1
|
Wheeler R, Gomperts Boneca I. The hidden base of the iceberg: gut peptidoglycome dynamics is foundational to its influence on the host. Gut Microbes 2024; 16:2395099. [PMID: 39239828 PMCID: PMC11382707 DOI: 10.1080/19490976.2024.2395099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 07/01/2024] [Accepted: 08/16/2024] [Indexed: 09/07/2024] Open
Abstract
The intestinal microbiota of humans includes a highly diverse range of bacterial species. All these bacteria possess a cell wall, composed primarily of the macromolecule peptidoglycan. As such, the gut also harbors an abundant and varied peptidoglycome. A remarkable range of host physiological pathways are regulated by peptidoglycan fragments that originate from the gut microbiota and enter the host system. Interactions between the host system and peptidoglycan can influence physiological development and homeostasis, promote health, or contribute to inflammatory disease. Underlying these effects is the interplay between microbiota composition and enzymatic processes that shape the intestinal peptidoglycome, dictating the types of peptidoglycan generated, that subsequently cross the gut barrier. In this review, we highlight and discuss the hidden and emerging functional aspects of the microbiome, i.e. the hidden base of the iceberg, that modulate the composition of gut peptidoglycan, and how these fundamental processes are drivers of physiological outcomes for the host.
Collapse
Affiliation(s)
- Richard Wheeler
- Institut Pasteur, Université Paris Cité, Paris, France
- Hauts-de-Seine, Arthritis Research and Development, Neuilly-sur-Seine, France
| | | |
Collapse
|
2
|
Guryanova SV. Regulation of Immune Homeostasis via Muramyl Peptides-Low Molecular Weight Bioregulators of Bacterial Origin. Microorganisms 2022; 10:1526. [PMID: 36013944 PMCID: PMC9413341 DOI: 10.3390/microorganisms10081526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 12/10/2022] Open
Abstract
Metabolites and fragments of bacterial cells play an important role in the formation of immune homeostasis. Formed in the course of evolution, symbiotic relationships between microorganisms and a macroorganism are manifested, in particular, in the regulation of numerous physiological functions of the human body by the innate immunity receptors. Low molecular weight bioregulators of bacterial origin have recently attracted more and more attention as drugs in the prevention and composition of complex therapy for a wide range of diseases of bacterial and viral etiology. Signaling networks show cascades of causal relationships of deterministic phenomena that support the homeostasis of multicellular organisms at different levels. To create networks, data from numerous biomedical and clinical research databases were used to prepare expert systems for use in pharmacological and biomedical research with an emphasis on muramyl dipeptides. Muramyl peptides are the fragments of the cell wall of Gram-positive and Gram-negative bacteria. Binding of muramyl peptides with intracellular NOD2 receptors is crucial for an immune response on pathogens. Depending on the microenvironment and duration of action, muramyl peptides possess positive or negative regulation of inflammation. Other factors, such as genetic, pollutions, method of application and stress also contribute and should be taken into account. A system biology approach should be used in order to systemize all experimental data for rigorous analysis, with the aim of understanding intrinsic pathways of homeostasis, in order to define precise medicine therapy and drug design.
Collapse
Affiliation(s)
- Svetlana V Guryanova
- Medical Institute, Peoples' Friendship University of Russia (RUDN University) of the Ministry of Science and Higher Education of the Russian Federation, 117198 Moscow, Russia
| |
Collapse
|
3
|
Trindade BC, Chen GY. NOD1 and NOD2 in inflammatory and infectious diseases. Immunol Rev 2020; 297:139-161. [PMID: 32677123 DOI: 10.1111/imr.12902] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 12/12/2022]
Abstract
It has been long recognized that NOD1 and NOD2 are critical players in the host immune response, primarily by their sensing bacterial peptidoglycan-conserved motifs. Significant advances have been made from efforts that characterize their upstream activators, assembly of signaling complexes, and activation of downstream signaling pathways. Disruption in NOD1 and NOD2 signaling has also been associated with impaired host defense and resistance to the development of inflammatory diseases. In this review, we will describe how NOD1 and NOD2 sense microbes and cellular stress to regulate host responses that can affect disease pathogenesis and outcomes.
Collapse
Affiliation(s)
- Bruno C Trindade
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Grace Y Chen
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
4
|
Sharma A. Persistence of Tannerella forsythia and Fusobacterium nucleatum in dental plaque: a strategic alliance. CURRENT ORAL HEALTH REPORTS 2020; 7:22-28. [PMID: 36779221 PMCID: PMC9917731 DOI: 10.1007/s40496-020-00254-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
PURPOSE OF REVIEW The Gram-negative oral pathogen Tannerella forsythia is implicated in the pathogenesis of periodontitis, an inflammatory disease characterized by progressive destruction of the tooth supporting structures affecting over 700 million people worldwide. This review highlights the basis of why and how T. forsythia interacts with Fusobacterium nucleatum, a bacterium considered to be a bridge between the early and late colonizing bacteria of the dental plaque. RECENT FINDINGS The recent findings indicate that these two organisms have a strong mutualistic relationship that involves foraging by T. forsythia on F. nucleatum peptidoglycan and utilization of glucose, released by the hydrolytic activity of T. forsythia glucanase, as a nutrient by F. nucleatum. In addition, T. forsythia has the unique ability to generate a toxic and inflammogenic compound, methylglyoxal, from glucose. This compound can induce inflammation, leading to the degradation of periodontal tissues and release of host components as nutrients for bacteria to further exacerbate the disease. SUMMARY In summary, this article will present our current understanding of mechanisms underpinning T. forsythia-F. nucleatum mutualism, and how this mutualism might impact periodontal disease progression.
Collapse
Affiliation(s)
- Ashu Sharma
- Department of Oral Biology, School of Dental Medicine, 3435 Main Street, University at Buffalo, State University of New York, Buffalo, NY 14214
| |
Collapse
|
5
|
Borrelia burgdorferi peptidoglycan is a persistent antigen in patients with Lyme arthritis. Proc Natl Acad Sci U S A 2019; 116:13498-13507. [PMID: 31209025 PMCID: PMC6613144 DOI: 10.1073/pnas.1904170116] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Lyme disease, caused by the spirochete Borrelia burgdorferi, is the most common vector-borne disease in North America. If early infection is untreated, it can result in late-stage manifestations, including arthritis. Although antibiotics are generally effective at all stages of the disease, arthritis may persist in some patients for months to several years despite oral and intravenous antibiotic treatment. Excessive, dysregulated host immune responses are thought to play an important role in this outcome, but the underlying mechanisms are not completely understood. This study identifies the B. burgdorferi peptidoglycan, a major component of the cell wall, as an immunogen likely to contribute to inflammation during infection and in cases of postinfectious Lyme arthritis. Lyme disease is a multisystem disorder caused by the spirochete Borrelia burgdorferi. A common late-stage complication of this disease is oligoarticular arthritis, often involving the knee. In ∼10% of cases, arthritis persists after appropriate antibiotic treatment, leading to a proliferative synovitis typical of chronic inflammatory arthritides. Here, we provide evidence that peptidoglycan (PG), a major component of the B. burgdorferi cell envelope, may contribute to the development and persistence of Lyme arthritis (LA). We show that B. burgdorferi has a chemically atypical PG (PGBb) that is not recycled during cell-wall turnover. Instead, this pathogen sheds PGBb fragments into its environment during growth. Patients with LA mount a specific immunoglobulin G response against PGBb, which is significantly higher in the synovial fluid than in the serum of the same patient. We also detect PGBb in 94% of synovial fluid samples (32 of 34) from patients with LA, many of whom had undergone oral and intravenous antibiotic treatment. These same synovial fluid samples contain proinflammatory cytokines, similar to those produced by human peripheral blood mononuclear cells stimulated with PGBb. In addition, systemic administration of PGBb in BALB/c mice elicits acute arthritis. Altogether, our study identifies PGBb as a likely contributor to inflammatory responses in LA. Persistence of this antigen in the joint may contribute to synovitis after antibiotics eradicate the pathogen. Furthermore, our finding that B. burgdorferi sheds immunogenic PGBb fragments during growth suggests a potential role for PGBb in the immunopathogenesis of other Lyme disease manifestations.
Collapse
|
6
|
Mallick S, Das J, Verma J, Mathew S, Maiti TK, Ghosh AS. Role of Escherichia coli endopeptidases and dd-carboxypeptidases in infection and regulation of innate immune response. Microbes Infect 2019; 21:464-474. [PMID: 31085336 DOI: 10.1016/j.micinf.2019.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 04/21/2019] [Accepted: 04/23/2019] [Indexed: 01/13/2023]
Abstract
The low-molecular-mass penicillin-binding proteins, involved in peptidoglycan recycling can also produce peptidoglycan fragments capable of activating an innate immune response in host. To investigate how these proteins in Enterobacteriaceae play a role to elicit/evade innate immune responses during infections, we deleted certain endopeptidases and dd-carboxypeptidases from Escherichia coli CS109 and studied the viability of these mutants in macrophages. The ability of infected macrophages to exert oxidative killing, express surface activation markers TLR2, MHC class II and release TNFα, were assessed. Immune responses were elevated in macrophages infected with dd-carboxypeptidase mutants but reduced for endopeptidase mutants. However, the NFκB, iNOS, and TLR2 transcripts remained elevated in macrophages infected with both mutant types. Overall, we have shown, under normal conditions endopeptidases have a tendency to elicit the immune response but their effect is suppressed by the presence of dd-carboxypeptidases. Conversely, DD-carboxypeptidases, normally, tend to reduce immune responses, as their deletions enhanced the same in macrophages. Therefore, we conclude that the roles of endopeptidases and dd-carboxypeptidases are possibly counter-active in wild-type cells where either class of enzymes suppresses each other's immunogenic properties rendering overall maintenance of low immunogenicity that helps E. coli in evading the host immune responses.
Collapse
Affiliation(s)
- Sathi Mallick
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal, 721302, India
| | - Joyjyoti Das
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal, 721302, India
| | - Jyoti Verma
- Advanced Technology Development Centre, Indian Institute of Technology Kharagpur, West Bengal, 721302, India
| | - Samatha Mathew
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal, 721302, India
| | - Tapas K Maiti
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal, 721302, India
| | - Anindya S Ghosh
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal, 721302, India.
| |
Collapse
|
7
|
Irazoki O, Hernandez SB, Cava F. Peptidoglycan Muropeptides: Release, Perception, and Functions as Signaling Molecules. Front Microbiol 2019; 10:500. [PMID: 30984120 PMCID: PMC6448482 DOI: 10.3389/fmicb.2019.00500] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 02/27/2019] [Indexed: 12/12/2022] Open
Abstract
Peptidoglycan (PG) is an essential molecule for the survival of bacteria, and thus, its biosynthesis and remodeling have always been in the spotlight when it comes to the development of antibiotics. The peptidoglycan polymer provides a protective function in bacteria, but at the same time is continuously subjected to editing activities that in some cases lead to the release of peptidoglycan fragments (i.e., muropeptides) to the environment. Several soluble muropeptides have been reported to work as signaling molecules. In this review, we summarize the mechanisms involved in muropeptide release (PG breakdown and PG recycling) and describe the known PG-receptor proteins responsible for PG sensing. Furthermore, we overview the role of muropeptides as signaling molecules, focusing on the microbial responses and their functions in the host beyond their immunostimulatory activity.
Collapse
Affiliation(s)
| | | | - Felipe Cava
- Laboratory for Molecular Infection Medicine Sweden, Department of Molecular Biology, Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| |
Collapse
|
8
|
Schaub RE, Dillard JP. The Pathogenic Neisseria Use a Streamlined Set of Peptidoglycan Degradation Proteins for Peptidoglycan Remodeling, Recycling, and Toxic Fragment Release. Front Microbiol 2019; 10:73. [PMID: 30766523 PMCID: PMC6365954 DOI: 10.3389/fmicb.2019.00073] [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: 10/06/2018] [Accepted: 01/15/2019] [Indexed: 12/26/2022] Open
Abstract
Neisseria gonorrhoeae and Neisseria meningitidis release peptidoglycan (PG) fragments from the cell as the bacteria grow. For N. gonorrhoeae these PG fragments are known to cause damage to human Fallopian tube tissue in organ culture that mimics the damage seen in patients with pelvic inflammatory disease. N. meningitidis also releases pro-inflammatory PG fragments, but in smaller amounts than those from N. gonorrhoeae. It is not yet known if PG fragment release contributes to the highly inflammatory conditions of meningitis and meningococcemia caused by N. meningitidis. Examination of the mechanisms of PG degradation and recycling identified proteins required for these processes. In comparison to the model organism E. coli, the pathogenic Neisseria have far fewer PG degradation proteins, and some of these proteins show differences in subcellular localization compared to their E. coli homologs. In particular, some N. gonorrhoeae PG degradation proteins were demonstrated to be in the outer membrane while their homologs in E. coli were found free in the periplasm or in the cytoplasm. The localization of two of these proteins was demonstrated to affect PG fragment release. Another major factor for PG fragment release is the allele of ampG. Gonococcal AmpG was found to be slightly defective compared to related PG fragment permeases, thus leading to increased release of PG. A number of additional PG-related factors affect other virulence functions in Neisseria. Endopeptidases and carboxypeptidases were found to be required for type IV pilus production and resistance to hydrogen peroxide. Also, deacetylation of PG was required for virulence of N. meningitidis as well as normal cell size. Overall, we describe the processes involved in PG degradation and recycling and how certain characteristics of these proteins influence the interactions of these pathogens with their host.
Collapse
Affiliation(s)
- Ryan E Schaub
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Joseph P Dillard
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
9
|
Role of a fluid-phase PRR in fighting an intracellular pathogen: PTX3 in Shigella infection. PLoS Pathog 2018; 14:e1007469. [PMID: 30532257 PMCID: PMC6317801 DOI: 10.1371/journal.ppat.1007469] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/03/2019] [Accepted: 11/15/2018] [Indexed: 12/31/2022] Open
Abstract
Shigella spp. are pathogenic bacteria that cause bacillary dysentery in humans by invading the colonic and rectal mucosa where they induce dramatic inflammation. Here, we have analyzed the role of the soluble PRR Pentraxin 3 (PTX3), a key component of the humoral arm of innate immunity. Mice that had been intranasally infected with S. flexneri were rescued from death by treatment with recombinant PTX3. In vitro PTX3 exerts the antibacterial activity against Shigella, impairing epithelial cell invasion and contributing to the bactericidal activity of serum. PTX3 is produced upon LPS-TLR4 stimulation in accordance with the lipid A structure of Shigella. In the plasma of infected patients, the level of PTX3 amount only correlates strongly with symptom severity. These results signal PTX3 as a novel player in Shigella pathogenesis and its potential role in fighting shigellosis. Finally, we suggest that the plasma level of PTX3 in shigellosis patients could act as a biomarker for infection severity. Soluble pattern recognition molecules, PRMs, are components of the humoral arm of innate immunity. The long pentraxin 3, PTX3, is a prototypic soluble PRM that is produced in response to primary inflammatory signals. Shigella spp. are human entero-pathogens which invade colonic and rectal mucosa where they cause deleterious inflammation. We show that PTX3 acts as an ante-antibody and contributes to the clearance of extracellular Shigella. As a countermeasure, Shigella uses invasiveness and low-inflammatory LPS to control PTX3 release in infected cells. This study highlights that the extracellular phase of the invasion process can be considered the “Achille heels” of Shigella pathogenesis.
Collapse
|
10
|
Juan C, Torrens G, Barceló IM, Oliver A. Interplay between Peptidoglycan Biology and Virulence in Gram-Negative Pathogens. Microbiol Mol Biol Rev 2018; 82:e00033-18. [PMID: 30209071 PMCID: PMC6298613 DOI: 10.1128/mmbr.00033-18] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The clinical and epidemiological threat of the growing antimicrobial resistance in Gram-negative pathogens, particularly for β-lactams, the most frequently used and relevant antibiotics, urges research to find new therapeutic weapons to combat the infections caused by these microorganisms. An essential previous step in the development of these therapeutic solutions is to identify their potential targets in the biology of the pathogen. This is precisely what we sought to do in this review specifically regarding the barely exploited field analyzing the interplay among the biology of the peptidoglycan and related processes, such as β-lactamase regulation and virulence. Hence, here we gather, analyze, and integrate the knowledge derived from published works that provide information on the topic, starting with those dealing with the historically neglected essential role of the Gram-negative peptidoglycan in virulence, including structural, biogenesis, remodeling, and recycling aspects, in addition to proinflammatory and other interactions with the host. We also review the complex link between intrinsic β-lactamase production and peptidoglycan metabolism, as well as the biological costs potentially associated with the expression of horizontally acquired β-lactamases. Finally, we analyze the existing evidence from multiple perspectives to provide useful clues for identifying targets enabling the future development of therapeutic options attacking the peptidoglycan-virulence interconnection as a key weak point of the Gram-negative pathogens to be used, if not to kill the bacteria, to mitigate their capacity to produce severe infections.
Collapse
Affiliation(s)
- Carlos Juan
- Servicio de Microbiología and Unidad de Investigación, Hospital Son Espases, Instituto de Investigación Sanitaria de Baleares (IdISBa), Palma, Spain
| | - Gabriel Torrens
- Servicio de Microbiología and Unidad de Investigación, Hospital Son Espases, Instituto de Investigación Sanitaria de Baleares (IdISBa), Palma, Spain
| | - Isabel Maria Barceló
- Servicio de Microbiología and Unidad de Investigación, Hospital Son Espases, Instituto de Investigación Sanitaria de Baleares (IdISBa), Palma, Spain
| | - Antonio Oliver
- Servicio de Microbiología and Unidad de Investigación, Hospital Son Espases, Instituto de Investigación Sanitaria de Baleares (IdISBa), Palma, Spain
| |
Collapse
|
11
|
The Structure of ampG Gene in Pseudomonas aeruginosa and Its Effect on Drug Resistance. CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY 2018; 2018:7170416. [PMID: 30598711 PMCID: PMC6287161 DOI: 10.1155/2018/7170416] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 08/20/2018] [Accepted: 09/24/2018] [Indexed: 11/18/2022]
Abstract
In order to study the relationship between the structure and function of AmpG, structure, site-specific mutation, and gene complementary experiments have been performed against the clinical isolates of Pseudomonas aeruginosa. We found that there are 51 nucleotide variations at 34 loci over the ampG genes from 24 of 35 P. aeruginosa strains detected, of which 7 nucleotide variations resulted in amino acid change. The ampG variants with the changed nucleotides (amino acids) could complement the function of ampG deleted PA01 (PA01ΔG). The ampicillin minimum inhibitory concentration (MIC) of PA01ΔG complemented with 32 ampG variants was up to 512 μg/ml, similar to the original PA01 (P. aeruginosa PA01). Furthermore, site-directed mutation of two conservative amino acids (I53 and W90) showed that when I53 was mutated to 53S or 53T (I53S or I53T), the ampicillin MIC level dropped drastically, and the activity of AmpC β-lactamase decreased as well. By contrast, the ampicillin MIC and the activity of AmpC β-lactamase remained unchanged for W90R and W90S mutants. Our studies demonstrated that although nucleotide variations occurred in most of the ampG genes, the structure of AmpG protein in clinical isolates is stable, and conservative amino acid is necessary to maintain normal function of AmpG.
Collapse
|
12
|
Antibiotic Targets in Gonococcal Cell Wall Metabolism. Antibiotics (Basel) 2018; 7:antibiotics7030064. [PMID: 30037076 PMCID: PMC6164560 DOI: 10.3390/antibiotics7030064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 07/19/2018] [Accepted: 07/19/2018] [Indexed: 12/14/2022] Open
Abstract
The peptidoglycan cell wall that encloses the bacterial cell and provides structural support and protection is remodeled by multiple enzymes that synthesize and cleave the polymer during growth. This essential and dynamic structure has been targeted by multiple antibiotics to treat gonococcal infections. Up until now, antibiotics have been used against the biosynthetic machinery and the therapeutic potential of inhibiting enzymatic activities involved in peptidoglycan breakdown has not been explored. Given the major antibiotic resistance problems we currently face, it is crucial to identify other possible targets that are key to maintaining cell integrity and contribute to disease development. This article reviews peptidoglycan as an antibiotic target, how N. gonorrhoeae has developed resistance to currently available antibiotics, and the potential of continuing to target this essential structure to combat gonococcal infections by attacking alternative enzymatic activities involved in cell wall modification and metabolism.
Collapse
|
13
|
Corridoni D, Chapman T, Ambrose T, Simmons A. Emerging Mechanisms of Innate Immunity and Their Translational Potential in Inflammatory Bowel Disease. Front Med (Lausanne) 2018. [PMID: 29515999 PMCID: PMC5825991 DOI: 10.3389/fmed.2018.00032] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Activation of the innate immune system through pattern-recognition receptor (PRR) signaling plays a pivotal role in the early induction of host defense following exposure to pathogens. Loss of intestinal innate immune regulation leading aberrant immune responses has been implicated in the pathogenesis of inflammatory bowel disease (IBD). The precise role of PRRs in gut inflammation is not well understood, but considering their role as bacterial sensors and their genetic association with IBD, they likely contribute to dysregulated immune responses to the commensal microbiota. The purpose of this review is to evaluate the emerging functions of PRRs including their functional cross-talk, how they respond to mitochondrial damage, induce mitophagy or autophagy, and influence adaptive immune responses by interacting with the antigen presentation machinery. The review also summarizes some of the recent attempts to harness these pathways for therapeutic approaches in intestinal inflammation.
Collapse
Affiliation(s)
- Daniele Corridoni
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom.,Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Thomas Chapman
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom.,Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Tim Ambrose
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom.,Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Alison Simmons
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom.,Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
14
|
Ruscitto A, Sharma A. Peptidoglycan synthesis in Tannerella forsythia: Scavenging is the modus operandi. Mol Oral Microbiol 2018; 33:125-132. [PMID: 29247483 DOI: 10.1111/omi.12210] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2017] [Indexed: 01/05/2023]
Abstract
Tannerella forsythia is a Gram-negative oral pathogen strongly associated with periodontitis. This bacterium has an absolute requirement for exogenous N-acetylmuramic acid (MurNAc), an amino sugar that forms the repeating disaccharide unit with amino sugar N-acetylglucosamine (GlcNAc) of the peptidoglycan backbone. In silico genome analysis indicates that T. forsythia lacks the key biosynthetic enzymes needed for the de novo synthesis of MurNAc, and so relies on alternative ways to meet its requirement for peptidoglycan biosynthesis. In the subgingival niche, the bacterium can acquire MurNAc and peptidoglycan fragments (muropeptides) released by the cohabiting bacteria during their cell wall breakdown associated with cell division. Tannerella forsythia is able to also use host sialic acid (Neu5Ac) in lieu of MurNAc or muropeptides for its survival during the biofilm growth. Evidence suggests that the bacterium might be able to shunt sialic acid into a metabolic pathway leading to peptidoglycan synthesis. In this review, we explore the mechanisms by which T. forsythia is able to scavenge MurNAc, muropeptide and sialic acid for its peptidoglycan synthesis, and the impact of these scavenging activities on pathogenesis.
Collapse
Affiliation(s)
| | - A Sharma
- Department of Oral Biology, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
15
|
Chan JM, Dillard JP. Attention Seeker: Production, Modification, and Release of Inflammatory Peptidoglycan Fragments in Neisseria Species. J Bacteriol 2017; 199:e00354-17. [PMID: 28674065 PMCID: PMC5637178 DOI: 10.1128/jb.00354-17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Maintenance of the structural macromolecule peptidoglycan (PG), which involves regulated cycles of PG synthesis and PG degradation, is pivotal for cellular integrity and survival. PG fragments generated from the degradation process are usually efficiently recycled by Gram-negative bacteria. However, Neisseria gonorrhoeae and a limited number of Gram-negative bacteria release PG fragments in amounts sufficient to induce host tissue inflammation and damage during an infection. Due to limited redundancy in PG-modifying machineries and genetic tractability, N. gonorrhoeae serves as a great model organism for the study of biological processes related to PG. This review summarizes the generation, modification, and release of inflammatory PG molecules by N. gonorrhoeae and related species and discusses these findings in the context of understanding bacterial physiology and pathogenesis.
Collapse
Affiliation(s)
- Jia Mun Chan
- Department of Medical Microbiology & Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Joseph P Dillard
- Department of Medical Microbiology & Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
16
|
Abstract
Lysozyme is a cornerstone of innate immunity. The canonical mechanism for bacterial killing by lysozyme occurs through the hydrolysis of cell wall peptidoglycan (PG). Conventional type (c-type) lysozymes are also highly cationic and can kill certain bacteria independently of PG hydrolytic activity. Reflecting the ongoing arms race between host and invading microorganisms, both gram-positive and gram-negative bacteria have evolved mechanisms to thwart killing by lysozyme. In addition to its direct antimicrobial role, more recent evidence has shown that lysozyme modulates the host immune response to infection. The degradation and lysis of bacteria by lysozyme enhance the release of bacterial products, including PG, that activate pattern recognition receptors in host cells. Yet paradoxically, lysozyme is important for the resolution of inflammation at mucosal sites. This review will highlight recent advances in our understanding of the diverse mechanisms that bacteria use to protect themselves against lysozyme, the intriguing immunomodulatory function of lysozyme, and the relationship between these features in the context of infection.
Collapse
Affiliation(s)
- Stephanie A. Ragland
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Alison K. Criss
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
17
|
Balasubramanian I, Gao N. From sensing to shaping microbiota: insights into the role of NOD2 in intestinal homeostasis and progression of Crohn's disease. Am J Physiol Gastrointest Liver Physiol 2017; 313:G7-G13. [PMID: 28450278 PMCID: PMC5538831 DOI: 10.1152/ajpgi.00330.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 04/06/2017] [Accepted: 04/20/2017] [Indexed: 01/31/2023]
Abstract
NOD2 was the first susceptibility gene identified for Crohn's disease (CD), one of the major forms of inflammatory bowel disease (IBD). The field of NOD2 research has opened up many questions critical to understanding the complexities of microbiota-host interactions. In addition to sensing its specific bacterial components as a cytosolic pattern recognition receptor, NOD2 also appears to shape the colonization of intestinal microbiota. Activated NOD2 triggers downstream signaling cascades exampled by the NF-κB pathway to induce antimicrobial activities, however, defective or loss of NOD2 functions incur a similarly activated inflammatory response. Additional studies have identified the involvement of NOD2 in protection against non-microbiota-related intestinal damages as well as extraintestinal infections. We survey recent molecular and genetic studies of NOD2-mediated bacterial sensing and immunological modulation, and integrate evidence to suggest a highly reciprocal but still poorly understood cross talk between enteric microbiota and host cells.
Collapse
Affiliation(s)
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark, New Jersey
| |
Collapse
|
18
|
Lorenzo FD, Palmigiano A, Paciello I, Pallach M, Garozzo D, Bernardini ML, Cono VL, Yakimov MM, Molinaro A, Silipo A. The Deep-Sea Polyextremophile Halobacteroides lacunaris TB21 Rough-Type LPS: Structure and Inhibitory Activity towards Toxic LPS. Mar Drugs 2017; 15:md15070201. [PMID: 28653982 PMCID: PMC5532643 DOI: 10.3390/md15070201] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 06/12/2017] [Accepted: 06/22/2017] [Indexed: 11/24/2022] Open
Abstract
The structural characterization of the lipopolysaccharide (LPS) from extremophiles has important implications in several biomedical and therapeutic applications. The polyextremophile Gram-negative bacterium Halobacteroideslacunaris TB21, isolated from one of the most extreme habitats on our planet, the deep-sea hypersaline anoxic basin Thetis, represents a fascinating microorganism to investigate in terms of its LPS component. Here we report the elucidation of the full structure of the R-type LPS isolated from H. lacunaris TB21 that was attained through a multi-technique approach comprising chemical analyses, NMR spectroscopy, and Matrix-Assisted Laser Desorption Ionization (MALDI) mass spectrometry. Furthermore, cellular immunology studies were executed on the pure R-LPS revealing a very interesting effect on human innate immunity as an inhibitor of the toxic Escherichia coli LPS.
Collapse
Affiliation(s)
- Flaviana Di Lorenzo
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy.
| | - Angelo Palmigiano
- CNR-Istituto per i Polimeri, Compositi e Biomateriali IPCB-Unità di Catania, 95126 Catania, Italy.
| | - Ida Paciello
- Department of Biology and Biotechnology "Charles Darwin", Sapienza-University of Rome, 00185 Rome, Italy.
| | - Mateusz Pallach
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy.
| | - Domenico Garozzo
- CNR-Istituto per i Polimeri, Compositi e Biomateriali IPCB-Unità di Catania, 95126 Catania, Italy.
| | - Maria-Lina Bernardini
- Department of Biology and Biotechnology "Charles Darwin", Sapienza-University of Rome, 00185 Rome, Italy.
| | - Violetta La Cono
- Marine Molecular Microbiology & Biotechnology, CNR-Institute for Coastal Marine Environment, 98122 Messina, Italy.
| | - Michail M Yakimov
- Marine Molecular Microbiology & Biotechnology, CNR-Institute for Coastal Marine Environment, 98122 Messina, Italy.
- Immanuel Kant Baltic Federal University, 236040 Kaliningrad, Russia.
| | - Antonio Molinaro
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy.
| | - Alba Silipo
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy.
| |
Collapse
|
19
|
Leuzzi A, Grossi M, Di Martino ML, Pasqua M, Micheli G, Colonna B, Prosseda G. Role of the SRRz/Rz 1 lambdoid lysis cassette in the pathoadaptive evolution of Shigella. Int J Med Microbiol 2017; 307:268-275. [PMID: 28389211 DOI: 10.1016/j.ijmm.2017.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 03/21/2017] [Accepted: 03/27/2017] [Indexed: 12/25/2022] Open
Abstract
Shigella, the etiological agent of bacillary dysentery (shigellosis), is a highly adapted human pathogen. It evolved from an innocuous ancestor resembling the Escherichia coli strain by gain and loss of genes and functions. While the gain process concerns the acquisition of the genetic determinants of virulence, the loss is related to the adaptation of the genome to the new pathogenic status and occurs by pathoadaptive mutation of antivirulence genes. In this study, we highlight that the SRRz/Rz1 lambdoid lysis cassette, even though stably adopted in E. coli K12 by virtue of its beneficial effect on cell physiology, has undergone a significant decay in Shigella. Moreover, we show the antivirulence nature of the SRRz/Rz1 lysis cassette in Shigella. In fact, by restoring the SRRz/Rz1 expression in this pathogen, we observe an increased release of peptidoglycan fragments, causing an unbalance in the fine control exerted by Shigella on host innate immunity and a mitigation of its virulence. This strongly affects the virulence of Shigella and allows to consider the loss of SRRz/Rz1 lysis cassette as another pathoadaptive event in the life of Shigella.
Collapse
Affiliation(s)
- Adriano Leuzzi
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Dipartimento di Biologia e Biotecnologie "C. Darwin", Sapienza Università di Roma, Roma, Italy
| | - Milena Grossi
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Dipartimento di Biologia e Biotecnologie "C. Darwin", Sapienza Università di Roma, Roma, Italy
| | - Maria Letizia Di Martino
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Dipartimento di Biologia e Biotecnologie "C. Darwin", Sapienza Università di Roma, Roma, Italy
| | - Martina Pasqua
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Dipartimento di Biologia e Biotecnologie "C. Darwin", Sapienza Università di Roma, Roma, Italy
| | - Gioacchino Micheli
- Istituto di Biologia e Patologia Molecolari, Consiglio Nazionale delle Ricerche, Roma, Italy
| | - Bianca Colonna
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Dipartimento di Biologia e Biotecnologie "C. Darwin", Sapienza Università di Roma, Roma, Italy
| | - Gianni Prosseda
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Dipartimento di Biologia e Biotecnologie "C. Darwin", Sapienza Università di Roma, Roma, Italy.
| |
Collapse
|
20
|
Darzi Y, Jiao Y, Hasegawa M, Moon H, Núñez G, Inohara N, Raes J. The Genomic Sequence of the Oral Pathobiont Strain NI1060 Reveals Unique Strategies for Bacterial Competition and Pathogenicity. PLoS One 2016; 11:e0158866. [PMID: 27409077 PMCID: PMC4943601 DOI: 10.1371/journal.pone.0158866] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 06/23/2016] [Indexed: 11/19/2022] Open
Abstract
Strain NI1060 is an oral bacterium responsible for periodontitis in a murine ligature-induced disease model. To better understand its pathogenicity, we have determined the complete sequence of its 2,553,982 bp genome. Although closely related to Pasteurella pneumotropica, a pneumonia-associated rodent commensal based on its 16S rRNA, the NI1060 genomic content suggests that they are different species thriving on different energy sources via alternative metabolic pathways. Genomic and phylogenetic analyses showed that strain NI1060 is distinct from the genera currently described in the family Pasteurellaceae, and is likely to represent a novel species. In addition, we found putative virulence genes involved in lipooligosaccharide synthesis, adhesins and bacteriotoxic proteins. These genes are potentially important for host adaption and for the induction of dysbiosis through bacterial competition and pathogenicity. Importantly, strain NI1060 strongly stimulates Nod1, an innate immune receptor, but is defective in two peptidoglycan recycling genes due to a frameshift mutation. The in-depth analysis of its genome thus provides critical insights for the development of NI1060 as a prime model system for infectious disease.
Collapse
Affiliation(s)
- Youssef Darzi
- Department of Bioengineering Sciences, Microbiology Unit, Vrije Universiteit Brussel, Brussels, Belgium
- Center for the Biology of Disease, VIB, Leuven, Belgium
- Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium
| | - Yizu Jiao
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Mizuho Hasegawa
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Henry Moon
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Gabriel Núñez
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States of America
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Naohiro Inohara
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail: (JR); (NI)
| | - Jeroen Raes
- Department of Bioengineering Sciences, Microbiology Unit, Vrije Universiteit Brussel, Brussels, Belgium
- Center for the Biology of Disease, VIB, Leuven, Belgium
- Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium
- * E-mail: (JR); (NI)
| |
Collapse
|
21
|
The Orchestra and Its Maestro: Shigella's Fine-Tuning of the Inflammasome Platforms. Curr Top Microbiol Immunol 2016; 397:91-115. [PMID: 27460806 DOI: 10.1007/978-3-319-41171-2_5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Shigella spp. are the causative agents of bacillary dysentery, leading to extensive mortality and morbidity worldwide. These facultative intracellular bacteria invade the epithelium of the colon and the rectum, inducing a severe inflammatory response from which the symptoms of the disease originate. Shigella are human pathogens able to manipulate and subvert the innate immune system surveillance. Shigella dampens inflammasome activation in epithelial cells. In infected macrophages, inflammasome activation and IL-1β and IL-18 release lead to massive neutrophil recruitment and greatly contribute to inflammation. Here, we describe how Shigella hijacks and finely tunes inflammasome activation in the different cell populations involved in pathogenesis: epithelial cells, macrophages, neutrophils, DCs, and B and T lymphocytes. Shigella emerges as a "sly" pathogen that switches on/off the inflammasome mechanisms in order to optimize the interaction with the host and establish a successful infection.
Collapse
|
22
|
Bryant CE, Orr S, Ferguson B, Symmons MF, Boyle JP, Monie TP. International Union of Basic and Clinical Pharmacology. XCVI. Pattern recognition receptors in health and disease. Pharmacol Rev 2015; 67:462-504. [PMID: 25829385 DOI: 10.1124/pr.114.009928] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Since the discovery of Toll, in the fruit fly Drosophila melanogaster, as the first described pattern recognition receptor (PRR) in 1996, many families of these receptors have been discovered and characterized. PRRs play critically important roles in pathogen recognition to initiate innate immune responses that ultimately link to the generation of adaptive immunity. Activation of PRRs leads to the induction of immune and inflammatory genes, including proinflammatory cytokines and chemokines. It is increasingly clear that many PRRs are linked to a range of inflammatory, infectious, immune, and chronic degenerative diseases. Several drugs to modulate PRR activity are already in clinical trials and many more are likely to appear in the near future. Here, we review the different families of mammalian PRRs, the ligands they recognize, the mechanisms of activation, their role in disease, and the potential of targeting these proteins to develop the anti-inflammatory therapeutics of the future.
Collapse
Affiliation(s)
- Clare E Bryant
- Departments of Veterinary Medicine (C.E.B., J.P.B., T.P.M.), Pathology (B.F.), and Biochemistry (M.F.S., J.P.B.), University of Cambridge, Cambridge, United Kingdom; and Institute of Infection and Immunity, Cardiff University, Cardiff, United Kingdom (S.O.)
| | - Selinda Orr
- Departments of Veterinary Medicine (C.E.B., J.P.B., T.P.M.), Pathology (B.F.), and Biochemistry (M.F.S., J.P.B.), University of Cambridge, Cambridge, United Kingdom; and Institute of Infection and Immunity, Cardiff University, Cardiff, United Kingdom (S.O.)
| | - Brian Ferguson
- Departments of Veterinary Medicine (C.E.B., J.P.B., T.P.M.), Pathology (B.F.), and Biochemistry (M.F.S., J.P.B.), University of Cambridge, Cambridge, United Kingdom; and Institute of Infection and Immunity, Cardiff University, Cardiff, United Kingdom (S.O.)
| | - Martyn F Symmons
- Departments of Veterinary Medicine (C.E.B., J.P.B., T.P.M.), Pathology (B.F.), and Biochemistry (M.F.S., J.P.B.), University of Cambridge, Cambridge, United Kingdom; and Institute of Infection and Immunity, Cardiff University, Cardiff, United Kingdom (S.O.)
| | - Joseph P Boyle
- Departments of Veterinary Medicine (C.E.B., J.P.B., T.P.M.), Pathology (B.F.), and Biochemistry (M.F.S., J.P.B.), University of Cambridge, Cambridge, United Kingdom; and Institute of Infection and Immunity, Cardiff University, Cardiff, United Kingdom (S.O.)
| | - Tom P Monie
- Departments of Veterinary Medicine (C.E.B., J.P.B., T.P.M.), Pathology (B.F.), and Biochemistry (M.F.S., J.P.B.), University of Cambridge, Cambridge, United Kingdom; and Institute of Infection and Immunity, Cardiff University, Cardiff, United Kingdom (S.O.)
| |
Collapse
|
23
|
Determination of the structure of the O-antigen and the lipid A from the entomopathogenic bacterium Pseudomonas entomophila lipopolysaccharide along with its immunological properties. Carbohydr Res 2015; 412:20-7. [DOI: 10.1016/j.carres.2015.04.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 03/31/2015] [Accepted: 04/17/2015] [Indexed: 02/07/2023]
|
24
|
Boyle JP, Parkhouse R, Monie TP. Insights into the molecular basis of the NOD2 signalling pathway. Open Biol 2015; 4:rsob.140178. [PMID: 25520185 PMCID: PMC4281710 DOI: 10.1098/rsob.140178] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The cytosolic pattern recognition receptor NOD2 is activated by the peptidoglycan fragment muramyl dipeptide to generate a proinflammatory immune response. Downstream effects include the secretion of cytokines such as interleukin 8, the upregulation of pro-interleukin 1β, the induction of autophagy, the production of antimicrobial peptides and defensins, and contributions to the maintenance of the composition of the intestinal microbiota. Polymorphisms in NOD2 are the cause of the inflammatory disorder Blau syndrome and act as susceptibility factors for the inflammatory bowel condition Crohn's disease. The complexity of NOD2 signalling is highlighted by the observation that over 30 cellular proteins interact with NOD2 directly and influence or regulate its functional activity. Previously, the majority of reviews on NOD2 function have focused upon the role of NOD2 in inflammatory disease or in its interaction with and response to microbes. However, the functionality of NOD2 is underpinned by its biochemical interactions. Consequently, in this review, we have taken the opportunity to address the more ‘basic’ elements of NOD2 signalling. In particular, we have focused upon the core interactions of NOD2 with protein factors that influence and modulate the signal transduction pathways involved in NOD2 signalling. Further, where information exists, such as in relation to the role of RIP2, we have drawn comparison with the closely related, but functionally discrete, pattern recognition receptor NOD1. Overall, we provide a comprehensive resource targeted at understanding the complexities of NOD2 signalling.
Collapse
Affiliation(s)
- Joseph P Boyle
- Department of Biochemistry, University of Cambridge, Cambridge, UK Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | | | - Tom P Monie
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK MRC Human Nutrition Research, Elsie Widdowson Laboratory, 120 Fulbourn Road, Cambridge, UK
| |
Collapse
|
25
|
Tomás A, Lery L, Regueiro V, Pérez-Gutiérrez C, Martínez V, Moranta D, Llobet E, González-Nicolau M, Insua JL, Tomas JM, Sansonetti PJ, Tournebize R, Bengoechea JA. Functional Genomic Screen Identifies Klebsiella pneumoniae Factors Implicated in Blocking Nuclear Factor κB (NF-κB) Signaling. J Biol Chem 2015; 290:16678-97. [PMID: 25971969 PMCID: PMC4505419 DOI: 10.1074/jbc.m114.621292] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Indexed: 01/01/2023] Open
Abstract
Klebsiella pneumoniae is an etiologic agent of community-acquired and nosocomial pneumonia. It has been shown that K. pneumoniae infections are characterized by reduced early inflammatory response. Recently our group has shown that K. pneumoniae dampens the activation of inflammatory responses by antagonizing the activation of the NF-κB canonical pathway. Our results revealed that K. pneumoniae capsule polysaccharide (CPS) was necessary but not sufficient to attenuate inflammation. To identify additional Klebsiella factors required to dampen inflammation, we standardized and applied a high-throughput gain-of-function screen to examine a Klebsiella transposon mutant library. We identified 114 mutants that triggered the activation of NF-κB. Two gene ontology categories accounted for half of the loci identified in the screening: metabolism and transport genes (32% of the mutants) and envelope-related genes (17%). Characterization of the mutants revealed that the lack of the enterobactin siderophore was linked to a reduced CPS expression, which in turn underlined the NF-κB activation induced by the mutant. The lipopolysaccharide (LPS) O-polysaccharide and the pullulanase (PulA) type 2 secretion system (T2SS) are required for full effectiveness of the immune evasion. Importantly, these factors do not play a redundant role. The fact that LPS O-polysaccharide and T2SS mutant-induced responses were dependent on TLR2-TLR4-MyD88 activation suggested that LPS O-polysaccharide and PulA perturbed Toll-like receptor (TLR)-dependent recognition of K. pneumoniae. Finally, we demonstrate that LPS O-polysaccharide and pulA mutants are attenuated in the pneumonia mouse model. We propose that LPS O-polysaccharide and PulA T2SS could be new targets for the design of new antimicrobials. Increasing TLR-governed defense responses might provide also selective alternatives for the management of K. pneumoniae pneumonia.
Collapse
Affiliation(s)
- Anna Tomás
- From the Infection and Immunity Program, Fundación de Investigación Sanitaria de las Islas Baleares (FISIB), 07110 Mallorca, Spain, the Instituto de Investigación Sanitaria de Palma (IdisPa), 07120 Mallorca, Spain, the Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| | - Leticia Lery
- the Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, 75724 Paris, France, INSERM U786, 75724 Paris, France
| | - Verónica Regueiro
- From the Infection and Immunity Program, Fundación de Investigación Sanitaria de las Islas Baleares (FISIB), 07110 Mallorca, Spain, the Instituto de Investigación Sanitaria de Palma (IdisPa), 07120 Mallorca, Spain, the Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| | - Camino Pérez-Gutiérrez
- From the Infection and Immunity Program, Fundación de Investigación Sanitaria de las Islas Baleares (FISIB), 07110 Mallorca, Spain, the Instituto de Investigación Sanitaria de Palma (IdisPa), 07120 Mallorca, Spain, the Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| | - Verónica Martínez
- From the Infection and Immunity Program, Fundación de Investigación Sanitaria de las Islas Baleares (FISIB), 07110 Mallorca, Spain, the Instituto de Investigación Sanitaria de Palma (IdisPa), 07120 Mallorca, Spain, the Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| | - David Moranta
- From the Infection and Immunity Program, Fundación de Investigación Sanitaria de las Islas Baleares (FISIB), 07110 Mallorca, Spain, the Instituto de Investigación Sanitaria de Palma (IdisPa), 07120 Mallorca, Spain, the Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| | - Enrique Llobet
- From the Infection and Immunity Program, Fundación de Investigación Sanitaria de las Islas Baleares (FISIB), 07110 Mallorca, Spain, the Instituto de Investigación Sanitaria de Palma (IdisPa), 07120 Mallorca, Spain, the Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| | - Mar González-Nicolau
- From the Infection and Immunity Program, Fundación de Investigación Sanitaria de las Islas Baleares (FISIB), 07110 Mallorca, Spain, the Instituto de Investigación Sanitaria de Palma (IdisPa), 07120 Mallorca, Spain, the Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| | - Jose L Insua
- the Centre for Infection and Immunity, Queen's University Belfast, Belfast BT9 7AE, United Kingdom
| | - Juan M Tomas
- the Departamento de Microbiología, Facultad de Biología, Universidad de Barcelona, 08028 Barcelona, Spain
| | - Philippe J Sansonetti
- the Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, 75724 Paris, France, INSERM U786, 75724 Paris, France, Chaire de Microbiologie et Maladies Infectieuses, Collège de France, 75231 Paris, France
| | - Régis Tournebize
- the Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, 75724 Paris, France, INSERM U786, 75724 Paris, France, Imagopole, Plateforme d'Imagerie Dynamique, Institut Pasteur, 75724 Paris, France, and
| | - José A Bengoechea
- the Centre for Infection and Immunity, Queen's University Belfast, Belfast BT9 7AE, United Kingdom, the Consejo Superior de Investigaciones Científicas (CSIC), 28008 Madrid, Spain
| |
Collapse
|
26
|
Bertsche U, Mayer C, Götz F, Gust AA. Peptidoglycan perception--sensing bacteria by their common envelope structure. Int J Med Microbiol 2014; 305:217-23. [PMID: 25596887 DOI: 10.1016/j.ijmm.2014.12.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Most Eubacteria possess peptidoglycan (PGN) or murein that surrounds the cytoplasmic membrane. While on the one hand this PGN sacculus is a very protective shield that provides resistance to the internal turgor and adverse effects of the environment, it serves on the other hand as a major pattern of recognition due to its unique structure. Eukaryotes harness this particular bacterial macromolecule to perceive (pathogenic) microorganisms and initiate their immune defence. PGN fragments are generated by bacteria as turnover products during bacterial cell wall growth and these fragments can be sensed by plants and animals to assess a potential bacterial threat. To increase the sensitivity the concentration of PGN fragments can be amplified by host hydrolytic enzymes such as lysozyme or amidase. But also bacteria themselves are able to perceive information about the state of their cell wall by sensing small soluble fragments released from its PGN, which eventually leads to the induction of antibiotic responses or cell differentiation. How PGN is sensed by bacteria, plants and animals, and how the antibacterial defence is modulated by PGN perception is the issue of this review.
Collapse
Affiliation(s)
- Ute Bertsche
- Microbial Genetics, Interfaculty Institute for Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, 72076 Tübingen, Germany
| | - Christoph Mayer
- Microbiology/Biotechnology, Interfaculty Institute for Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, 72076 Tübingen, Germany
| | - Friedrich Götz
- Microbial Genetics, Interfaculty Institute for Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, 72076 Tübingen, Germany
| | - Andrea A Gust
- Plant Biochemistry, Center for Plant Molecular Biology (ZMBP), University of Tübingen, 72076 Tübingen, Germany.
| |
Collapse
|
27
|
Abstract
Pattern recognition receptors are essential mediators of host defense and inflammation in the gastrointestinal system. Recent data have revealed that toll-like receptors and nucleotide-binding domain and leucine-rich repeat-containing proteins (NLRs) function to maintain homeostasis between the host microbiome and mucosal immunity. The NLR proteins are a diverse class of cytoplasmic pattern recognition receptors. In humans, only about half of the identified NLRs have been adequately characterized. The majority of well-characterized NLRs participate in the formation of a multiprotein complex, termed the inflammasome, which is responsible for the maturation of interleukin-1β and interleukin-18. However, recent observations have also uncovered the presence of a novel subgroup of NLRs that function as positive or negative regulators of inflammation through modulating critical signaling pathways, including NF-κB. Dysregulation of specific NLRs from both proinflammatory and inhibitory subgroups have been associated with the development of inflammatory bowel disease (IBD) in genetically susceptible human populations. Our own preliminary retrospective data mining efforts have identified a diverse range of NLRs that are significantly altered at the messenger RNA level in colons from patients with IBD. Likewise, studies using genetically modified mouse strains have revealed that multiple NLR family members have the potential to dramatically modulate the immune response during IBD. Targeting NLR signaling represents a promising and novel therapeutic strategy. However, significant effort is necessary to translate the current understanding of NLR biology into effective therapies.
Collapse
|
28
|
Yang TC, Chen TF, Tsai JJ, Hu RM. NagZ is required for beta-lactamase expression and full pathogenicity in Xanthomonas campestris pv. campestris str. 17. Res Microbiol 2014; 165:612-9. [DOI: 10.1016/j.resmic.2014.08.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 08/21/2014] [Accepted: 08/22/2014] [Indexed: 01/17/2023]
|
29
|
Di Lorenzo F, Paciello I, Fazio LL, Albuquerque L, Sturiale L, da Costa MS, Lanzetta R, Parrilli M, Garozzo D, Bernardini ML, Silipo DA, Molinaro A. Thermophiles as potential source of novel endotoxin antagonists: the full structure and bioactivity of the lipo-oligosaccharide from Thermomonas hydrothermalis. Chembiochem 2014; 15:2146-55. [PMID: 25171597 DOI: 10.1002/cbic.201402233] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Indexed: 01/10/2023]
Abstract
Thermomonas hydrothermalis is a Gram-negative thermophilic bacterium that is able to live at 50 °C. This ability is attributed to chemical modifications, involving those to bacterial cell-wall components, such as proteins and (glyco)lipids. As the main component of the outer membrane of Gram-negative bacteria, lipopolysaccharides (LPSs) are exposed to the environment, thus they can undergo structural chemical changes to allow thermophilic bacteria to live at their optimal growth temperature. Furthermore, as one of the major target of the eukaryotic innate immune system, LPS elicits host immune response in a structure-dependent mode; thus the uncommon chemical features of thermophilic bacterial LPSs might exert a different biological action on the innate immune system-an antagonistic effect, as shown in studies of LPS structure-activity relationship in the ongoing research into antagonist LPS candidates. Here, we report the complete structural and biological activity analysis of the lipo-oligosaccharide isolated from Thermomonas hydrothermalis, achieved by a multidisciplinary approach (chemical analysis, NMR, MALDI MS and cellular immunology). We demonstrate a tricky and interesting structure combined with a very interesting effect on human innate immunity.
Collapse
Affiliation(s)
- Flaviana Di Lorenzo
- Dipartimento di Scienze Chimiche, Università di Napoli "Federico II", Complesso Universitario Monte S.Angelo, Via Cintia 4, 80126 Napoli (Italy)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Liu X, Grabherr HM, Willmann R, Kolb D, Brunner F, Bertsche U, Kühner D, Franz-Wachtel M, Amin B, Felix G, Ongena M, Nürnberger T, Gust AA. Host-induced bacterial cell wall decomposition mediates pattern-triggered immunity in Arabidopsis. eLife 2014; 3:e01990. [PMID: 24957336 PMCID: PMC4103680 DOI: 10.7554/elife.01990] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 06/20/2014] [Indexed: 12/22/2022] Open
Abstract
Peptidoglycans (PGNs) are immunogenic bacterial surface patterns that trigger immune activation in metazoans and plants. It is generally unknown how complex bacterial structures such as PGNs are perceived by plant pattern recognition receptors (PRRs) and whether host hydrolytic activities facilitate decomposition of bacterial matrices and generation of soluble PRR ligands. Here we show that Arabidopsis thaliana, upon bacterial infection or exposure to microbial patterns, produces a metazoan lysozyme-like hydrolase (lysozyme 1, LYS1). LYS1 activity releases soluble PGN fragments from insoluble bacterial cell walls and cleavage products are able to trigger responses typically associated with plant immunity. Importantly, LYS1 mutant genotypes exhibit super-susceptibility to bacterial infections similar to that observed on PGN receptor mutants. We propose that plants employ hydrolytic activities for the decomposition of complex bacterial structures, and that soluble pattern generation might aid PRR-mediated immune activation in cell layers adjacent to infection sites.
Collapse
Affiliation(s)
- Xiaokun Liu
- Department of Plant Biochemistry, Center for Plant Molecular Biology, University of Tübingen, Tübingen, Germany
| | - Heini M Grabherr
- Department of Plant Biochemistry, Center for Plant Molecular Biology, University of Tübingen, Tübingen, Germany
| | - Roland Willmann
- Department of Plant Biochemistry, Center for Plant Molecular Biology, University of Tübingen, Tübingen, Germany
| | - Dagmar Kolb
- Department of Plant Biochemistry, Center for Plant Molecular Biology, University of Tübingen, Tübingen, Germany
| | - Frédéric Brunner
- Department of Plant Biochemistry, Center for Plant Molecular Biology, University of Tübingen, Tübingen, Germany
| | - Ute Bertsche
- Department of Microbial Genetics, University of Tübingen, Tübingen, Germany
| | - Daniel Kühner
- Department of Microbial Genetics, University of Tübingen, Tübingen, Germany
| | | | - Bushra Amin
- Medical and Natural Sciences Research Centre, University of Tübingen, Tübingen, Germany
| | - Georg Felix
- Department of Plant Biochemistry, Center for Plant Molecular Biology, University of Tübingen, Tübingen, Germany
| | - Marc Ongena
- Wallon Centre for Industrial Biology, University of Liege-Gembloux Agro-Bio Tech, Gembloux, Belgium
| | - Thorsten Nürnberger
- Department of Plant Biochemistry, Center for Plant Molecular Biology, University of Tübingen, Tübingen, Germany
| | - Andrea A Gust
- Department of Plant Biochemistry, Center for Plant Molecular Biology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
31
|
Mutations in Ralstonia solanacearum loci involved in lipopolysaccharide biogenesis, phospholipid trafficking and peptidoglycan recycling render bacteriophage infection. Arch Microbiol 2014; 196:667-74. [DOI: 10.1007/s00203-014-1002-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 05/13/2014] [Accepted: 05/30/2014] [Indexed: 10/25/2022]
|
32
|
Dworkin J. The medium is the message: interspecies and interkingdom signaling by peptidoglycan and related bacterial glycans. Annu Rev Microbiol 2014; 68:137-54. [PMID: 24847956 DOI: 10.1146/annurev-micro-091213-112844] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Peptidoglycan serves as a key structure of the bacterial cell by determining cell shape and providing resistance to internal turgor pressure. However, in addition to these essential and well-studied functions, bacterial signaling by peptidoglycan fragments, or muropeptides, has been demonstrated by recent work. Actively growing bacteria release muropeptides as a consequence of cell wall remodeling during elongation and division. Therefore, the presence of muropeptide synthesis is indicative of growth-promoting conditions and may serve as a broadly conserved signal for nongrowing cells to reinitiate growth. In addition, muropeptides serve as signals between bacteria and eukaryotic organisms during both pathogenic and symbiotic interactions. The increasingly appreciated role of the microbiota in metazoan organisms suggests that muropeptide signaling likely has important implications for homeostatic mammalian physiology.
Collapse
Affiliation(s)
- Jonathan Dworkin
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032;
| |
Collapse
|
33
|
Atilano ML, Pereira PM, Vaz F, Catalão MJ, Reed P, Grilo IR, Sobral RG, Ligoxygakis P, Pinho MG, Filipe SR. Bacterial autolysins trim cell surface peptidoglycan to prevent detection by the Drosophila innate immune system. eLife 2014; 3:e02277. [PMID: 24692449 PMCID: PMC3971415 DOI: 10.7554/elife.02277] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Bacteria have to avoid recognition by the host immune system in order to establish a successful infection. Peptidoglycan, the principal constituent of virtually all bacterial surfaces, is a specific molecular signature recognized by dedicated host receptors, present in animals and plants, which trigger an immune response. Here we report that autolysins from Gram-positive pathogenic bacteria, enzymes capable of hydrolyzing peptidoglycan, have a major role in concealing this inflammatory molecule from Drosophila peptidoglycan recognition proteins (PGRPs). We show that autolysins trim the outermost peptidoglycan fragments and that in their absence bacterial virulence is impaired, as PGRPs can directly recognize leftover peptidoglycan extending beyond the external layers of bacterial proteins and polysaccharides. The activity of autolysins is not restricted to the producer cells but can also alter the surface of neighboring bacteria, facilitating the survival of the entire population in the infected host. DOI:http://dx.doi.org/10.7554/eLife.02277.001 While most bacteria are harmless, some can cause diseases as varied as food poisoning and meningitis, so our immune system has developed various ways of detecting and eliminating bacteria and other pathogens. Receptor proteins belonging to the immune system detect molecules that give away the presence of the bacteria and trigger an immune response targeted at the invading pathogen. Peptidoglycan is one telltale molecule that betrays the presence of bacteria. Peptidoglycan is found in the bacterial cell wall, and for many years it was assumed that the immune system detected stray fragments of peptidoglycan that were accidentally shed by the bacteria. However, it was later shown that the immune system could, under certain conditions, detect peptidoglycan when it is still part of the cell wall. This raised an interesting question: do bacteria use other methods to stop peptidoglycan being detected by the immune system? Now, Atilano, Pereira et al. have found that enzymes called autolysins can conceal bacteria from the receptor proteins that detect peptidoglycan. These enzymes are needed to break the bonds within the peptidoglycan present in the rigid bacterial cell wall to allow the bacteria to grow and divide. ‘Knocking out’ the genes for autolysins allowed the receptor proteins from the fruit fly, Drosophila, to bind to the bacteria; however, the mutant bacteria were able to evade the immune system after they had been treated with the purified enzymes. Atilano, Pereira et al. suggest that the autolysins trim the exposed ends of the peptidoglycan molecules on the surface of the cell wall, which could otherwise be detected by the host. The experiments also show that bacterial pathogens—including a strain of MRSA—with mutations that knock out autolysin activity trigger a stronger immune response in fruit flies, and are therefore less able to infect this host. Autolysins also help to conceal Streptococcus pneumoniae—a bacterial pathogen that is a common cause of pneumonia and infant deaths in developing countries—from detection by fruit flies. The findings of Atilano, Pereira et al. highlight how bacteria employ a number of ways to evade detection. If similar behavior is observed when bacteria infect humans, autolysins could represent a new drug target for the treatment of bacterial diseases. DOI:http://dx.doi.org/10.7554/eLife.02277.002
Collapse
Affiliation(s)
- Magda Luciana Atilano
- Laboratory of Bacterial Cell Surfaces and Pathogenesis, Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa (ITQB-UNL), Oeiras, Portugal.,Genes and Development Laboratory, Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Pedro Matos Pereira
- Laboratory of Bacterial Cell Biology, Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa (ITQB-UNL), Oeiras, Portugal
| | - Filipa Vaz
- Laboratory of Bacterial Cell Surfaces and Pathogenesis, Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa (ITQB-UNL), Oeiras, Portugal
| | - Maria João Catalão
- Laboratory of Bacterial Cell Surfaces and Pathogenesis, Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa (ITQB-UNL), Oeiras, Portugal
| | - Patricia Reed
- Laboratory of Bacterial Cell Biology, Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa (ITQB-UNL), Oeiras, Portugal
| | - Inês Ramos Grilo
- Laboratory of Molecular Genetics, Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa (ITQB-UNL), Oeiras, Portugal
| | - Rita Gonçalves Sobral
- Departamento de Ciências da Vida, Centro de Recursos Microbiologicos (CREM), Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal
| | - Petros Ligoxygakis
- Genes and Development Laboratory, Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Mariana Gomes Pinho
- Laboratory of Bacterial Cell Biology, Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa (ITQB-UNL), Oeiras, Portugal
| | - Sérgio Raposo Filipe
- Laboratory of Bacterial Cell Surfaces and Pathogenesis, Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa (ITQB-UNL), Oeiras, Portugal
| |
Collapse
|
34
|
Jiao Y, Hasegawa M, Inohara N. Emerging roles of immunostimulatory oral bacteria in periodontitis development. Trends Microbiol 2014; 22:157-63. [PMID: 24433922 DOI: 10.1016/j.tim.2013.12.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 12/06/2013] [Accepted: 12/13/2013] [Indexed: 12/19/2022]
Abstract
Periodontitis is a common dental disease which results in irreversible alveolar bone loss around teeth, and subsequent tooth loss. Previous studies have focused on bacteria that damage the host and the roles of commensals to facilitate their colonization. Although some immune responses targeting oral bacteria protect the host from alveolar bone loss, recent studies show that particular host defense responses to oral bacteria can induce alveolar bone loss. Host-damaging and immunostimulatory oral bacteria cooperatively induce bone loss by inducing gingival damage followed by immunostimulation. In mouse models of experimental periodontitis induced by either Porphyromonas gingivalis or ligature, γ-proteobacteria accumulate and stimulate host immune responses to induce host damage. Here we review the differential roles of individual bacterial groups in promoting bone loss through the induction of host damage and immunostimulation.
Collapse
Affiliation(s)
- Yizu Jiao
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Mizuho Hasegawa
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Naohiro Inohara
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
35
|
Intracellular Shigella remodels its LPS to dampen the innate immune recognition and evade inflammasome activation. Proc Natl Acad Sci U S A 2013; 110:E4345-54. [PMID: 24167293 DOI: 10.1073/pnas.1303641110] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
LPS is a potent bacterial effector triggering the activation of the innate immune system following binding with the complex CD14, myeloid differentiation protein 2, and Toll-like receptor 4. The LPS of the enteropathogen Shigella flexneri is a hexa-acylated isoform possessing an optimal inflammatory activity. Symptoms of shigellosis are produced by severe inflammation caused by the invasion process of Shigella in colonic and rectal mucosa. Here we addressed the question of the role played by the Shigella LPS in eliciting a dysregulated inflammatory response of the host. We unveil that (i) Shigella is able to modify the LPS composition, e.g., the lipid A and core domains, during proliferation within epithelial cells; (ii) the LPS of intracellular bacteria (iLPS) and that of bacteria grown in laboratory medium differ in the number of acyl chains in lipid A, with iLPS being the hypoacylated; (iii) the immunopotential of iLPS is dramatically lower than that of bacteria grown in laboratory medium; (iv) both LPS forms mainly signal through the Toll-like receptor 4/myeloid differentiation primary response gene 88 pathway; (v) iLPS down-regulates the inflammasome-mediated release of IL-1β in Shigella-infected macrophages; and (vi) iLPS exhibits a reduced capacity to prime polymorfonuclear cells for an oxidative burst. We propose a working model whereby the two forms of LPS might govern different steps of the invasive process of Shigella. In the first phases, the bacteria, decorated with hypoacylated LPS, are able to lower the immune system surveillance, whereas, in the late phases, shigellae harboring immunopotent LPS are fully recognized by the immune system, which can then successfully resolve the infection.
Collapse
|
36
|
Mavrogiorgos N, Mekasha S, Yang Y, Kelliher MA, Ingalls RR. Activation of NOD receptors by Neisseria gonorrhoeae modulates the innate immune response. Innate Immun 2013; 20:377-89. [PMID: 23884094 DOI: 10.1177/1753425913493453] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
NOD1 and NOD2 are members of the NOD-like receptor family of cytosolic pattern recognition receptors that recognize specific fragments of the bacterial cell wall component peptidoglycan. Neisseria species are unique amongst Gram-negative bacteria in that they turn over large amounts of peptidoglycan during growth. We examined the ability of NOD1 and NOD2 to recognize Neisseria gonorrhoeae, and determined the role of NOD-dependent signaling in regulating the immune response to gonococcal infection. Gonococci, as well as conditioned medium from mid-logarithmic phase grown bacteria, were capable of activating both human NOD1 and NOD2, as well as mouse NOD2, leading to the activation of the transcription factor NF-κB and polyubiquitination of the adaptor receptor-interacting serine-threonine kinase 2. We identified a number of cytokines and chemokines that were differentially expressed in wild type versus NOD2-deficient macrophages in response to gonococcal infection. Moreover, NOD2 signaling up-regulated complement pathway components and cytosolic nucleic acid sensors, suggesting a broad impact of NOD activation on innate immunity. Thus, NOD1 and NOD2 are important intracellular regulators of the immune response to infection with N. gonorrhoeae. Given the intracellular lifestyle of this pathogen, we believe these cytosolic receptors may provide a key innate immune defense mechanism for the host during gonococcal infection.
Collapse
Affiliation(s)
- Nikolaos Mavrogiorgos
- 1Section of Infectious Diseases, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA
| | | | | | | | | |
Collapse
|
37
|
Abstract
Neisseria meningitidis (meningococcus) is a symbiont of the human nasopharynx. On occasion, meningococci disseminate from the nasopharynx to cause invasive disease. Previous work showed that purified meningococcal peptidoglycan (PG) stimulates human Nod1, which leads to activation of NF-κB and production of inflammatory cytokines. No studies have determined if meningococci release PG or activate Nod1 during infection. The closely related pathogen Neisseria gonorrhoeae releases PG fragments during normal growth. These fragments induce inflammatory cytokine production and ciliated cell death in human fallopian tubes. We determined that meningococci also release PG fragments during growth, including fragments known to induce inflammation. We found that N. meningitidis recycles PG fragments via the selective permease AmpG and that meningococcal PG recycling is more efficient than gonococcal PG recycling. Comparison of PG fragment release from N. meningitidis and N. gonorrhoeae showed that meningococci release less of the proinflammatory PG monomers than gonococci and degrade PG to smaller fragments. The decreased release of PG monomers by N. meningitidis relative to N. gonorrhoeae is partly due to ampG, since replacement of gonococcal ampG with the meningococcal allele reduced PG monomer release. Released PG fragments in meningococcal supernatants induced significantly less Nod1-dependent NF-κB activity than released fragments in gonococcal supernatants and tended to induce less interleukin-8 (IL-8) secretion in primary human fallopian tube explants. These results support a model in which efficient PG recycling and extensive degradation of PG fragments lessen inflammatory responses and may be advantageous for maintaining meningococcal carriage in the nasopharynx.
Collapse
|
38
|
Chandramouli KH, Dash S, Zhang Y, Ravasi T, Qian PY. Proteomic and metabolomic profiles of marine Vibrio sp. 010 in response to an antifoulant challenge. BIOFOULING 2013; 29:789-802. [PMID: 23822634 DOI: 10.1080/08927014.2013.805209] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Vibrio spp. have the ability to form biofilms, which may contribute to the subsequent successful colonization by microfouling and macrofouling organisms. The effects of an antifouling compound, poly-ether B, on Vibrio sp. 010 were investigated using flow cytometry, proteomics, and metabolomics. A 2-D gel-based proteomic analysis was used to identify proteins responsive to poly-ether B treatment. The profiles of biofilm metabolites were analyzed by ultra-performance liquid chromatography-mass spectrometry. Poly-ether B caused a significant reduction in viability. The proteins affected by the treatment were related to nucleotide metabolism, the glyoxylate cycle, and stress responses. Metabolites such as tripeptides, fatty acids, and quorum-sensing molecules were regulated differentially. Down-regulation of proteins and metabolites potentially led to a loss in colonisation ability, thereby affecting the structure of the biofilm. These results suggest that the proteins and metabolites identified may serve as target molecules for potent antifouling compounds.
Collapse
Affiliation(s)
- Kondethimmanahalli H Chandramouli
- KAUST Global Collaborative Research, Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | | | | | | | | |
Collapse
|
39
|
Lorenzo FD, Sturiale L, Palmigiano A, Fazio LL, Paciello I, Coutinho CP, Sá-Correia I, Bernardini M, Lanzetta R, Garozzo D, Silipo A, Molinaro A. Chemistry and Biology of the Potent Endotoxin from aBurkholderia dolosaClinical Isolate from a Cystic Fibrosis Patient. Chembiochem 2013; 14:1105-15. [DOI: 10.1002/cbic.201300062] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Indexed: 01/10/2023]
|
40
|
Silipo A, Di Lorenzo F, Fazio LL, Paciello I, Sturiale L, Palmigiano A, Parrilli M, Grant WD, Garozzo D, Lanzetta R, Bernardini ML, Molinaro A. Structure and Immunological Activity of the Lipopolysaccharide Isolated from the SpeciesAlkalimonas delamerensis. European J Org Chem 2013. [DOI: 10.1002/ejoc.201201702] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
41
|
Mating pair formation homologue TraG is a variable membrane protein essential for contact-independent type IV secretion of chromosomal DNA by Neisseria gonorrhoeae. J Bacteriol 2013; 195:1666-79. [PMID: 23378511 DOI: 10.1128/jb.02098-12] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Neisseria gonorrhoeae uses a type IV secretion system (T4SS) to secrete chromosomal DNA into the surrounding milieu. The DNA is effective in transforming gonococci in the population, and this mechanism of DNA donation may contribute to the high degree of genetic diversity in this species. Similar to other F-like T4SSs, the gonococcal T4SS requires a putative membrane protein, TraG, for DNA transfer. In F-plasmid and related systems, the homologous protein acts in pilus production, mating pair stabilization, and entry exclusion. We characterized the localization, membrane topology, and variation of TraG in N. gonorrhoeae. TraG was found to be an inner-membrane protein with one large periplasmic region and one large cytoplasmic region. Each gonococcal strain carried one of three different alleles of traG. Strains that carried the smallest allele of traG were found to lack the peptidoglycanase gene atlA but carried a peptidoglycan endopeptidase gene in place of atlA. The purified endopeptidase degraded gonococcal peptidoglycan in vitro, cutting the peptide cross-links. Although the other two traG alleles functioned for DNA secretion in strain MS11, the smallest traG did not support DNA secretion. Despite the requirement for a mating pair stabilization homologue, static coculture transformation experiments demonstrated that DNA transfer was nuclease sensitive and required active uptake by the recipient, thus demonstrating that transfer occurred by transformation and not conjugation. Together, these results demonstrate the TraG acts in a process of DNA export not specific to conjugation and that different forms of TraG affect what substrates can be transported.
Collapse
|
42
|
Liu M, Haenssler E, Uehara T, Losick VP, Park JT, Isberg RR. The Legionella pneumophila EnhC protein interferes with immunostimulatory muramyl peptide production to evade innate immunity. Cell Host Microbe 2013; 12:166-76. [PMID: 22901537 PMCID: PMC3678716 DOI: 10.1016/j.chom.2012.06.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 04/27/2012] [Accepted: 06/04/2012] [Indexed: 12/22/2022]
Abstract
Successful pathogens have evolved to evade innate immune recognition of microbial molecules by pattern recognition receptors (PRR), which control microbial growth in host tissues. Upon Legionella pneumophila infection of macrophages, the cytosolic PRR Nod1 recognizes anhydro-disaccharide-tetrapeptide (anhDSTP) generated by soluble lytic transglycosylase (SltL), the predominant bacterial peptidoglycan degrading enzyme, to activate NF-κB-dependent innate immune responses. We show that L. pneumophila periplasmic protein EnhC, which is uniquely required for bacterial replication within macrophages, interferes with SltL to lower anhDSTP production. L. pneumophila mutant strains lacking EnhC (ΔenhC) increase Nod1-dependent NF-κB activation in host cells, while reducing SltL activity in a ΔenhC strain restores intracellular bacterial growth. Further, L. pneumophila ΔenhC is specifically rescued in Nod1- but not Nod2-deficient macrophages, arguing that EnhC facilitates evasion from Nod1 recognition. These results indicate that a bacterial pathogen regulates peptidoglycan degradation to control the production of PRR ligands and evade innate immune recognition.
Collapse
Affiliation(s)
- Mingyu Liu
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
Many Gram-negative and Gram-positive bacteria recycle a significant proportion of the peptidoglycan components of their cell walls during their growth and septation. In many--and quite possibly all--bacteria, the peptidoglycan fragments are recovered and recycled. Although cell-wall recycling is beneficial for the recovery of resources, it also serves as a mechanism to detect cell-wall-targeting antibiotics and to regulate resistance mechanisms. In several Gram-negative pathogens, anhydro-MurNAc-peptide cell-wall fragments regulate AmpC β-lactamase induction. In some Gram-positive organisms, short peptides derived from the cell wall regulate the induction of both β-lactamase and β-lactam-resistant penicillin-binding proteins. The involvement of peptidoglycan recycling with resistance regulation suggests that inhibitors of the enzymes involved in the recycling might synergize with cell-wall-targeted antibiotics. Indeed, such inhibitors improve the potency of β-lactams in vitro against inducible AmpC β-lactamase-producing bacteria. We describe the key steps of cell-wall remodeling and recycling, the regulation of resistance mechanisms by cell-wall recycling, and recent advances toward the discovery of cell-wall-recycling inhibitors.
Collapse
Affiliation(s)
- Jarrod W Johnson
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | | | | |
Collapse
|
44
|
Wyckoff TJ, Taylor JA, Salama NR. Beyond growth: novel functions for bacterial cell wall hydrolases. Trends Microbiol 2012; 20:540-7. [PMID: 22944244 DOI: 10.1016/j.tim.2012.08.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2012] [Revised: 08/03/2012] [Accepted: 08/09/2012] [Indexed: 12/18/2022]
Abstract
The peptidoglycan cell wall maintains turgor pressure and cell shape of most bacteria. Cell wall hydrolases are essential, together with synthases, for growth and daughter cell separation. Recent work in diverse organisms has uncovered new cell wall hydrolases that act autonomously or on neighboring cells to modulate invasion of prey cells, cell shape, innate immune detection, intercellular communication, and competitor lysis. The hydrolases involved in these processes catalyze the cleavage of bonds throughout the sugar and peptide moities of peptidoglycan. Phenotypes associated with these diverse hydrolases reveal new functions of the bacterial cell wall beyond growth and division.
Collapse
Affiliation(s)
- Timna J Wyckoff
- Division of Science and Mathematics, University of Minnesota, Morris, MN, USA
| | | | | |
Collapse
|
45
|
Jehl SP, Nogueira CV, Zhang X, Starnbach MN. IFNγ inhibits the cytosolic replication of Shigella flexneri via the cytoplasmic RNA sensor RIG-I. PLoS Pathog 2012; 8:e1002809. [PMID: 22912573 PMCID: PMC3415441 DOI: 10.1371/journal.ppat.1002809] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 06/05/2012] [Indexed: 12/02/2022] Open
Abstract
The activation of host cells by interferon gamma (IFNγ) is essential for inhibiting the intracellular replication of most microbial pathogens. Although significant advances have been made in identifying IFNγ-dependent host factors that suppress intracellular bacteria, little is known about how IFNγ enables cells to recognize, or restrict, the growth of pathogens that replicate in the host cytoplasm. The replication of the cytosolic bacterial pathogen Shigella flexneri is significantly inhibited in IFNγ-stimulated cells, however the specific mechanisms that mediate this inhibition have remained elusive. We found that S. flexneri efficiently invades IFNγ-activated mouse embryonic fibroblasts (MEFs) and escapes from the vacuole, suggesting that IFNγ acts by blocking S. flexneri replication in the cytosol. This restriction on cytosolic growth was dependent on interferon regulatory factor 1 (IRF1), an IFNγ-inducible transcription factor capable of inducing IFNγ-mediated cell-autonomous immunity. To identify host factors that restrict S. flexneri growth, we used whole genome microarrays to identify mammalian genes whose expression in S. flexneri-infected cells is controlled by IFNγ and IRF1. Among the genes we identified was the pattern recognition receptor (PRR) retanoic acid-inducible gene I (RIG-I), a cytoplasmic sensor of foreign RNA that had not been previously known to play a role in S. flexneri infection. We found that RIG-I and its downstream signaling adaptor mitochondrial antiviral signaling protein (MAVS)—but not cytosolic Nod-like receptors (NLRs)—are critically important for IFNγ-mediated S. flexneri growth restriction. The recently described RNA polymerase III pathway, which transcribes foreign cytosolic DNA into the RIG-I ligand 5′-triphosphate RNA, appeared to be involved in this restriction. The finding that RIG-I responds to S. flexneri infection during the IFNγ response extends the range of PRRs that are capable of recognizing this bacterium. Additionally, these findings expand our understanding of how IFNγ recognizes, and ultimately restricts, bacterial pathogens within host cells. Shigella flexneri, the major cause of bacillary dysentery worldwide, invades and replicates within the cytoplasm of intestinal epithelial cells, where it disseminates to neighboring cells and ultimately increases the likelihood of transmission to uninfected hosts. A hallmark of the mammalian immune system is its ability to inhibit the growth of such intracellular pathogens by upregulating intracellular resistance mechanisms in response to the cytokine IFNγ. We found that in non-myeloid host cells stimulated with IFNγ S. flexneri remains able to invade the cells efficiently and gain access to the host cytoplasm. Once in the cytoplasm of IFγ-activated cells, the RIG-I/ MAVS immunosurveillance pathway is activated, enabling the stimulated host cells to inhibit S. flexneri replication. Interestingly, RIG-I only played a minor role in the cellular response to this pathogen in the absence of IFNγ, suggesting that the IFNγ response ensures the recognition of the infection through an immunosurveillance pathway that is otherwise dispensable for controlling S. flexneri growth. Together, these findings implicate the RIG-I pathway as a crucial component in the cellular response to this devastating bacterial pathogen.
Collapse
Affiliation(s)
- Stephanie P. Jehl
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Catarina V. Nogueira
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Xuqing Zhang
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Michael N. Starnbach
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
46
|
Huang FC. Regulation of Salmonella flagellin-induced interleukin-8 in intestinal epithelial cells by muramyl dipeptide. Cell Immunol 2012; 278:1-9. [PMID: 23121969 DOI: 10.1016/j.cellimm.2012.06.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 05/24/2012] [Accepted: 06/19/2012] [Indexed: 12/31/2022]
Abstract
Toll-like receptor 5 (TLR5) and nucleotide-binding oligomerization domain 2 (Nod2) are two important pattern recognition receptors involved in innate immunity to invading pathogens. Flagellin, recognized by TLR5, is Salmonella's dominant pro-inflammatory determinant in intestinal epithelial cells (IECs). Nod2 has played a pivotal role in protecting against intestinal bacterial infection. Therefore the aim of the study is to investigate regulation of Salmonella flagellin-induced interleukin (IL)-8 (IL-8) in IECs by Nod2 agonist, muramyl dipeptide (MDP). We found that MDP by itself induced only a weak IL-8 secretion in Caco-2 cells. However, it did show synergistic enhancement on flagellin-induced IL-8 production in Caco-2 cells, possibly caused by flagellin-mediated enhanced Nod2 recruitment into cell membrane. By Western blot and siRNA, we showed ERK and NF-κB, Nod2 and Rip2 were involved in the synergistic effect of MDP. These findings suggested that the cooperation of TLR5 and Nod2 in IECs regulates inflammatory response to Salmonella infection.
Collapse
Affiliation(s)
- Fu-Chen Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| |
Collapse
|
47
|
Lautz K, Damm A, Menning M, Wenger J, Adam AC, Zigrino P, Kremmer E, Kufer TA. NLRP10 enhances Shigella-induced pro-inflammatory responses. Cell Microbiol 2012; 14:1568-83. [PMID: 22672233 DOI: 10.1111/j.1462-5822.2012.01822.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 05/18/2012] [Accepted: 05/26/2012] [Indexed: 01/01/2023]
Abstract
Members of the NLR family evolved as intracellular sensors for bacterial and viral infection. However, our knowledge on the implication of most of the human NLR proteins in innate immune responses still remains fragmentary. Here we characterized the role of human NLRP10 in bacterial infection. Our data revealed that NLRP10 is a cytoplasmic localized protein that positively contributes to innate immune responses induced by the invasive bacterial pathogen Shigella flexneri. SiRNA-mediated knock-down studies showed that NLRP10 contributes to pro-inflammatory cytokine release triggered by Shigella in epithelial cells and primary dermal fibroblasts, by influencing p38 and NF-κB activation. This effect is dependent on the ATPase activity of NLRP10 and its PYD domain. Mechanistically, NLRP10 interacts with NOD1, a NLR that is pivotally involved in sensing of invasive microbes, and both proteins are recruited to the bacterial entry point at the plasma membrane. Moreover, NLRP10 physically interacts with downstream components of the NOD1 signalling pathway, such as RIP2, TAK1 and NEMO. Taken together, our data revealed a novel role of NLRP10 in innate immune responses towards bacterial infection and suggest that NLRP10 functions as a scaffold for the formation of the NOD1-Nodosome.
Collapse
Affiliation(s)
- Katja Lautz
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Boudreau MA, Fisher JF, Mobashery S. Messenger functions of the bacterial cell wall-derived muropeptides. Biochemistry 2012; 51:2974-90. [PMID: 22409164 DOI: 10.1021/bi300174x] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Bacterial muropeptides are soluble peptidoglycan structures central to recycling of the bacterial cell wall and messengers in diverse cell signaling events. Bacteria sense muropeptides as signals that antibiotics targeting cell-wall biosynthesis are present, and eukaryotes detect muropeptides during the innate immune response to bacterial infection. This review summarizes the roles of bacterial muropeptides as messengers, with a special emphasis on bacterial muropeptide structures and the relationship of structure to the biochemical events that the muropeptides elicit. Muropeptide sensing and recycling in both Gram-positive and Gram-negative bacteria are discussed, followed by muropeptide sensing by eukaryotes as a crucial event in the innate immune response of insects (via peptidoglycan-recognition proteins) and mammals (through Nod-like receptors) to bacterial invasion.
Collapse
Affiliation(s)
- Marc A Boudreau
- Department of Chemistry and Biochemistry, Nieuwland Science Hall, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | | | | |
Collapse
|
49
|
Chan YA, Hackett KT, Dillard JP. The lytic transglycosylases of Neisseria gonorrhoeae. Microb Drug Resist 2012; 18:271-9. [PMID: 22432703 DOI: 10.1089/mdr.2012.0001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Neisseria gonorrhoeae encodes five lytic transglycosylases (LTs) in the core genome, and most gonococcal strains also carry the gonococcal genetic island that encodes one or two additional LTs. These peptidoglycan (PG)-degrading enzymes are required for a number of processes that are either involved in the normal growth of the bacteria or affect the pathogenesis and gene transfer aspects of this species that make N. gonorrhoeae highly inflammatory and highly genetically variable. Systematic mutagenesis determined that two LTs are involved in producing the 1,6-anhydro PG monomers that cause the death of ciliated cells in Fallopian tubes. Here, we review the information available on these enzymes and discuss their roles in bacterial growth, cell separation, autolysis, type IV secretion, and pathogenesis.
Collapse
Affiliation(s)
- Yolande A Chan
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin 53706, USA
| | | | | |
Collapse
|
50
|
Abstract
Much is known about the molecular effectors of pathogenicity of gram-negative enteric pathogens, among which Shigella can be considered a model. This is due to its capacity to recapitulate the multiple steps required for a pathogenic microbe to survive close to its mucosal target, colonize and then invade its epithelial surface, cause its inflammatory destruction and simultaneously regulate the extent of the elicited innate response to likely survive the encounter and achieve successful subsequent transmission. These various steps of the infectious process represent an array of successive environmental conditions to which the bacteria need to successfully adapt. These conditions represent the selective pressure that triggered the "arms race" in which Shigella acquired the genetic and molecular effectors of its pathogenic armory, including the regulatory hierarchies that regulate the expression and function of these effectors. They also represent cues through which Shigella achieves the temporo-spatial expression and regulation of its virulence effectors. The role of such environmental cues has recently become obvious in the case of the major virulence effector of Shigella, the type three secretion system (T3SS) and its dedicated secreted virulence effectors. It needs to be better defined for other major virulence components such as the LPS and peptidoglycan which are used as examples here, in addition to the T3SS as models of regulation as it relates to the assembly and functional regulation of complex macromolecular systems of the bacterial surface. This review also stresses the need to better define what the true and relevant environmental conditions can be at the various steps of the progression of infection. The "identity" of the pathogen differs depending whether it is cultivated under in vitro or in vivo conditions. Moreover, this "identity" may quickly change during its progression into the infected tissue. Novel concepts and relevant tools are needed to address this challenge in microbial pathogenesis.
Collapse
Affiliation(s)
- Benoit Marteyn
- Unité de Pathogénie Microbienne Moléculaire; Institut Pasteur; Paris, France,Unité INSERM 786; Institut Pasteur; Paris, France
| | - Anastasia Gazi
- Unité de Pathogénie Microbienne Moléculaire; Institut Pasteur; Paris, France,Unité INSERM 786; Institut Pasteur; Paris, France
| | - Philippe Sansonetti
- Unité de Pathogénie Microbienne Moléculaire; Institut Pasteur; Paris, France,Unité INSERM 786; Institut Pasteur; Paris, France,Chaire de Microbiologie et Maladies Infectieuses; Collège de France; Paris, France,Correspondence to: Philippe Sansonetti,
| |
Collapse
|