration of a single L1S dose was found to significantly reduce the number and area of "metastatic" tumor nodules on the lungs of mice with established B16.F10 murine melanomas. Depletion studies showed that these antitumor effects were dependent on NK cells and IFNγ. These data provide proof of concept that administration of a single immune-modulating microbial polypeptide can be used to therapeutically boost NK cell in vivo activation and promote anti-tumor responses.
Collapse
Affiliation(s)
- Amber L Ortiz
- Department of Immunology and Microbiology. University of Colorado School of Medicine. Aurora, CO. 80045
| | - Laurel L Lenz
- Department of Biomedical Research. National Jewish Health. Denver, CO 80206
| |
Collapse
|
16
|
Kapustyan А, Cherno N. Immunological properties of the bacterial origin compounds. FOOD SCIENCE AND TECHNOLOGY 2016. [DOI: 10.15673/fst.v10i3.175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It is shown that microorganisms are an integral element of the mаcroorganism immune system.Peptidoglycan, muramyldypeptyd, teichoic acids are structural components of cell walls of microorganisms. These componentsare an object for recognition of the innate immunity system. The necessity of the bacteria cell walls destruction with a view toobtain the immunotropic products for enteral consumption, able to overcome the intestinal barrier, was substantiated.The use of lactic acid bacteria (ICD) for such purposes is perspective and safe, since the considerable experience oftheir cultivation was accumulated, in addition, ICD have got «GRAS» (Generally Recognized As Safe) status. Waste productsof ICD are organic acids, hydrogen peroxide, bacteriocins and others. These substances have got antagonist activity, implicitlyaffect on the immune system, reducing the antigenic load caused by pathogenic microorganisms.A number of physical, chemical and biochemical methods of bacteria cell walls destruction were considered. Thepriority methods is the soft influence, namely the use of specific enzymes or hydrolases, own autolyzins with a combination ofphysical destruction methods.
Collapse
|
17
|
Clark SE, Filak HC, Guthrie BS, Schmidt RL, Jamieson A, Merkel P, Knight V, Cole CM, Raulet DH, Lenz LL. Bacterial Manipulation of NK Cell Regulatory Activity Increases Susceptibility to Listeria monocytogenes Infection. PLoS Pathog 2016; 12:e1005708. [PMID: 27295349 PMCID: PMC4905663 DOI: 10.1371/journal.ppat.1005708] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 05/25/2016] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells produce interferon (IFN)-γ and thus have been suggested to promote type I immunity during bacterial infections. Yet, Listeria monocytogenes (Lm) and some other pathogens encode proteins that cause increased NK cell activation. Here, we show that stimulation of NK cell activation increases susceptibility during Lm infection despite and independent from robust NK cell production of IFNγ. The increased susceptibility correlated with IL-10 production by responding NK cells. NK cells produced IL-10 as their IFNγ production waned and the Lm virulence protein p60 promoted induction of IL-10 production by mouse and human NK cells. NK cells consequently exerted regulatory effects to suppress accumulation and activation of inflammatory myeloid cells. Our results reveal new dimensions of the role played by NK cells during Lm infection and demonstrate the ability of this bacterial pathogen to exploit the induction of regulatory NK cell activity to increase host susceptibility. Natural killer (NK) cells are an innate immune cell population known to promote antiviral immunity through cytolysis and production of cytokines. Yet, some pathogens encode proteins that cause increased NK cell activation. Here, using a model of systemic infection by the bacterial pathogen Listeria monocytogenes (Lm), we show that NK cell activation increases host susceptibility. Activated NK cells increased bacterial burdens in infected tissues despite their early production of the pro-inflammatory cytokine IFNγ. We found that the ability of NK cells to exacerbate infection was independent from their production of IFNγ and instead due to subsequent production of the anti-inflammatory cytokine IL-10. A single bacterial protein, p60, was sufficient to elicit NK cell production of both early IFNγ and delayed IL-10. IL-10-production by NK cells has been shown to occur in other systems, but our studies are first to show how this “regulatory” response impacts the course of a bacterial infection. We found that IL-10 producing NK cells suppress accumulation and activation of inflammatory myeloid cells. Our studies suggest that the exploitation of NK cell regulatory activity provides selective pressure for the evolution of pathogen proteins that promote NK cell activation.
Collapse
Affiliation(s)
- Sarah E. Clark
- Department of Biomedical Sciences, National Jewish Health, Denver, Colorado, United States of America
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Holly C. Filak
- Department of Biomedical Sciences, National Jewish Health, Denver, Colorado, United States of America
| | - Brandon S. Guthrie
- Department of Biomedical Sciences, National Jewish Health, Denver, Colorado, United States of America
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Rebecca L. Schmidt
- Department of Biomedical Sciences, National Jewish Health, Denver, Colorado, United States of America
| | - Amanda Jamieson
- Department of Molecular and Cell Biology, Division of Immunology, University of California, Berkeley, Berkeley, California, United States of America
| | - Patricia Merkel
- Division of Pathology, Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
| | - Vijaya Knight
- Division of Pathology, Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
| | - Caroline M. Cole
- Department of Pediatrics, National Jewish Health, Denver, Colorado, United States of America
| | - David H. Raulet
- Department of Molecular and Cell Biology, Division of Immunology, University of California, Berkeley, Berkeley, California, United States of America
| | - Laurel L. Lenz
- Department of Biomedical Sciences, National Jewish Health, Denver, Colorado, United States of America
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
18
|
Domain function dissection and catalytic properties of Listeria monocytogenes p60 protein with bacteriolytic activity. Appl Microbiol Biotechnol 2015; 99:10527-37. [PMID: 26363556 DOI: 10.1007/s00253-015-6967-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 08/12/2015] [Accepted: 08/23/2015] [Indexed: 02/07/2023]
Abstract
The major extracellular protein p60 of Listeria monocytogenes (Lm-p60) is an autolysin that can hydrolyze the peptidoglycan of bacterial cell wall and has been shown to be required for L. monocytogenes virulence. The predicted three-dimensional structure of Lm-p60 showed that Lm-p60 could be split into two independent structural domains at the amino acid residue 270. Conserved motif analysis showed that V30, D207, S395, and H444 are the key amino acid residues of the corresponding motifs. However, not only the actual functions of these two domains but also the catalytic properties of Lm-p60 are unclear. We try to express recombinant Lm-p60 and identify the functions of two domains by residue substitution (V30A, D207A, S395A, and H444A) and peptide truncation. The C-terminal domain was identified as catalytic element and N-terminal domain as substrate recognition and binding element. Either N-terminal domain truncation or C-terminal domain truncation presents corresponding biological activity. The catalytic activity of Lm-p60 with a malfunctioned substrate-binding domain was decreased, while the substrate binding was not affected by a mulfunctioned catalytic domain. With turbidimetric method, we determined the optimal conditions for the bacteriolytic activity of Lm-p60 against Micrococcus lysodeikficus. The assay for the effect of Lm-p60 on the bacteriolytic activity of lysozyme revealed that the combined use of Lm-p60 protein with lysozyme showed a strong synergistic effect on the bacteriolytic activity.
Collapse
|
19
|
Dhama K, Karthik K, Tiwari R, Shabbir MZ, Barbuddhe S, Malik SVS, Singh RK. Listeriosis in animals, its public health significance (food-borne zoonosis) and advances in diagnosis and control: a comprehensive review. Vet Q 2015; 35:211-35. [PMID: 26073265 DOI: 10.1080/01652176.2015.1063023] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Listeriosis is an infectious and fatal disease of animals, birds, fish, crustaceans and humans. It is an important food-borne zoonosis caused by Listeria monocytogenes, an intracellular pathogen with unique potential to spread from cell to cell, thereby crossing blood-brain, intestinal and placental barriers. The organism possesses a pile of virulence factors that help to infect the host and evade from host immune machinery. Though disease occurrence is sporadic throughout the world, it can result in severe damage during an outbreak. Listeriosis is characterized by septicaemia, encephalitis, meningitis, meningoencephalitis, abortion, stillbirth, perinatal infections and gastroenteritis with the incubation period varying with the form of infection. L. monocytogenes has been isolated worldwide from humans, animals, poultry, environmental sources like soil, river, decaying plants, and food sources like milk, meat and their products, seafood and vegetables. Since appropriate vaccines are not available and infection is mainly transmitted through foods in humans and animals, hygienic practices can prevent its spread. The present review describes etiology, epidemiology, transmission, clinical signs, post-mortem lesions, pathogenesis, public health significance, and advances in diagnosis, vaccines and treatment of this disease. Special attention has been given to novel as well as prospective emerging therapies that include bacteriophage and cytokine therapy, avian egg yolk antibodies and herbal therapy. Various vaccines, including advances in recombinant and DNA vaccines and their modes of eliciting immune response, are also discussed. Due focus has also been given regarding appropriate prevention and control strategies to be adapted for better management of this zoonotic disease.
Collapse
Affiliation(s)
- Kuldeep Dhama
- a Division of Pathology , Indian Veterinary Research Institute (IVRI) , Izatnagar, Bareilly 243122 , UP , India
| | - Kumaragurubaran Karthik
- b Division of Bacteriology and Mycology , Indian Veterinary Research Institute (IVRI) , Izatnagar, Bareilly 243122 , UP , India
| | - Ruchi Tiwari
- c Department of Veterinary Microbiology and Immunology , College of Veterinary Sciences , Uttar Pradesh Pandit Deen Dayal Upadhayay Pashu Chikitsa Vigyan Vishwavidyalay Evum Go-Anusandhan Sansthan (DUVASU) , Mathura 281001 , India
| | - Muhammad Zubair Shabbir
- d Quality Operations Laboratory , University of Veterinary and Animal Sciences , Lahore 54600, Pakistan
| | - Sukhadeo Barbuddhe
- e Indian Council of Agricultural Research Complex for Goa , Old Goa, Goa 403402, India
| | - Satya Veer Singh Malik
- f Division of Veterinary Public Health , Indian Veterinary Research Institute (IVRI) , Izatnagar, Bareilly 243122 , UP , India
| | - Raj Kumar Singh
- g Indian Veterinary Research Institute (IVRI) , Izatnagar, Bareilly 243122 , UP , India
| |
Collapse
|
20
|
Carvalho F, Sousa S, Cabanes D. How Listeria monocytogenes organizes its surface for virulence. Front Cell Infect Microbiol 2014; 4:48. [PMID: 24809022 PMCID: PMC4010754 DOI: 10.3389/fcimb.2014.00048] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 04/02/2014] [Indexed: 02/04/2023] Open
Abstract
Listeria monocytogenes is a Gram-positive pathogen responsible for the manifestation of human listeriosis, an opportunistic foodborne disease with an associated high mortality rate. The key to the pathogenesis of listeriosis is the capacity of this bacterium to trigger its internalization by non-phagocytic cells and to survive and even replicate within phagocytes. The arsenal of virulence proteins deployed by L. monocytogenes to successfully promote the invasion and infection of host cells has been progressively unveiled over the past decades. A large majority of them is located at the cell envelope, which provides an interface for the establishment of close interactions between these bacterial factors and their host targets. Along the multistep pathways carrying these virulence proteins from the inner side of the cytoplasmic membrane to their cell envelope destination, a multiplicity of auxiliary proteins must act on the immature polypeptides to ensure that they not only maturate into fully functional effectors but also are placed or guided to their correct position in the bacterial surface. As the major scaffold for surface proteins, the cell wall and its metabolism are critical elements in listerial virulence. Conversely, the crucial physical support and protection provided by this structure make it an ideal target for the host immune system. Therefore, mechanisms involving fine modifications of cell envelope components are activated by L. monocytogenes to render it less recognizable by the innate immunity sensors or more resistant to the activity of antimicrobial effectors. This review provides a state-of-the-art compilation of the mechanisms used by L. monocytogenes to organize its surface for virulence, with special focus on those proteins that work “behind the frontline”, either supporting virulence effectors or ensuring the survival of the bacterium within its host.
Collapse
Affiliation(s)
- Filipe Carvalho
- Group of Molecular Microbiology, Unit of Infection and Immunity, Instituto de Biologia Molecular e Celular, University of Porto Porto, Portugal
| | - Sandra Sousa
- Group of Molecular Microbiology, Unit of Infection and Immunity, Instituto de Biologia Molecular e Celular, University of Porto Porto, Portugal
| | - Didier Cabanes
- Group of Molecular Microbiology, Unit of Infection and Immunity, Instituto de Biologia Molecular e Celular, University of Porto Porto, Portugal
| |
Collapse
|
21
|
Tracing innate immune defences along the path of Listeria monocytogenes infection. Immunol Cell Biol 2014; 92:563-9. [PMID: 24732075 DOI: 10.1038/icb.2014.27] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 03/10/2014] [Accepted: 03/13/2014] [Indexed: 12/21/2022]
Abstract
The pathogenic gram-positive bacteria, Listeria monocytogenes is a facultative infectious intracellular pathogen that causes listeriosis. Effective elimination of infection is dependent upon a functioning innate immune system and activation of inflammatory responses by pathogen recognition receptors (PRRs). In this review, we trace the route of L. monocytogenes invasion as it disseminates from the intestinal epithelium, through the bloodstream of the host, to the liver and spleen. Along this route, we highlight the diverse, region specific, innate defences in place throughout the course of infection. We provide an overview of recent advances in our knowledge of key innate immune defences against L. monocytogenes, focusing on the PRRs in various cell types known to be critical in the detection of this pathogen.
Collapse
|
22
|
Kearney S, Delgado C, Lenz LL. Differential effects of type I and II interferons on myeloid cells and resistance to intracellular bacterial infections. Immunol Res 2013; 55:187-200. [PMID: 22983898 DOI: 10.1007/s12026-012-8362-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The type I and II interferons (IFNs) play important roles in regulating immune responses during viral and bacterial infections and in the context of autoimmune and neoplastic diseases. These two IFN types bind to distinct cell surface receptors that are expressed by nearly all cells to trigger signal transduction events and elicit diverse cellular responses. In some cases, type I and II IFNs trigger similar cellular responses, while in other cases, the IFNs have unique or antagonistic effects on host cells. Negative regulators of IFN signaling also modulate cellular responses to the IFNs and play important roles in maintaining immunological homeostasis. In this review, we provide an overview of how IFNs stimulate cellular responses. We discuss the disparate effects of type I and II IFNs on host resistance to certain intracellular bacterial infections and provide an overview of models that have been proposed to account for these disparate effects. Mechanisms of antagonistic cross talk between type I and II IFNs are also introduced.
Collapse
Affiliation(s)
- Staci Kearney
- Integrated Department of Immunology, University of Colorado School of Medicine, Aurora, CO, USA
| | | | | |
Collapse
|
23
|
Williams MA, Schmidt RL, Lenz LL. Early events regulating immunity and pathogenesis during Listeria monocytogenes infection. Trends Immunol 2012; 33:488-95. [PMID: 22677184 PMCID: PMC3440530 DOI: 10.1016/j.it.2012.04.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2012] [Revised: 04/18/2012] [Accepted: 04/22/2012] [Indexed: 11/29/2022]
Abstract
Listeria monocytogenes (Lm) is both a life-threatening pathogen of humans and a model organism that is widely used to dissect the mechanisms of innate and adaptive immune resistance to infection. Specific aspects of the immune response to systemic Lm infection can be protective, neutral, or in some cases deleterious. In this review, we seek to provide an overview of the early events during Lm infection that dictate or regulate host innate and adaptive immune responses. We highlight several recent developments that add to our understanding of the complex interplay between inflammatory responses, host susceptibility to infection, and the development of protective immunity.
Collapse
Affiliation(s)
- Matthew A Williams
- Department of Pathology, University of Utah, 15 North Medical Drive East, Salt Lake City, UT 84112, USA
| | | | | |
Collapse
|
24
|
Luo X, Cai X. A Combined Use of Autolysin p60 and Listeriolysin O Antigens Induces High Protective Immune Responses Against Listeria monocytogenes Infection. Curr Microbiol 2012; 65:813-8. [DOI: 10.1007/s00284-012-0238-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 08/31/2012] [Indexed: 12/11/2022]
|
25
|
Schmidt RL, Lenz LL. Distinct licensing of IL-18 and IL-1β secretion in response to NLRP3 inflammasome activation. PLoS One 2012; 7:e45186. [PMID: 23028835 PMCID: PMC3445464 DOI: 10.1371/journal.pone.0045186] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 08/14/2012] [Indexed: 01/09/2023] Open
Abstract
Inflammasome activation permits processing of interleukins (IL)-1β and 18 and elicits cell death (pyroptosis). Whether these responses are independently licensed or are “hard-wired” consequences of caspase-1 (casp1) activity has not been clear. Here, we show that that each of these responses is independently regulated following activation of NLRP3 inflammasomes by a “non-canonical” stimulus, the secreted Listeria monocytogenes (Lm) p60 protein. Primed murine dendritic cells (DCs) responded to p60 stimulation with reactive oxygen species (ROS) production and secretion of IL-1β and IL-18 but not pyroptosis. Inhibitors of ROS production inhibited secretion of IL-1β, but did not impair IL-18 secretion. Furthermore, DCs from caspase-11 (casp11)-deficient 129S6 mice failed to secrete IL-1β in response to p60 but were fully responsive for IL-18 secretion. These findings reveal that there are distinct licensing requirements for processing of IL-18 versus IL-1β by NLRP3 inflammasomes.
Collapse
Affiliation(s)
- Rebecca L. Schmidt
- Integrated Department of Immunology, National Jewish Health, Denver, Colorado, United States of America
| | - Laurel L. Lenz
- Integrated Department of Immunology, National Jewish Health, Denver, Colorado, United States of America
- Departments of Immunology and Microbiology, University of Colorado - Denver School of Medicine, Denver, Colorado, United States of America
- * E-mail:
| |
Collapse
|
26
|
Regulski K, Courtin P, Meyrand M, Claes IJJ, Lebeer S, Vanderleyden J, Hols P, Guillot A, Chapot-Chartier MP. Analysis of the peptidoglycan hydrolase complement of Lactobacillus casei and characterization of the major γ-D-glutamyl-L-lysyl-endopeptidase. PLoS One 2012; 7:e32301. [PMID: 22384208 PMCID: PMC3288076 DOI: 10.1371/journal.pone.0032301] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Accepted: 01/24/2012] [Indexed: 01/01/2023] Open
Abstract
Peptidoglycan (PG) is the major component of Gram positive bacteria cell wall and is essential for bacterial integrity and shape. Bacteria synthesize PG hydrolases (PGHs) which are able to cleave bonds in their own PG and play major roles in PG remodelling required for bacterial growth and division. Our aim was to identify the main PGHs in Lactobacillus casei BL23, a lactic acid bacterium with probiotic properties. The PGH complement was first identified in silico by amino acid sequence similarity searches of the BL23 genome sequence. Thirteen PGHs were detected with different predicted hydrolytic specificities. Transcription of the genes was confirmed by RT-PCR. A proteomic analysis combining the use of SDS-PAGE and LC-MS/MS revealed the main seven PGHs synthesized during growth of L. casei BL23. Among these PGHs, LCABL_02770 (renamed Lc-p75) was identified as the major one. This protein is the homolog of p75 (Msp1) major secreted protein of Lactobacillus rhamnosus GG, which was shown to promote survival and growth of intestinal epithelial cells. We identified its hydrolytic specificity on PG and showed that it is a γ-D-glutamyl-L-lysyl-endopeptidase. It has a marked specificity towards PG tetrapeptide chains versus tripeptide chains and for oligomers rather than monomers. Immunofluorescence experiments demonstrated that Lc-p75 localizes at cell septa in agreement with its role in daughter cell separation. It is also secreted under an active form as detected in zymogram. Comparison of the muropeptide profiles of wild type and Lc-p75-negative mutant revealed a decrease of the amount of disaccharide-dipeptide in the mutant PG in agreement with Lc-p75 activity. As a conclusion, Lc-p75 is the major L. casei BL23 PGH with endopeptidase specificity and a key role in daughter cell separation. Further studies will aim at investigating the role of Lc-p75 in the anti-inflammatory potential of L. casei BL23.
Collapse
Affiliation(s)
- Krzysztof Regulski
- Institut National de la Recherche Agronomique, UMR1319 Micalis, Jouy-en-Josas, France
- AgroParisTech, UMR Micalis, Jouy-en-Josas, France
| | - Pascal Courtin
- Institut National de la Recherche Agronomique, UMR1319 Micalis, Jouy-en-Josas, France
- AgroParisTech, UMR Micalis, Jouy-en-Josas, France
| | - Mickael Meyrand
- Institut National de la Recherche Agronomique, UMR1319 Micalis, Jouy-en-Josas, France
- AgroParisTech, UMR Micalis, Jouy-en-Josas, France
| | | | - Sarah Lebeer
- Centre of Microbial and Plant Genetics, K.U. Leuven, Leuven, Belgium
| | - Jos Vanderleyden
- Centre of Microbial and Plant Genetics, K.U. Leuven, Leuven, Belgium
| | - Pascal Hols
- Biochimie et Génétique Moléculaire Bactérienne, Institut des Sciences de la Vie, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Alain Guillot
- Institut National de la Recherche Agronomique, UMR1319 Micalis, Jouy-en-Josas, France
- AgroParisTech, UMR Micalis, Jouy-en-Josas, France
| | - Marie-Pierre Chapot-Chartier
- Institut National de la Recherche Agronomique, UMR1319 Micalis, Jouy-en-Josas, France
- AgroParisTech, UMR Micalis, Jouy-en-Josas, France
- * E-mail:
| |
Collapse
|
27
|
Schmidt RL, Filak HC, Lemon JD, Potter TA, Lenz LL. A LysM and SH3-domain containing region of the Listeria monocytogenes p60 protein stimulates accessory cells to promote activation of host NK cells. PLoS Pathog 2011; 7:e1002368. [PMID: 22072975 PMCID: PMC3207947 DOI: 10.1371/journal.ppat.1002368] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Accepted: 09/22/2011] [Indexed: 11/21/2022] Open
Abstract
Listeria monocytogenes (Lm) infection induces rapid and robust activation of host natural killer (NK) cells. Here we define a region of the abundantly secreted Lm endopeptidase, p60, that potently but indirectly stimulates NK cell activation in vitro and in vivo. Lm expression of p60 resulted in increased IFNγ production by naïve NK cells co-cultured with treated dendritic cells (DCs). Moreover, recombinant p60 protein stimulated activation of naive NK cells when co-cultured with TLR or cytokine primed DCs in the absence of Lm. Intact p60 protein weakly digested bacterial peptidoglycan (PGN), but neither muropeptide recognition by RIP2 nor the catalytic activity of p60 was required for NK cell activation. Rather, the immune stimulating activity mapped to an N-terminal region of p60, termed L1S. Treatment of DCs with a recombinant L1S polypeptide stimulated them to activate naïve NK cells in a cell culture model. Further, L1S treatment activated NK cells in vivo and increased host resistance to infection with Francisella tularensis live vaccine strain (LVS). These studies demonstrate an immune stimulating function for a bacterial LysM domain-containing polypeptide and suggest that recombinant versions of L1S or other p60 derivatives can be used to promote NK cell activation in therapeutic contexts.
Collapse
Affiliation(s)
- Rebecca L. Schmidt
- Integrated Department of Immunology, National Jewish Health and University of Colorado School of Medicine, Denver, Colorado, United States of America
| | - Holly C. Filak
- Integrated Department of Immunology, National Jewish Health and University of Colorado School of Medicine, Denver, Colorado, United States of America
| | - Jack D. Lemon
- Integrated Department of Immunology, National Jewish Health and University of Colorado School of Medicine, Denver, Colorado, United States of America
| | - Terry A. Potter
- Integrated Department of Immunology, National Jewish Health and University of Colorado School of Medicine, Denver, Colorado, United States of America
| | - Laurel L. Lenz
- Integrated Department of Immunology, National Jewish Health and University of Colorado School of Medicine, Denver, Colorado, United States of America
| |
Collapse
|
28
|
Jin L, Hill KK, Filak H, Mogan J, Knowles H, Zhang B, Perraud AL, Cambier JC, Lenz LL. MPYS is required for IFN response factor 3 activation and type I IFN production in the response of cultured phagocytes to bacterial second messengers cyclic-di-AMP and cyclic-di-GMP. THE JOURNAL OF IMMUNOLOGY 2011; 187:2595-601. [PMID: 21813776 DOI: 10.4049/jimmunol.1100088] [Citation(s) in RCA: 232] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cyclic-di-GMP and cyclic-di-AMP are second messengers produced by bacteria and influence bacterial cell survival, differentiation, colonization, biofilm formation, virulence, and bacteria-host interactions. In this study, we show that in both RAW264.7 macrophage cells and primary bone marrow-derived macrophages, the production of IFN-β and IL-6, but not TNF, in response to cyclic-di-AMP and cyclic-di-GMP requires MPYS (also known as STING, MITA, and TMEM173). Furthermore, expression of MPYS was required for IFN response factor 3 but not NF-κB activation in response to these bacterial metabolites. We also confirm that MPYS is required for type I IFN production by cultured macrophages infected with the intracellular pathogens Listeria monocytogenes and Francisella tularensis. However, during systemic infection with either pathogen, MPYS deficiency did not impact bacterial burdens in infected spleens. Serum IFN-β and IL-6 concentrations in the infected control and MPYS(-/-) mice were also similar at 24 h postinfection, suggesting that these pathogens stimulate MPYS-independent cytokine production during in vivo infection. Our findings indicate that bifurcating MPYS-dependent and -independent pathways mediate sensing of cytosolic bacterial infections.
Collapse
Affiliation(s)
- Lei Jin
- Integrated Department of Immunology, University of Colorado Denver School of Medicine, Denver, CO 80206, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Stavru F, Archambaud C, Cossart P. Cell biology and immunology of Listeria monocytogenes infections: novel insights. Immunol Rev 2011; 240:160-84. [DOI: 10.1111/j.1600-065x.2010.00993.x] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
30
|
Humann J, Lenz LL. Bacterial peptidoglycan degrading enzymes and their impact on host muropeptide detection. J Innate Immun 2010; 1:88-97. [PMID: 19319201 DOI: 10.1159/000181181] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Peptidoglycan (PGN) is a major component of the bacterial cell envelope in both Gram-positive and Gram-negative bacteria. These muropeptides can be produced or modified by the activity of bacterial glycolytic and peptidolytic enzymes referred to as PGN hydrolases and autolysins. Some of these bacterial enzymes are crucial for bacterial pathogenicity and have been shown to modulate muropeptide release and/or host innate immune responses. The ability of muropeptides to modulate host responses is due to the fact that eukaryotes do not produce PGN and have instead evolved numerous strategies to detect intact PGN and PGN fragments (muropeptides). Here we review the structure of PGN and introduce the various bacterial enzymes known to degrade or modify bacterial PGN. Host factors involved in PGN and muropeptide detection are also briefly discussed, as are examples of how specific bacterial pathogens use PGN degradation and modification to subvert host innate immunity.
Collapse
|
31
|
Drevets DA, Schawang JE, Mandava VK, Dillon MJ, Leenen PJM. Severe Listeria monocytogenes infection induces development of monocytes with distinct phenotypic and functional features. THE JOURNAL OF IMMUNOLOGY 2010; 185:2432-41. [PMID: 20631315 DOI: 10.4049/jimmunol.1000486] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Monocytes perform diverse roles during infection with the facultative intracellular bacterium Listeria monocytogenes. They are essential as bactericidal cells in host defense but can also become Trojan horses transporting bacteria into the brain. To explain these contrasting roles, we characterized bone marrow (BM) monocytes in steady state and generated during lethal and sublethal L. monocytogenes infection. Ly-6C(high)CD11b(+) BM monocytes expressed high amounts of M-CSFR/CD115 in steady state and 72 h following sublethal infection. However, infection with increasing numbers of bacteria resulted in progressive loss of CD115 and strongly decreased CD115-encoding c-fms mRNA expression. Conversely, analysis of regulatory molecules showed de novo expression of the nonsignaling IL-1RII, CD121b, under the same conditions. Ly-6C(high)CD11b(+) monocytes in circulation also acquired a CD115(neg/low)CD121b(high) phenotype during lethal infection. These BM monocytes showed upregulation of suppressor of cytokine signaling 1 and 3 and IL-1R-"associated kinase-M to a greater extent and/or earlier compared with cells from sublethal infection and showed decreased LPS-induced IL-6 production despite similar levels of surface TLR4 expression. BM monocytes from uninfected or sublethally infected mice bound and internalized very few L. monocytogenes in vitro. However, both functions were significantly increased in monocytes developing during lethal infection. Nonetheless, these cells did not produce reactive oxygen intermediates, suggesting an inability to kill L. monocytogenes. Together, these data show that systemic infections with lethal and sublethal amounts of bacteria differentially shape developing BM monocytes. This results in distinct phenotypic and functional properties consistent with being Trojan horses rather than bactericidal effector cells.
Collapse
Affiliation(s)
- Douglas A Drevets
- Department of Medicine, Oklahoma University Health Sciences Center, Oklahoma City, OK 73014, USA.
| | | | | | | | | |
Collapse
|
32
|
Humann J, Lenz LL. Activation of naive NK cells in response to Listeria monocytogenes requires IL-18 and contact with infected dendritic cells. THE JOURNAL OF IMMUNOLOGY 2010; 184:5172-8. [PMID: 20351186 DOI: 10.4049/jimmunol.0903759] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The mechanisms for NK cell activation during infection by intracellular bacterial pathogens are not clearly defined. To dissect how Listeria monocytogenes infection elicits NK cell activation, we evaluated the requirements for activation of naive splenic NK cells by infected bone marrow-derived dendritic cells (BMDCs). We found that NK cell activation in this setting required infection of BMDCs by live wild type bacteria. NK cells were not activated when BMDCs were infected with a live hemolysin deficient (Deltahly) strain. Neutralization of IL-12, TNF-alpha, or caspase-1 each dramatically reduced NK cell IFN-gamma production in response to live wt L. monocytogenes infection. Addition of recombinant IL-18, but not IL-1beta, reversed the effects of caspase-1 inhibition. Recombinant IL-18 also restored NK cell activation by BMDCs infected with Deltahly L. monocytogenes, which produced IL-12 but not IL-18. IL-18 acted on NK cells because MyD88 expression was required in responding NK cells, but not infected BMDC. However, secreted cytokines were not sufficient for activation of naive NK cells by infected BMDCs. Rather, NK cell activation additionally required contact between infected BMDCs and NK cells. These data suggest that the activation of NK cells during L. monocytogenes infection requires both secreted cytokines and ligation of NK activating receptors during direct contact with infected DCs.
Collapse
Affiliation(s)
- Jessica Humann
- National Jewish Health and Integrated Department of Immunology, University of Colorado, Denver, CO 80206, USA
| | | |
Collapse
|
33
|
Sashinami H, Hu DL, Li SJ, Mitsui T, Hakamada KI, Ishiguro Y, Fukuda S, Nakane A. Virulence factor p60 of Listeria monocytogenes modulates innate immunity by inducing tumor necrosis factor alpha. ACTA ACUST UNITED AC 2010; 59:100-7. [PMID: 20337701 DOI: 10.1111/j.1574-695x.2010.00666.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
We investigated the effect of p60, a virulence factor of Listeria monocytogenes, on host immune response in vitro and in vivo. Administration of p60 before a sublethal infection with L. monocytogenes enhanced innate host resistance in naïve mice. Mouse macrophage RAW264.7 cells produced tumor necrosis factor (TNF)-alpha in response to stimulation with recombinant p60. Toll-like receptor 4 may be involved in TNF-alpha production from RAW264.7 cells and enhanced host resistance induced by p60 administration. Our findings demonstrated that p60 modulates innate immune responses against L. monocytogenes infection.
Collapse
Affiliation(s)
- Hiroshi Sashinami
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Characterization of Acp, a peptidoglycan hydrolase of Clostridium perfringens with N-acetylglucosaminidase activity that is implicated in cell separation and stress-induced autolysis. J Bacteriol 2010; 192:2373-84. [PMID: 20190047 DOI: 10.1128/jb.01546-09] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
This work reports the characterization of the first known peptidoglycan hydrolase (Acp) produced mainly during vegetative growth of Clostridium perfringens. Acp has a modular structure with three domains: a signal peptide domain, an N-terminal domain with repeated sequences, and a C-terminal catalytic domain. The purified recombinant catalytic domain of Acp displayed lytic activity on the cell walls of several Gram-positive bacterial species. Its hydrolytic specificity was established by analyzing the Bacillus subtilis peptidoglycan digestion products by coupling reverse phase-high-pressure liquid chromatography (RP-HPLC) and matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS) analysis, which displayed an N-acetylglucosaminidase activity. The study of acp expression showed a constant expression during growth, which suggested an important role of Acp in growth of C. perfringens. Furthermore, cell fractionation and indirect immunofluorescence staining using anti-Acp antibodies revealed that Acp is located at the septal peptidoglycan of vegetative cells during exponential growth phase, indicating a role in cell separation or division of C. perfringens. A knockout acp mutant strain was obtained by using the insertion of mobile group II intron strategy (ClosTron). The microscopic examination indicated a lack of vegetative cell separation in the acp mutant strain, as well as the wild-type strain incubated with anti-Acp antibodies, demonstrating the critical role of Acp in cell separation. The comparative responses of wild-type and acp mutant strains to stresses induced by Triton X-100, bile salts, and vancomycin revealed an implication of Acp in autolysis induced by these stresses. Overall, Acp appears as a major cell wall N-acetylglucosaminidase implicated in both vegetative growth and stress-induced autolysis.
Collapse
|
35
|
Rayamajhi M, Humann J, Penheiter K, Andreasen K, Lenz LL. Induction of IFN-alphabeta enables Listeria monocytogenes to suppress macrophage activation by IFN-gamma. J Exp Med 2010; 207:327-37. [PMID: 20123961 PMCID: PMC2822610 DOI: 10.1084/jem.20091746] [Citation(s) in RCA: 163] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Accepted: 01/05/2010] [Indexed: 01/25/2023] Open
Abstract
Production of type I interferon (IFN; IFN-alphabeta) increases host susceptibility to Listeria monocytogenes, whereas type II IFN (IFN-gamma) activates macrophages to resist infection. We show that these opposing immunological effects of IFN-alphabeta and IFN-gamma occur because of cross talk between the respective signaling pathways. We found that cultured macrophages infected with L. monocytogenes were refractory to IFN-gamma treatment as a result of down-regulation of the IFN-gamma receptor (IFNGR). The soluble factor responsible for these effects was identified as host IFN-alphabeta. Accordingly, macrophages and dendritic cells (DCs) showed reduced IFNGR1 expression and reduced responsiveness to IFN-gamma during systemic infection of IFN-alphabeta-responsive mice. Furthermore, the increased resistance of mice lacking the IFN-alphabeta receptor (IFNAR(-/-)) to L. monocytogenes correlated with increased expression of IFN-gamma-dependent activation markers by macrophages and DCs and was reversed by depletion of IFN-gamma. Thus, IFN-alphabeta produced in response to bacterial infection and other stimuli antagonizes the host response to IFN-gamma by down-regulating the IFNGR. Such cross talk permits prioritization of IFN-alphabeta-type immune responses and may contribute to the beneficial effects of IFN-beta in treatment of inflammatory diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Manira Rayamajhi
- National Jewish Health, Denver, CO 80206
- Integrated Department of Immunology, University of Colorado, Denver, Denver, CO 80206
| | - Jessica Humann
- National Jewish Health, Denver, CO 80206
- Integrated Department of Immunology, University of Colorado, Denver, Denver, CO 80206
| | | | | | - Laurel L. Lenz
- National Jewish Health, Denver, CO 80206
- Integrated Department of Immunology, University of Colorado, Denver, Denver, CO 80206
| |
Collapse
|
36
|
Eberlein J, Nguyen TT, Victorino F, Golden-Mason L, Rosen HR, Homann D. Comprehensive assessment of chemokine expression profiles by flow cytometry. J Clin Invest 2010; 120:907-23. [PMID: 20197626 DOI: 10.1172/jci40645] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Accepted: 12/14/2009] [Indexed: 11/17/2022] Open
Abstract
The chemokines are a large family of mainly secreted molecules involved in the regulation of numerous physiological and pathophysiological processes. Despite many years of investigation, the precise cellular sources of most chemokines have remained incompletely defined as a consequence of the limited availability of suitable reagents to visualize the expression of chemokine proteins at the single-cell level. Here, we developed a simple flow cytometry-based assay using commercially available chemokine-specific antibodies for efficient cell-associated detection of 37 of 39 murine chemokines. To demonstrate the utility of this methodology, we used it to reevaluate the nature of homeostatic chemokines in the hematopoietic compartment, to delineate the complete chemokine profiles of NK cells and B cells in response to major polyclonal stimuli, and to assess the chemokine response of DCs to bacterial infection. The versatility of this analytical methodology was further demonstrated by its application to selected human chemokines and should greatly facilitate any future investigation into chemokine biology at large.
Collapse
Affiliation(s)
- Jens Eberlein
- Barbara Davis Center for Childhood Diabetes, Department of Medicine, University of Colorado Denver, Aurora, 80045-0511, USA
| | | | | | | | | | | |
Collapse
|
37
|
Hyland KA, Brennan R, Olmsted SB, Rojas E, Murphy E, Wang B, Cleary PP. The early interferon response of nasal-associated lymphoid tissue to Streptococcus pyogenes infection. ACTA ACUST UNITED AC 2009; 55:422-31. [PMID: 19243434 DOI: 10.1111/j.1574-695x.2009.00540.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Streptococcus pyogenes is a major causative agent of tonsillitis or pharyngitis in children. Streptococcus pyogenes can persist in tonsils, and one-third of children treated with antibiotics continue to shed streptococci and have recurrent infections. Mouse nasal-associated lymphoid tissue (NALT) is functionally analogous to human oropharyngeal lymphoid tissues, and serves as a model for characterization of the mucosal innate immune response to S. pyogenes. Wild-type S. pyogenes induces transcription of both type I and interferon-gamma (IFN-gamma)-responsive genes, proinflammatory genes and acute-phase response proteins 24 h after intranasal infection. Invasion of NALT and the induction of the interferon response were not dependent on expression of antiphagocytic M protein. Intranasal infection induces a substantial influx of neutrophils into NALT at 24 h, which declines by 48 h after infection. Infection of IFN-gamma(-/-) [IFN-gamma knock-out mouse (GKO)] C57BL/6 mice with wild-type S. pyogenes resulted in local dissemination of bacteria to draining lymph nodes (LN), but did not lead to systemic infection by 48 h after infection. Infected GKO mice had an increased influx of neutrophils into NALT compared with immunocompetent mice. Thus, IFN-gamma-induced responses are required to prevent local dissemination of streptococci to the draining LN.
Collapse
Affiliation(s)
- Kendra A Hyland
- Department of Microbiology, University of Minnesota, Minneapolis, MN 55413, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Stable integration vector for nutrient broth-based selection of attenuated Listeria monocytogenes strains with recombinant antigen expression. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2008; 15:1414-9. [PMID: 18650400 DOI: 10.1128/cvi.00208-08] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Recombinant Listeria monocytogenes strains induce strong cellular immune responses and may prove useful for antigen delivery for the vaccination of humans. However, the genetic systems currently available for the stable expression of recombinant antigens by L. monocytogenes rely on the use of antibiotic resistance genes. We report on a derivative, pPL2dalGlnA, of the Listeria monocytogenes pPL2 integration vector that completely lacks drug resistance genes. The selectable markers in pPL2dalGlnA are glutamine synthetase (GlnA) and alanine racemase (Dal). This novel vector was stably maintained in auxotropic L. monocytogenes strains that normally require d-alanine. The pPL2dalGlnA vector also partially restored the ability of an L. monocytogenes Deltadal Deltadat strain to colonize the spleens and livers of infected mice. A novel, highly attenuated strain of L. monocytogenes with quadruple deletions was also engineered by deleting the L. monocytogenes actA and plcB virulence genes from a Deltadal Deltadat strain. Infection of mice with recombinants of this mutant strain that express the antigen from pPL2dalGlnA were shown to elicit CD8(+) T-cell responses to human immunodeficiency virus Tat. This vector system is thus useful for stable antigen expression and vaccination studies.
Collapse
|
39
|
Noor S, Goldfine H, Tucker DE, Suram S, Lenz LL, Akira S, Uematsu S, Girotti M, Bonventre JV, Breuel K, Williams DL, Leslie CC. Activation of cytosolic phospholipase A2alpha in resident peritoneal macrophages by Listeria monocytogenes involves listeriolysin O and TLR2. J Biol Chem 2008; 283:4744-55. [PMID: 18083708 PMCID: PMC4741307 DOI: 10.1074/jbc.m709956200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Eicosanoid production by macrophages is an early response to microbial infection that promotes acute inflammation. The intracellular pathogen Listeria monocytogenes stimulates arachidonic acid release and eicosanoid production from resident mouse peritoneal macrophages through activation of group IVA cytosolic phospholipase A2 (cPLA2alpha). The ability of wild type L. monocytogenes (WTLM) to stimulate arachidonic acid release is partially dependent on the virulence factor listeriolysin O; however, WTLM and L. monocytogenes lacking listeriolysin O (DeltahlyLM) induce similar levels of cyclooxygenase 2. Arachidonic acid release requires activation of MAPKs by WTLM and DeltahlyLM. The attenuated release of arachidonic acid that is observed in TLR2-/- and MyD88-/- macrophages infected with WTLM and DeltahlyLM correlates with diminished MAPK activation. WTLM but not DeltahlyLM increases intracellular calcium, which is implicated in regulation of cPLA2alpha. Prostaglandin E2, prostaglandin I2, and leukotriene C4 are produced by cPLA2alpha+/+ but not cPLA2alpha-/- macrophages in response to WTLM and DeltahlyLM. Tumor necrosis factor (TNF)-alpha production is significantly lower in cPLA2alpha+/+ than in cPLA2alpha-/- macrophages infected with WTLM and DeltahlyLM. Treatment of infected cPLA2alpha+/+ macrophages with the cyclooxygenase inhibitor indomethacin increases TNFalpha production to the level produced by cPLA2alpha-/- macrophages implicating prostaglandins in TNFalpha down-regulation. Therefore activation of cPLA2alpha in macrophages may impact immune responses to L. monocytogenes.
Collapse
Affiliation(s)
- Shahid Noor
- Department of Pediatrics, National Jewish Medical and Research Center, Denver, Colorado 80206
| | - Howard Goldfine
- Department of Microbiology, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Dawn E. Tucker
- Department of Pediatrics, National Jewish Medical and Research Center, Denver, Colorado 80206
| | - Saritha Suram
- Department of Pediatrics, National Jewish Medical and Research Center, Denver, Colorado 80206
| | - Laurel L. Lenz
- Department of Immunology, National Jewish Medical and Research Center, Denver, Colorado 80206
| | - Shizuo Akira
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Satoshi Uematsu
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Milena Girotti
- Department of Psychology, University of Colorado, Boulder, Colorado 80309
| | | | - Kevin Breuel
- ETSU Clinical Labs, ETSU Physicians and Associates, Johnson City, Tennessee 37604
| | - David L. Williams
- Department of Surgery, James H. Quillen College of Medicine, Johnson City, Tennessee 37614
| | - Christina C. Leslie
- Department of Pediatrics, National Jewish Medical and Research Center, Denver, Colorado 80206
- Departments of Pathology and Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| |
Collapse
|
40
|
Dussurget O. Chapter 1 New Insights into Determinants of Listeria Monocytogenes Virulence. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2008; 270:1-38. [DOI: 10.1016/s1937-6448(08)01401-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
41
|
Bierne H, Cossart P. Listeria monocytogenes surface proteins: from genome predictions to function. Microbiol Mol Biol Rev 2007; 71:377-97. [PMID: 17554049 PMCID: PMC1899877 DOI: 10.1128/mmbr.00039-06] [Citation(s) in RCA: 179] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The genome of the human food-borne pathogen Listeria monocytogenes is predicted to encode a high number of surface proteins. This abundance likely reflects the ability of this bacterium to survive in diverse environments, including soil, food, and the human host. This review focuses on the various mechanisms by which listerial proteins are attached at the bacterial surface and their many functions, including peptidoglycan metabolism, protein processing, adhesion to host cells, and invasion of host tissues. Extensive in silico analysis of the domains or motifs present in these mosaic proteins reveals that diverse structural features allow the surface proteome to interact with diverse bacterial or host components. This diversity offers new clues about the molecular bases of Listeria pathogenesis.
Collapse
Affiliation(s)
- Hélène Bierne
- Institut Pasteur, Unité des Interactions Bactéries Cellules, Paris F-75015, France.
| | | |
Collapse
|