1
|
Kim JK, Sapkota A, Roh T, Jo EK. The intricate interactions between inflammasomes and bacterial pathogens: Roles, mechanisms, and therapeutic potentials. Pharmacol Ther 2025; 265:108756. [PMID: 39581503 DOI: 10.1016/j.pharmthera.2024.108756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/06/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Abstract
Inflammasomes are intracellular multiprotein complexes that consist of a sensor, an adaptor, and a caspase enzyme to cleave interleukin (IL)-1β and IL-18 into their mature forms. In addition, caspase-1 and -11 activation results in the cleavage of gasdermin D to form pores, thereby inducing pyroptosis. Activation of the inflammasome and pyroptosis promotes host defense against pathogens, whereas dysregulation of the inflammasome can result in various pathologies. Inflammasomes exhibit versatile microbial signal detection, directly or indirectly, through cellular processes, such as ion fluctuations, reactive oxygen species generation, and the disruption of intracellular organelle function; however, bacteria have adaptive strategies to manipulate the inflammasome by altering microbe-associated molecular patterns, intercepting innate pathways with secreted effectors, and attenuating inflammatory and cell death responses. In this review, we summarize recent advances in the diverse roles of the inflammasome during bacterial infections and discuss how bacteria exploit inflammasome pathways to establish infections or persistence. In addition, we highlight the therapeutic potential of harnessing bacterial immune subversion strategies against acute and chronic bacterial infections. A more comprehensive understanding of the significance of inflammasomes in immunity and their intricate roles in the battle between bacterial pathogens and hosts will lead to the development of innovative strategies to address emerging threats posed by the expansion of drug-resistant bacterial infections.
Collapse
Affiliation(s)
- Jin Kyung Kim
- Department of Microbiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Asmita Sapkota
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea; Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Taylor Roh
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea; Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea; Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
| |
Collapse
|
2
|
Resta SC, Guerra F, Talà A, Bucci C, Alifano P. Beyond Inflammation: Role of Pyroptosis Pathway Activation by Gram-Negative Bacteria and Their Outer Membrane Vesicles (OMVs) in the Interaction with the Host Cell. Cells 2024; 13:1758. [PMID: 39513865 PMCID: PMC11545737 DOI: 10.3390/cells13211758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Pyroptosis is a gasdermin-mediated pro-inflammatory programmed cell death that, during microbial infections, aims to restrict the spreading of bacteria. Nevertheless, excessive pyroptosis activation leads to inflammation levels that are detrimental to the host. Pathogen-associated molecular patterns (PAMPs) present in bacteria and outer membrane vesicles (OMVs) can trigger pyroptosis pathways in different cell types with different outcomes. Moreover, some pathogens have evolved virulence factors that directly interfere with pyroptosis pathways, like Yersinia pestis YopM and Shigella flexneri IpaH7.8. Other virulence factors, such as those of Neisseria meningitidis, Neisseria gonorrhoeae, Salmonella enterica, and Helicobacter pylori affect pyroptosis pathways indirectly with important differences between pathogenic and commensal species of the same family. These pathogens deserve special attention because of the increasing antimicrobial resistance of S. flexneri and N. gonorrhoeae, the high prevalence of S. enterica and H. pylori, and the life-threatening diseases caused by N. meningitidis and Y. pestis. While inflammation due to macrophage pyroptosis has been extensively addressed, the effects of activation of pyroptosis pathways on modulation of cell cytoskeleton and cell-cell junctions in epithelia and endothelia and on the bacterial crossing of epithelial and endothelial barriers have only been partly investigated. Another important point is the diverse consequences of pyroptosis pathways on calcium influx, like activation of calcium-dependent enzymes and mitochondria dysregulation. This review will discuss the pyroptotic pathways activated by Gram-negative bacteria and their OMVs, analyzing the differences between pathogens and commensal bacteria. Particular attention will also be paid to the experimental models adopted and the main results obtained in the different models. Finally, strategies adopted by pathogens to modulate these pathways will be discussed with a perspective on the use of pyroptosis inhibitors as adjuvants in the treatment of infections.
Collapse
Affiliation(s)
- Silvia Caterina Resta
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy; (S.C.R.); (F.G.); (A.T.)
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy; (S.C.R.); (F.G.); (A.T.)
| | - Adelfia Talà
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy; (S.C.R.); (F.G.); (A.T.)
| | - Cecilia Bucci
- Department of Experimental Medicine (DiMeS), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy;
| | - Pietro Alifano
- Department of Experimental Medicine (DiMeS), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy;
| |
Collapse
|
3
|
Rytter H, Roger K, Chhuon C, Ding X, Coureuil M, Jamet A, Henry T, Guerrera IC, Charbit A. Dual proteomics of infected macrophages reveal bacterial and host players involved in the Francisella intracellular life cycle and cell to cell dissemination by merocytophagy. Sci Rep 2024; 14:7797. [PMID: 38565565 PMCID: PMC10987565 DOI: 10.1038/s41598-024-58261-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/27/2024] [Indexed: 04/04/2024] Open
Abstract
Bacterial pathogens adapt and replicate within host cells, while host cells develop mechanisms to eliminate them. Using a dual proteomic approach, we characterized the intra-macrophage proteome of the facultative intracellular pathogen, Francisella novicida. More than 900 Francisella proteins were identified in infected macrophages after a 10-h infection. Biotin biosynthesis-related proteins were upregulated, emphasizing the role of biotin-associated genes in Francisella replication. Conversely, proteins encoded by the Francisella pathogenicity island (FPI) were downregulated, supporting the importance of the F. tularensis Type VI Secretion System for vacuole escape, not cytosolic replication. In the host cell, over 300 proteins showed differential expression among the 6200 identified during infection. The most upregulated host protein was cis-aconitate decarboxylase IRG1, known for itaconate production with antimicrobial properties in Francisella. Surprisingly, disrupting IRG1 expression did not impact Francisella's intracellular life cycle, suggesting redundancy with other immune proteins or inclusion in larger complexes. Over-representation analysis highlighted cell-cell contact and actin polymerization in macrophage deregulated proteins. Using flow cytometry and live cell imaging, we demonstrated that merocytophagy involves diverse cell-to-cell contacts and actin polymerization-dependent processes. These findings lay the groundwork for further exploration of merocytophagy and its molecular mechanisms in future research.Data are available via ProteomeXchange with identifier PXD035145.
Collapse
Affiliation(s)
- Héloïse Rytter
- Université Paris CitéINSERM UMR-S1151, CNRS UMR-S8253Institut Necker Enfants Malades, 156-160 rue de Vaugirard, 75015, Paris, France
- INSERM U1151-CNRS UMR 8253, Team 7: Pathogénie des Infections Systémiques, 75015, Paris, France
| | - Kevin Roger
- INSERM US24/CNRS UAR3633, Proteomic Platform Necker, UniversitéParis-Cité, Federative Research Structure Necker, Paris, France
| | - Cerina Chhuon
- INSERM US24/CNRS UAR3633, Proteomic Platform Necker, UniversitéParis-Cité, Federative Research Structure Necker, Paris, France
| | - Xiongqi Ding
- Université Paris CitéINSERM UMR-S1151, CNRS UMR-S8253Institut Necker Enfants Malades, 156-160 rue de Vaugirard, 75015, Paris, France
- INSERM U1151-CNRS UMR 8253, Team 7: Pathogénie des Infections Systémiques, 75015, Paris, France
| | - Mathieu Coureuil
- Université Paris CitéINSERM UMR-S1151, CNRS UMR-S8253Institut Necker Enfants Malades, 156-160 rue de Vaugirard, 75015, Paris, France
- INSERM U1151-CNRS UMR 8253, Team 7: Pathogénie des Infections Systémiques, 75015, Paris, France
| | - Anne Jamet
- Université Paris CitéINSERM UMR-S1151, CNRS UMR-S8253Institut Necker Enfants Malades, 156-160 rue de Vaugirard, 75015, Paris, France
- INSERM U1151-CNRS UMR 8253, Team 7: Pathogénie des Infections Systémiques, 75015, Paris, France
| | - Thomas Henry
- CIRI, Centre International de Recherche en Infectiologie, Université Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Ida Chiara Guerrera
- INSERM US24/CNRS UAR3633, Proteomic Platform Necker, UniversitéParis-Cité, Federative Research Structure Necker, Paris, France.
| | - Alain Charbit
- Université Paris CitéINSERM UMR-S1151, CNRS UMR-S8253Institut Necker Enfants Malades, 156-160 rue de Vaugirard, 75015, Paris, France.
- INSERM U1151-CNRS UMR 8253, Team 7: Pathogénie des Infections Systémiques, 75015, Paris, France.
| |
Collapse
|
4
|
Zhang J, Zhu Y, Chen S, Xu Z, Zhang B, Liu A, He Q, Zhan J. Activation of cannabinoid receptors 2 alleviates myocardial damage in cecal ligation and puncture-induced sepsis by inhibiting pyroptosis. Immunol Lett 2023; 264:17-24. [PMID: 37918639 DOI: 10.1016/j.imlet.2023.10.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/15/2023] [Accepted: 10/28/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND It has been reported that cannabinoid receptors 2 (CB2 receptors) play an important role in the pathophysiological process of sepsis, which may also be associated with the regulation of pyroptosis, an inflammatory programmed cell death. The present study aimed to investigate the protective effect of CB2 receptors on myocardial damage in a model of septic mice by inhibiting pyroptosis. METHODS The C57BL/6 mice underwent cecal ligation and puncture (CLP) to induce sepsis. All mice were randomly divided into the sham, CLP, or CLP+HU308 group. Blood and heart tissue samples were collected 12 h after surgery. Hematoxylin and eosin staining was used for analyzing histopathological results. Creatine kinase isoenzymes (CK-MB) and IL-1β were measured using ELISA, while lactate dehydrogenase (LDH) level was determined using photoelectric colorimetry. The expression levels of CB2 receptors and pyroptosis-associated proteins (NLRP3, caspase-1, and GSDMD) were measured using western blotting. The location and distribution of CB2 receptors and caspase-1 in myocardial tissues were assessed by immunofluorescence. TUNEL staining was used to quantify the number of dead cells in myocardial tissues. RESULTS The CLP procedure increased CB2 receptor expression in mice. CB2 receptors were located in myocardial macrophages. Activating CB2 receptors decreased the levels of myocardial damage mediator LDH, CK-MB, and inflammatory cytokine IL-1β. The results also showed that CLP increased the pyroptosis in myocardial tissues, while CB2 agonist HU308 inhibited pyroptosis by decreasing the level of NLRP3 and activating caspase-1 and GSDMD. CONCLUSIONS CB2 receptor activation has a protective effect on the myocardium of mice with sepsis by inhibiting pyroptosis.
Collapse
Affiliation(s)
- Jingjing Zhang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China
| | - Yali Zhu
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China
| | - Shuxian Chen
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China
| | - Zujin Xu
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China
| | - Bin Zhang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China
| | - Anpeng Liu
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China
| | - Qianwen He
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China.
| | - Jia Zhan
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China.
| |
Collapse
|
5
|
Speir M, Tye H, Gottschalk TA, Simpson DS, Djajawi TM, Deo P, Ambrose RL, Conos SA, Emery J, Abraham G, Pascoe A, Hughes SA, Weir A, Hawkins ED, Kong I, Herold MJ, Pearson JS, Lalaoui N, Naderer T, Vince JE, Lawlor KE. A1 is induced by pathogen ligands to limit myeloid cell death and NLRP3 inflammasome activation. EMBO Rep 2023; 24:e56865. [PMID: 37846472 PMCID: PMC10626451 DOI: 10.15252/embr.202356865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 09/09/2023] [Accepted: 09/21/2023] [Indexed: 10/18/2023] Open
Abstract
Programmed cell death pathways play an important role in innate immune responses to infection. Activation of intrinsic apoptosis promotes infected cell clearance; however, comparatively little is known about how this mode of cell death is regulated during infections and whether it can induce inflammation. Here, we identify that the pro-survival BCL-2 family member, A1, controls activation of the essential intrinsic apoptotic effectors BAX/BAK in macrophages and monocytes following bacterial lipopolysaccharide (LPS) sensing. We show that, due to its tight transcriptional and post-translational regulation, A1 acts as a molecular rheostat to regulate BAX/BAK-dependent apoptosis and the subsequent NLRP3 inflammasome-dependent and inflammasome-independent maturation of the inflammatory cytokine IL-1β. Furthermore, induction of A1 expression in inflammatory monocytes limits cell death modalities and IL-1β activation triggered by Neisseria gonorrhoeae-derived outer membrane vesicles (NOMVs). Consequently, A1-deficient mice exhibit heightened IL-1β production in response to NOMV injection. These findings reveal that bacteria can induce A1 expression to delay myeloid cell death and inflammatory responses, which has implications for the development of host-directed antimicrobial therapeutics.
Collapse
|
6
|
Kroken AR, Klein KA, Mitchell PS, Nieto V, Jedel EJ, Evans DJ, Fleiszig SMJ. Intracellular replication of Pseudomonas aeruginosa in epithelial cells requires suppression of the caspase-4 inflammasome. mSphere 2023; 8:e0035123. [PMID: 37589460 PMCID: PMC10597407 DOI: 10.1128/msphere.00351-23] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 06/30/2023] [Indexed: 08/18/2023] Open
Abstract
Pathogenesis of Pseudomonas aeruginosa infections can include bacterial survival inside epithelial cells. Previously, we showed that this involves multiple roles played by the type three secretion system (T3SS), and specifically the effector ExoS. This includes ExoS-dependent inhibition of a lytic host cell response that subsequently enables intracellular replication. Here, we studied the underlying cell death response to intracellular P. aeruginosa, comparing wild-type to T3SS mutants varying in capacity to induce cell death and that localize to different intracellular compartments. Results showed that corneal epithelial cell death induced by intracellular P. aeruginosa lacking the T3SS, which remains in vacuoles, correlated with the activation of nuclear factor-κB as measured by p65 relocalization and tumor necrosis factor alpha transcription and secretion. Deletion of caspase-4 through CRISPR-Cas9 mutagenesis delayed cell death caused by these intracellular T3SS mutants. Caspase-4 deletion also countered more rapid cell death caused by T3SS effector-null mutants still expressing the T3SS apparatus that traffic to the host cell cytoplasm, and in doing so rescued intracellular replication normally dependent on ExoS. While HeLa cells lacked a lytic death response to T3SS mutants, it was found to be enabled by interferon gamma treatment. Together, these results show that epithelial cells can activate the noncanonical inflammasome pathway to limit proliferation of intracellular P. aeruginosa, not fully dependent on bacterially driven vacuole escape. Since ExoS inhibits the lytic response, the data implicate targeting of caspase-4, an intracellular pattern recognition receptor, as another contributor to the role of ExoS in the intracellular lifestyle of P. aeruginosa. IMPORTANCE Pseudomonas aeruginosa can exhibit an intracellular lifestyle within epithelial cells in vivo and in vitro. The type three secretion system (T3SS) effector ExoS contributes via multiple mechanisms, including extending the life of invaded host cells. Here, we aimed to understand the underlying cell death inhibited by ExoS when P. aeruginosa is intracellular. Results showed that intracellular P. aeruginosa lacking T3SS effectors could elicit rapid cell lysis via the noncanonical inflammasome pathway. Caspase-4 contributed to cell lysis even when the intracellular bacteria lacked the entire T33S and were consequently unable to escape vacuoles, representing a naturally occurring subpopulation during wild-type infection. Together, the data show the caspase-4 inflammasome as an epithelial cell defense against intracellular P. aeruginosa, and implicate its targeting as another mechanism by which ExoS preserves the host cell replicative niche.
Collapse
Affiliation(s)
- Abby R. Kroken
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, USA
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, USA
| | - Keith A. Klein
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, USA
| | - Patrick S. Mitchell
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Vincent Nieto
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, USA
| | - Eric J. Jedel
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, USA
| | - David J. Evans
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, USA
- College of Pharmacy, Touro University California, Vallejo, California, USA
| | - Suzanne M. J. Fleiszig
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, USA
- Graduate Groups in Vision Sciences, Microbiology, and Infectious Diseases & Immunity, University of California, Berkeley, California, USA
| |
Collapse
|
7
|
Liao XJ, He TT, Liu LY, Jiang XL, Sun SS, Deng YH, Zhang LQ, Xie HX, Nie P. Unraveling and characterization of novel T3SS effectors in Edwardsiella piscicida. mSphere 2023; 8:e0034623. [PMID: 37642418 PMCID: PMC10597406 DOI: 10.1128/msphere.00346-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 07/03/2023] [Indexed: 08/31/2023] Open
Abstract
Type III secretion system (T3SS) facilitates survival and replication of Edwardsiella piscicida in vivo. Identifying novel T3SS effectors and elucidating their functions are critical in understanding the pathogenesis of E. piscicida. E. piscicida T3SS effector EseG and EseJ was highly secreted when T3SS gatekeeper-containing protein complex EsaB-EsaL-EsaM was disrupted by EsaB deficiency. Based on this observation, concentrated secretomes of ΔesaB strain and ΔesaBΔesaN strain were purified by loading them into SDS-PAGE gel for a short electrophoresis to remove impurities prior to the in-the gel digestion and mass spectrometry. Four reported T3SS effectors and two novel T3SS effector candidates EseQ (ETAE_2009) and Trx2 (ETAE_0559) were unraveled by quantitative comparison of the identified peptides. EseQ and Trx2 were revealed to be secreted and translocated in a T3SS-dependent manner through CyaA-based translocation assay and immunofluorescent staining, demonstrating that EseQ and Trx2 are the novel T3SS effectors of E. piscicida. Trx2 was found to suppress macrophage apoptosis as revealed by TUNEL staining and cleaved caspase-3 of infected J774A.1 monolayers. Moreover, Trx2 has been shown to inhibit the p65 phosphorylation and p65 translocation into the nucleus, thus blocking the NF-κB pathway. Furthermore, depletion of Trx2 slightly but significantly attenuates E. piscicida virulence in a fish infection model. Taken together, an efficient method was established in unraveling T3SS effectors in E. piscicida, and Trx2, one of the novel T3SS effectors identified in this study, was demonstrated to suppress apoptosis and block NF- κB pathway during E. piscicida infection. IMPORTANCE Edwardsiella piscicida is an intracellular bacterial pathogen that causes intestinal inflammation and hemorrhagic sepsis in fish and human. Virulence depends on the Edwardsiella type III secretion system (T3SS). Identifying the bacterial effector proteins secreted by T3SS and defining their role is key to understanding Edwardsiella pathogenesis. EsaB depletion disrupts the T3SS gatekeeper-containing protein complex, resulting in increased secretion of T3SS effectors EseG and EseJ. EseQ and Trx2 were shown to be the novel T3SS effectors of E. piscicida by a secretome comparison between ∆esaB strain and ∆esaB∆esaN strain (T3SS mutant), together with CyaA-based translocation assay. In addition, Trx2 has been shown to suppress macrophage apoptosis and block the NF-κB pathway. Together, this work expands the known repertoire of T3SS effectors and sheds light on the pathogenic mechanism of E. piscicida.
Collapse
Affiliation(s)
- Xiao Jian Liao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Tian Tian He
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Lu Yi Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- Fisheries Research Institute, Wuhan Academy of Agricultural Sciences, Wuhan, China
| | - Xiu Long Jiang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Shan Shan Sun
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yu Hang Deng
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Li Qiang Zhang
- Fisheries Research Institute, Wuhan Academy of Agricultural Sciences, Wuhan, China
| | - Hai Xia Xie
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Pin Nie
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, China
| |
Collapse
|
8
|
Kroken AR, Klein KA, Mitchell PS, Nieto V, Jedel EJ, Evans DJ, Fleiszig SMJ. Intracellular replication of Pseudomonas aeruginosa in epithelial cells requires suppression of the caspase-4 inflammasome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.13.528260. [PMID: 36824932 PMCID: PMC9948977 DOI: 10.1101/2023.02.13.528260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Pathogenesis of Pseudomonas aeruginosa infections can include bacterial survival inside epithelial cells. Previously, we showed this involves multiple roles played by the type three-secretion system (T3SS), and specifically the effector ExoS. This includes ExoS-dependent inhibition of a lytic host cell response that subsequently enables intracellular replication. Here, we studied the underlying cell death response to intracellular P. aeruginosa, comparing wild-type to T3SS mutants varying in capacity to induce cell death and that localize to different intracellular compartments. Results showed that corneal epithelial cell death induced by intracellular P. aeruginosa lacking the T3SS, which remains in vacuoles, correlated with activation of NF-κB as measured by p65 relocalization and TNFα transcription and secretion. Deletion of caspase-4 through CRISPR-Cas9 mutagenesis delayed cell death caused by these intracellular T3SS mutants. Caspase-4 deletion also countered more rapid cell death caused by T3SS effector-null mutants still expressing the TSSS apparatus that traffic to the host cell cytoplasm, and in doing so rescued intracellular replication normally dependent on ExoS. While HeLa cells lacked a lytic death response to T3SS mutants, it was found to be enabled by interferon gamma treatment. Together, these results show that epithelial cells can activate the noncanonical inflammasome pathway to limit proliferation of intracellular P. aeruginosa, not fully dependent on bacterially-driven vacuole escape. Since ExoS inhibits the lytic response, the data implicate targeting of caspase-4, an intracellular pattern recognition receptor, as another contributor to the role of ExoS in the intracellular lifestyle of P. aeruginosa.
Collapse
Affiliation(s)
- Abby R Kroken
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL USA
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
| | - Keith A Klein
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL USA
| | | | - Vincent Nieto
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
| | - Eric J Jedel
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
| | - David J Evans
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
- College of Pharmacy, Touro University California, Vallejo, CA USA
| | - Suzanne M J Fleiszig
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, CA USA
- Graduate Groups in Vision Sciences, Microbiology, and Infectious Diseases & Immunity, University of California, Berkeley, CA USA
| |
Collapse
|
9
|
Marzhoseyni Z, Mousavi MJ, Saffari M, Ghotloo S. Immune escape strategies of Pseudomonas aeruginosa to establish chronic infection. Cytokine 2023; 163:156135. [PMID: 36724716 DOI: 10.1016/j.cyto.2023.156135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 01/08/2023] [Accepted: 01/12/2023] [Indexed: 02/02/2023]
Abstract
The infection caused by P. aeruginosa still is dangerous throughout the world. This is partly due to its immune escape mechanisms considerably increasing the bacterial survival in the host. By escape from recognition by TLRs, interference with complement system activation, phagocytosis inhibition, production of ROS, inhibition of NET production, interference with the generation of cytokines, inflammasome inhibition, reduced antigen presentation, interference with cellular and humoral immunity, and induction of apoptotic cell death and MDSc, P. aeruginosa breaks down the barriers of the immune system and causes lethal infections in the host. Recognition of other immune escape mechanisms of P. aeruginosa may provide a basis for the future treatment of the infection. This manuscript may provide new insights and information for the development of new strategies to combat P. aeruginosa infection. In the present manuscript, the escape mechanisms of P. aeruginosa against immune response would be reviewed.
Collapse
Affiliation(s)
- Zeynab Marzhoseyni
- Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Javad Mousavi
- Department of Hematology, Faculty of Allied Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mahmood Saffari
- Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Somayeh Ghotloo
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
10
|
A truncated mutation of MucA in Pseudomonas aeruginosa from a bronchiectasis patient affects T3SS expression and inflammasome activation. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1740-1747. [PMID: 36604139 PMCID: PMC9828237 DOI: 10.3724/abbs.2022169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that causes chronic airway infection in bronchiectasis patients and is closely associated with poor prognosis. Strains isolated from chronically infected patients typically have a mucoid phenotype due to the overproduction of alginate. In this study, we isolate a P. aeruginosa strain from the sputum of a patient with bronchiectasis and find that a truncated mutation occurred in mucA, which is named mucA117. mucA117 causes the strain to transform into a mucoid phenotype, downregulates the expression of T3SS and inflammasome ligands such as fliC and allows it to avoid inflammasome activation. The truncated mutation of the MucA protein may help P. aeruginosa escape clearance by the immune system, enabling long-term colonization.
Collapse
|
11
|
Oh C, Li L, Verma A, Reuven AD, Miao EA, Bliska JB, Aachoui Y. Neutrophil inflammasomes sense the subcellular delivery route of translocated bacterial effectors and toxins. Cell Rep 2022; 41:111688. [PMID: 36417874 PMCID: PMC9827617 DOI: 10.1016/j.celrep.2022.111688] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 05/23/2022] [Accepted: 10/27/2022] [Indexed: 11/23/2022] Open
Abstract
In neutrophils, caspase-11 cleaves gasdermin D (GSDMD), causing pyroptosis to clear cytosol-invasive bacteria. In contrast, caspase-1 also cleaves GSDMD but seems to not cause pyroptosis. Here, we show that this pyroptosis-resistant caspase-1 activation is specifically programmed by the site of translocation of the detected microbial virulence factors. We find that pyrin and NLRC4 agonists do not trigger pyroptosis in neutrophils when they access the cytosol from endosomal compartment. In contrast, when the same ligands penetrate through the plasma membrane, they cause pyroptosis. Consistently, pyrin detects extracellular Yersinia pseudotuberculosis ΔyopM in neutrophils, driving caspase-1-GSDMD pyroptosis. This pyroptotic response drives PAD4-dependent H3 citrullination and results in extrusion of neutrophil extracellular traps (NETs). Our data indicate that caspase-1, GSDMD, or PAD4 deficiency renders mice more susceptible to Y. pseudotuberculosis ΔyopM infection. Therefore, neutrophils induce pyroptosis in response to caspase-1-activating inflammasomes triggered by extracellular bacterial pathogens, but after they phagocytose pathogens, they are programmed to forego pyroptosis.
Collapse
Affiliation(s)
- Changhoon Oh
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Lupeng Li
- Department of Immunology and Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ambika Verma
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Arianna D Reuven
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH 03768, USA
| | - Edward A Miao
- Department of Immunology and Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| | - James B Bliska
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH 03768, USA
| | - Youssef Aachoui
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
12
|
Zhang S, Liang Y, Yao J, Li DF, Wang LS. Role of Pyroptosis in Inflammatory Bowel Disease (IBD): From Gasdermins to DAMPs. Front Pharmacol 2022; 13:833588. [PMID: 35677444 PMCID: PMC9168461 DOI: 10.3389/fphar.2022.833588] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 04/22/2022] [Indexed: 12/11/2022] Open
Abstract
Pyroptosis is a pro-inflammatory cell death executed by gasdermin family proteins that involve the formation of pores on cells, recognition of danger signals, and release of pro-inflammatory cytokines IL-1β and IL-18. Pyroptosis modulates mucosal innate immunity and enteropathogenic bacterial infection. Similarly, the gasdermin family has been reported to be involved in the defense of the intestinal epithelium against bacterial infection and in the regulation of intestinal inflammation. Pyroptosis initiates damage signals that activate multiple pathways to cause inflammation, which may be a potential cause of chronic intestinal inflammation. In this review, we discuss the impact of pyroptosis on inflammatory bowel disease (IBD), with a focus on the executive proteins of pyroptosis (GSDMB, GADMD, and GSDME) and IBD-related endogenous damage-associated molecular patterns (DAMPs) produced by pyroptosis.
Collapse
Affiliation(s)
- Shuxia Zhang
- School of Medicine, Southern University of Science and Technology, Shenzhen People Hospital, Shenzhen, China
| | | | - Jun Yao
- Department of Gastroenterology, The Second Clinical Medicine College (Shenzhen People's Hospital), Jinan University, Shenzhen, China
| | - De-Feng Li
- Department of Gastroenterology, The Second Clinical Medicine College (Shenzhen People's Hospital), Jinan University, Shenzhen, China
| | - Li-Sheng Wang
- Department of Gastroenterology, The Second Clinical Medicine College (Shenzhen People's Hospital), Jinan University, Shenzhen, China
| |
Collapse
|
13
|
Xie X, Zhang W, Chen X, Wu D, Cao Y. Norfloxacin suppresses Leptospira-induced inflammation through inhibiting p65 and ERK phosphorylation and NLRP3 inflammasome activation. Microb Pathog 2021; 162:105315. [PMID: 34826552 DOI: 10.1016/j.micpath.2021.105315] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/22/2021] [Accepted: 11/22/2021] [Indexed: 01/15/2023]
Abstract
Leptospirosis is a worldwide re-emerging zoonosis caused by pathogenic Leptospira. Inflammatory storms induced by Leptospira are the reason to induce immunoparalysis and organ failures. Antibiotics are still the current mainstream treatment for leptospirosis. In addition to their antibacterial action, the immunomodulatory function of antibiotics has been paid more and more attention. In this study, the role of norfloxacin on Leptospira-induced inflammation was investigated. Treatment with norfloxacin down-regulated Leptospira-induced IL-1β and TNF-α both in vivo and vitro models. Further study showed that norfloxacin inhibited Leptospira-induced phosphorylation of p65 and ERK. Norfloxacin also inhibited the Leptospira-induced NLRP3 inflammasome activation with the increased level of Na/K-ATPase Pump β1 subunit and decreased level of Kcnk6. These results indicated that norfloxacin suppressed Leptospira-induced inflammation through inhibiting p65 and ERK phosphorylation and NLRP3 inflammasome activation. Norfloxacin may be a potential candidate for suppressing inflammatory storms caused by Leptospira.
Collapse
Affiliation(s)
- Xufeng Xie
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, 130062, People's Republic of China.
| | - Wenlong Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, 130062, People's Republic of China.
| | - Xi Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, 130062, People's Republic of China.
| | - Dianjun Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, 130062, People's Republic of China.
| | - Yongguo Cao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, 130062, People's Republic of China.
| |
Collapse
|
14
|
Sireci G, Badami GD, Di Liberto D, Blanda V, Grippi F, Di Paola L, Guercio A, de la Fuente J, Torina A. Recent Advances on the Innate Immune Response to Coxiella burnetii. Front Cell Infect Microbiol 2021; 11:754455. [PMID: 34796128 PMCID: PMC8593175 DOI: 10.3389/fcimb.2021.754455] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/12/2021] [Indexed: 12/14/2022] Open
Abstract
Coxiella burnetii is an obligate intracellular Gram-negative bacterium and the causative agent of a worldwide zoonosis known as Q fever. The pathogen invades monocytes and macrophages, replicating within acidic phagolysosomes and evading host defenses through different immune evasion strategies that are mainly associated with the structure of its lipopolysaccharide. The main transmission routes are aerosols and ingestion of fomites from infected animals. The innate immune system provides the first host defense against the microorganism, and it is crucial to direct the infection towards a self-limiting respiratory disease or the chronic form. This review reports the advances in understanding the mechanisms of innate immunity acting during C. burnetii infection and the strategies that pathogen put in place to infect the host cells and to modify the expression of specific host cell genes in order to subvert cellular processes. The mechanisms through which different cell types with different genetic backgrounds are differently susceptible to C. burnetii intracellular growth are discussed. The subsets of cytokines induced following C. burnetii infection as well as the pathogen influence on an inflammasome-mediated response are also described. Finally, we discuss the use of animal experimental systems for studying the innate immune response against C. burnetii and discovering novel methods for prevention and treatment of disease in humans and livestock.
Collapse
Affiliation(s)
- Guido Sireci
- Central Laboratory of Advanced Diagnostic and Biological Research (CLADIBIOR), Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University Hospital "Paolo Giaccone", Università degli studi di Palermo, Palermo, Italy
| | - Giusto Davide Badami
- Central Laboratory of Advanced Diagnostic and Biological Research (CLADIBIOR), Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University Hospital "Paolo Giaccone", Università degli studi di Palermo, Palermo, Italy
| | - Diana Di Liberto
- Central Laboratory of Advanced Diagnostic and Biological Research (CLADIBIOR), Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University Hospital "Paolo Giaccone", Università degli studi di Palermo, Palermo, Italy
| | - Valeria Blanda
- Istituto Zooprofilattico Sperimentale della Sicilia, Palermo, Italy
| | - Francesca Grippi
- Istituto Zooprofilattico Sperimentale della Sicilia, Palermo, Italy
| | - Laura Di Paola
- Istituto Zooprofilattico Sperimentale della Sicilia, Palermo, Italy
| | - Annalisa Guercio
- Istituto Zooprofilattico Sperimentale della Sicilia, Palermo, Italy
| | - José de la Fuente
- SaBio Health and Biotechnology, Instituto de Investigación en Recursos Cinegéticos, IREC -Spanish National Research Council CSIC - University of Castilla-La Mancha UCLM - Regional Government of Castilla-La Mancha JCCM, Ciudad Real, Spain.,Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, United States
| | | |
Collapse
|
15
|
Zhang B, Zheng F, Liu A, Li Z, Zheng F, Liu Q, Yang L, Chen K, Wang Y, Zhang Z, He Q, Zhan J. Activation of CB2 receptor inhibits pyroptosis and subsequently ameliorates cecal ligation and puncture-induced sepsis. Int Immunopharmacol 2021; 99:108038. [PMID: 34364304 DOI: 10.1016/j.intimp.2021.108038] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/12/2021] [Accepted: 07/29/2021] [Indexed: 01/12/2023]
Abstract
BACKGROUND Cannabinoid receptor 2 (CB2), whose activities are upregulated during sepsis, may be related to the regulation of inflammatory programmed cell death called pyroptosis. The aim of this study is to investigate the role of CB2 activation in attenuation of inflammation through inhibiting pyroptosis in cecal ligation puncture (CLP)-induced sepsis andlipopolysaccharide (LPS) + ATP-stimulated macrophages. METHODS C57BL/6 mice were subjected to CLP procedure and treated with CB2 agonist HU308 and CB2 antagonist AM630. Lung tissues were collected for analyses of lung W/D ratio, inflammatory factors levels, and pyroptosis-related protein expression. Murine bone-marrow-derived macrophages (BMDM) were treated with LPS and ATP to construct a septic model in vitro in the presence of HU308 and AM630 for assessment of cell injury, cytokine levels and pyroptosis-related protein expression accordingly. To verify the relationship between CB2 receptors and pyroptosis in the process of inflammatory response, BMDM were transduced with CB2 receptors knockdown lentiviral vectors in the presence of HU308 and AM630 for assessment of pyroptosis-related protein expression. RESULTS CB2 activation ameliorated the release of inflammatory mediators. The results showed that CLP-induced pyroptosis was elevated, and CB2 agonist HU308 treatment inhibited the pyroptosis activity through a decrease of the protein levels of NLRP3 as well as caspase-1 and GSDMD activation. Similar results were obtained in BMDM after LPS and ATP treatment. Treatment with CB2 knockdown lentiviral particles prevented the HU308-induced decreases in cell pyroptosis, demonstrating that endogenous CB2 receptors are required for the cannabinoid-induced cell protection. CONCLUSIONS CB2 receptors activation plays a protective role in sepsis through inhibition of pyroptosis. The effect of CB2 receptors against pyroptosis depends on the existence of endogenous CB2 receptors.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 430071, Wuhan, Hubei, People's Republic of China
| | - Feng Zheng
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 430071, Wuhan, Hubei, People's Republic of China
| | - Anpeng Liu
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 430071, Wuhan, Hubei, People's Republic of China
| | - Zhen Li
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 430071, Wuhan, Hubei, People's Republic of China
| | - Fei Zheng
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 430071, Wuhan, Hubei, People's Republic of China
| | - Qiangsheng Liu
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 430071, Wuhan, Hubei, People's Republic of China
| | - Liu Yang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 430071, Wuhan, Hubei, People's Republic of China
| | - Kai Chen
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 430071, Wuhan, Hubei, People's Republic of China
| | - Yanlin Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 430071, Wuhan, Hubei, People's Republic of China
| | - Zongze Zhang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 430071, Wuhan, Hubei, People's Republic of China
| | - Qianwen He
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 430071, Wuhan, Hubei, People's Republic of China.
| | - Jia Zhan
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 430071, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|
16
|
Yang G, Zhu T, Wang D, Liu Z, Zhang R, Han G, Tian X, Liu B, Han MY, Zhang Z. Revealing the signaling regulation of hydrogen peroxide to cell pyroptosis using a ratiometric fluorescent probe in living cells. Chem Commun (Camb) 2021; 57:6628-6631. [PMID: 34124718 DOI: 10.1039/d1cc02008k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
A ratiometric fluorescent probe with a large emission shift was developed for the accurate measurement of hydrogen peroxide (H2O2) in sophisticated pyroptosis signaling pathways. The results reported here demonstrate that H2O2, as a principal member of ROS, is a critical upstream signaling molecule in regulating pyroptosis.
Collapse
Affiliation(s)
- Guanqing Yang
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, School of Chemistry and Chemical Engineering and Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, China.
| | - Tong Zhu
- School of Life Science, Anhui University, Hefei 230601, China
| | - Dong Wang
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, School of Chemistry and Chemical Engineering and Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, China.
| | - Zhengjie Liu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, School of Chemistry and Chemical Engineering and Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, China.
| | - Ruilong Zhang
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, School of Chemistry and Chemical Engineering and Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, China. and Key Laboratory of Structure and Functional Regulation of Hybrid Materials (Anhui University), Ministry of Education, Hefei, Anhui 230601, China
| | - Guangmei Han
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, School of Chemistry and Chemical Engineering and Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, China.
| | - Xiaohe Tian
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and molecular imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Bianhua Liu
- Key Lab of Photovoltaic and Energy Conservation Materials, Institute of Solid State Physics, HFIPS, Chinese Academy of Sciences, Hefei 230031, China
| | - Ming-Yong Han
- Key Lab of Photovoltaic and Energy Conservation Materials, Institute of Solid State Physics, HFIPS, Chinese Academy of Sciences, Hefei 230031, China
| | - Zhongping Zhang
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, School of Chemistry and Chemical Engineering and Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, China. and Key Laboratory of Structure and Functional Regulation of Hybrid Materials (Anhui University), Ministry of Education, Hefei, Anhui 230601, China
| |
Collapse
|
17
|
Walch P, Selkrig J, Knodler LA, Rettel M, Stein F, Fernandez K, Viéitez C, Potel CM, Scholzen K, Geyer M, Rottner K, Steele-Mortimer O, Savitski MM, Holden DW, Typas A. Global mapping of Salmonella enterica-host protein-protein interactions during infection. Cell Host Microbe 2021; 29:1316-1332.e12. [PMID: 34237247 PMCID: PMC8561747 DOI: 10.1016/j.chom.2021.06.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 02/24/2021] [Accepted: 05/21/2021] [Indexed: 11/16/2022]
Abstract
Intracellular bacterial pathogens inject effector proteins to hijack host cellular processes and promote their survival and proliferation. To systematically map effector-host protein-protein interactions (PPIs) during infection, we generated a library of 32 Salmonella enterica serovar Typhimurium (STm) strains expressing chromosomally encoded affinity-tagged effectors and quantified PPIs in macrophages and epithelial cells. We identified 446 effector-host PPIs, 25 of which were previously described, and validated 13 by reciprocal co-immunoprecipitation. While effectors converged on the same host cellular processes, most had multiple targets, which often differed between cell types. We demonstrate that SseJ, SseL, and SifA modulate cholesterol accumulation at the Salmonella-containing vacuole (SCV) partially via the cholesterol transporter Niemann-Pick C1 protein. PipB recruits the organelle contact site protein PDZD8 to the SCV, and SteC promotes actin bundling by phosphorylating formin-like proteins. This study provides a method for probing host-pathogen PPIs during infection and a resource for interrogating STm effector mechanisms.
Collapse
Affiliation(s)
- Philipp Walch
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany; Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Joel Selkrig
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Leigh A Knodler
- Paul G. Allen School for Global Health, College of Veterinary Medicine, Washington State University, Pullman, USA; Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Mandy Rettel
- EMBL, Proteomics Core Facility, Heidelberg, Germany
| | - Frank Stein
- EMBL, Proteomics Core Facility, Heidelberg, Germany
| | - Keith Fernandez
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Cristina Viéitez
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany; EMBL European Bioinformatics Institute, (EMBL-EBI), Hinxton, UK
| | - Clément M Potel
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Karoline Scholzen
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Matthias Geyer
- Institute of Structural Biology, University of Bonn, Bonn, Germany
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, TU Braunschweig, Braunschweig, Germany; Molecular Cell Biology Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Olivia Steele-Mortimer
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Mikhail M Savitski
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany; EMBL, Proteomics Core Facility, Heidelberg, Germany
| | - David W Holden
- MRC Centre for Molecular Bacteriology and Infection, Imperial College, London, UK
| | - Athanasios Typas
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany.
| |
Collapse
|
18
|
Delaney MA, Hartigh AD, Carpentier SJ, Birkland TP, Knowles DP, Cookson BT, Frevert CW. Avoidance of the NLRP3 Inflammasome by the Stealth Pathogen, Coxiella burnetii. Vet Pathol 2021; 58:624-642. [PMID: 33357072 DOI: 10.1177/0300985820981369] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Coxiella burnetii, a highly adapted obligate intracellular bacterial pathogen and the cause of the zoonosis Q fever, is a reemerging public health threat. C. burnetii employs a Type IV secretion system (T4SS) to establish and maintain its intracellular niche and modulate host immune responses including the inhibition of apoptosis. Interactions between C. burnetii and caspase-1-mediated inflammasomes are not fully elucidated. This study confirms that C. burnetii does not activate caspase-1 during infection of mouse macrophages in vitro. C. burnetii-infected cells did not develop NLRP3 and ASC foci indicating its ability to avoid cytosolic detection. C. burnetii is unable to inhibit the pyroptosis and IL-1β secretion that is induced by potent inflammasome stimuli but rather enhances these caspase-1-mediated effects. We found that C. burnetii upregulates pro-IL-1β and robustly primes NLRP3 inflammasomes via TLR2 and MyD88 signaling. As for wildtype C. burnetii, T4SS-deficient mutants primed and potentiated NLRP3 inflammasomes. An in vivo model of pulmonary infection in C57BL/6 mice was developed. Mice deficient in NLRP3 or caspase-1 were like wildtype mice in the development and resolution of splenomegaly due to red pulp hyperplasia, and histologic lesions and macrophage kinetics, but had slightly higher pulmonary bacterial burdens at the greatest measured time point. Together these findings indicate that C. burnetii primes but avoids cytosolic detection by NLRP3 inflammasomes, which are not required for the clinical resistance of C57BL/6 mice. Determining mechanisms employed by C. burnetii to avoid cytosolic detection via NLRP3 inflammasomes will be beneficial to the development of preventative and interventional therapies for Q fever.
Collapse
Affiliation(s)
- Martha A Delaney
- Departments of Comparative Medicine and Pathology, and the Comparative Pathology Program, 7284University of Washington, Seattle, WA
- Current address: Martha A. Delaney, Zoological Pathology Program, University of Illinois, Brookfield, IL, USA
| | - Andreas den Hartigh
- Departments of Microbiology and Lab Medicine, 7284University of Washington, Seattle, WA
| | - Samuel J Carpentier
- Departments of Microbiology and Lab Medicine, 7284University of Washington, Seattle, WA
| | - Timothy P Birkland
- Departments of Comparative Medicine and Pathology, and the Comparative Pathology Program, 7284University of Washington, Seattle, WA
| | - Donald P Knowles
- Animal Disease Research Unit, Agricultural Research Service, United States Department of Agriculture, Pullman, WA
- Department of Veterinary Microbiology and Pathology, 6760Washington State University, Pullman, WA
| | - Brad T Cookson
- Departments of Microbiology and Lab Medicine, 7284University of Washington, Seattle, WA
| | - Charles W Frevert
- Departments of Comparative Medicine and Pathology, and the Comparative Pathology Program, 7284University of Washington, Seattle, WA
| |
Collapse
|
19
|
Riera Romo M. Cell death as part of innate immunity: Cause or consequence? Immunology 2021; 163:399-415. [PMID: 33682112 DOI: 10.1111/imm.13325] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/11/2021] [Accepted: 02/25/2021] [Indexed: 12/13/2022] Open
Abstract
Regulated or programmed cell death plays a critical role in the development and tissue organization and function. In addition, it is intrinsically connected with immunity and host defence. An increasing cellular and molecular findings cause a change in the concept of cell death, revealing an expanding network of regulated cell death modalities and their biochemical programmes. Likewise, recent evidences demonstrate the interconnection between cell death pathways and how they are involved in different immune mechanisms. This work provides an overview of the main cell death programmes and their implication in innate immunity not only as an immunogenic/inflammatory process, but also as an active defence strategy during immune response and at the same time as a regulatory mechanism.
Collapse
Affiliation(s)
- Mario Riera Romo
- Radiology Department, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
20
|
Havira MS, Ta A, Kumari P, Wang C, Russo AJ, Ruan J, Rathinam VA, Vanaja SK. Shiga toxin suppresses noncanonical inflammasome responses to cytosolic LPS. Sci Immunol 2020; 5:5/53/eabc0217. [PMID: 33246946 DOI: 10.1126/sciimmunol.abc0217] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 10/02/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022]
Abstract
Inflammatory caspase-dependent cytosolic lipopolysaccharide (LPS) sensing is a critical arm of host defense against bacteria. How pathogens overcome this pathway to establish infections is largely unknown. Enterohemorrhagic Escherichia coli (EHEC) is a clinically important human pathogen causing hemorrhagic colitis and hemolytic uremic syndrome. We found that a bacteriophage-encoded virulence factor of EHEC, Shiga toxin (Stx), suppresses caspase-11-mediated activation of the cytosolic LPS sensing pathway. Stx was essential and sufficient to inhibit pyroptosis and interleukin-1 (IL-1) responses elicited specifically by cytosolic LPS. The catalytic activity of Stx was necessary for suppression of inflammasome responses. Stx impairment of inflammasome responses to cytosolic LPS occurs at the level of gasdermin D activation. Stx also suppresses inflammasome responses in vivo after LPS challenge and bacterial infection. Overall, this study assigns a previously undescribed inflammasome-subversive function to a well-known bacterial toxin, Stx, and reveals a new phage protein-based pathogen blockade of cytosolic immune surveillance.
Collapse
Affiliation(s)
- Morena S Havira
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Atri Ta
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Puja Kumari
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Chengliang Wang
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Ashley J Russo
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Jianbin Ruan
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Vijay A Rathinam
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Sivapriya Kailasan Vanaja
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA.
| |
Collapse
|
21
|
Involvement of the Inflammasome and Th17 Cells in Skin Lesions of Human Cutaneous Leishmaniasis Caused by Leishmania ( Viannia) panamensis. Mediators Inflamm 2020; 2020:9278931. [PMID: 33192178 PMCID: PMC7641710 DOI: 10.1155/2020/9278931] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/24/2020] [Accepted: 09/19/2020] [Indexed: 01/07/2023] Open
Abstract
Localized cutaneous leishmaniasis (LCL) caused by Leishmania (Viannia) panamensis is an endemic disease in Panama. This condition causes ulcerated skin lesions characterized by a mixed Th1/Th2 immune response that is responsible for disease pathology. However, the maintenance of the in situ inflammatory process involves other elements, such as Th17 and inflammasome responses. Although these processes are associated with parasite elimination, their role in the increase in disease pathology cannot be discarded. Thus, the role in Leishmania infection is still unclear. In this sense, the present study aimed at characterizing the Th17 and inflammasome responses in the skin lesions of patients with LCL caused by L. (V.) panamensis to help elucidate the pathogenesis of this disease in Panama. Th17 and inflammasome responses were evaluated by immunohistochemistry (IHQ) in 46 skin biopsies from patients with LCL caused by L. (V.) panamensis. The Th17 immune response was assessed using CD3, CD4, RoRγt, IL-17, IL-6, IL-23, and TGF-β1 antibodies, and the inflammasome response was assessed by IL-1β, IL-18, and caspase-1 antibodies. The presence of the Th17 and inflammasome responses was evidenced by a positive reaction for all immunological markers in the skin lesions. An inverse correlation between the density of amastigotes and the density of RoRγt+, IL-17+, IL-1β+, and caspase-1+ cells was observed, but no correlation between Th17 and the inflammasome response with evolutionary disease pathology was reported. These data showed the participation of Th17 cells and the inflammasome in the inflammatory response of the skin lesions of LCL caused by L. (V.) panamensis infection. These results suggest a role in the control of tissue parasitism of IL-17 and the activation of the NLRP3 inflammasome dependent on IL-1β but cannot exclude their role in the development of disease pathology.
Collapse
|
22
|
Orning P, Lien E. Multiple roles of caspase-8 in cell death, inflammation, and innate immunity. J Leukoc Biol 2020; 109:121-141. [PMID: 32531842 DOI: 10.1002/jlb.3mr0420-305r] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/16/2020] [Accepted: 04/30/2020] [Indexed: 12/20/2022] Open
Abstract
Caspase-8 is an apical caspase involved in the programmed form of cell death called apoptosis that is critically important for mammalian development and immunity. Apoptosis was historically described as immunologically silent in contrast to other types of programmed cell death such as necroptosis or pyroptosis. Recent reports suggest considerable crosstalk between these different forms of cell death. It is becoming increasingly clear that caspase-8 has many non-apoptotic roles, participating in multiple processes including regulation of necroptosis (mediated by receptor-interacting serine/threonine kinases, RIPK1-RIPK3), inflammatory cytokine expression, inflammasome activation, and cleavage of IL-1β and gasdermin D, and protection against shock and microbial infection. In this review, we discuss the involvement of caspase-8 in cell death and inflammation and highlight its role in innate immune responses and in the relationship between different forms of cell death. Caspase-8 is one of the central components in this type of crosstalk.
Collapse
Affiliation(s)
- Pontus Orning
- UMass Medical School, Program in Innate Immunity, Division of Infectious Diseases and Immunology, Department of Medicine, Worcester, Massachusetts, USA.,Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Egil Lien
- UMass Medical School, Program in Innate Immunity, Division of Infectious Diseases and Immunology, Department of Medicine, Worcester, Massachusetts, USA.,Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
23
|
Li LH, Chen TL, Chiu HW, Hsu CH, Wang CC, Tai TT, Ju TC, Chen FH, Chernikov OV, Tsai WC, Hua KF. Critical Role for the NLRP3 Inflammasome in Mediating IL-1β Production in Shigella sonnei-Infected Macrophages. Front Immunol 2020; 11:1115. [PMID: 32582195 PMCID: PMC7283925 DOI: 10.3389/fimmu.2020.01115] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 05/07/2020] [Indexed: 12/24/2022] Open
Abstract
Shigella is one of the leading bacterial causes of diarrhea worldwide, affecting more than 165 million people annually. Among the serotypes of Shigella, Shigella sonnei is physiologically unique and endemic in human immunodeficiency virus-infected men who have sex with men. The NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome, a protein complex composed of NLRP3, apoptosis-associated speck-like protein, and caspase-1, recognizes, and responds to pathogen infection and diverse sterile host-derived or environmental danger signals to induce IL-1β and IL-18 production. Although the Shigella flexneri-mediated activation of the NLRP3 inflammasome has been reported, the effect of S. sonnei on NLRP3 inflammasome activation remains unclear. We found that S. sonnei induced IL-1β production through NLRP3-dependent pathways in lipopolysaccharide-primed macrophages. A mechanistic study revealed that S. sonnei induced IL-1β production through P2X7 receptor-mediated potassium efflux, reactive oxygen species generation, lysosomal acidification, and mitochondrial damage. In addition, the phagocytosis of viable S. sonnei was important for IL-1β production. Furthermore, we demonstrated that NLRP3 negatively regulated phagocytosis and the bactericidal activity of macrophages against S. sonnei. These findings provide mechanistic insight into the activation of the NLRP3 inflammasome by S. sonnei in macrophages.
Collapse
Affiliation(s)
- Lan-Hui Li
- Department of Laboratory Medicine, Linsen, Chinese Medicine and Kunming Branch, Taipei City Hospital, Taipei, Taiwan.,Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Tzu-Ling Chen
- Department of Laboratory Medicine, Linsen, Chinese Medicine and Kunming Branch, Taipei City Hospital, Taipei, Taiwan.,Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hsiao-Wen Chiu
- Department of Biotechnology and Animal Science, National Ilan University, Ilan, Taiwan
| | - Chung-Hua Hsu
- Department of Laboratory Medicine, Linsen, Chinese Medicine and Kunming Branch, Taipei City Hospital, Taipei, Taiwan.,Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chien-Chun Wang
- Infectious Disease Division, Linsen, Chinese Medicine and Kunming Branch, Taipei City Hospital, Taipei, Taiwan
| | - Tzu-Ting Tai
- Department of Biotechnology and Animal Science, National Ilan University, Ilan, Taiwan
| | - Tz-Chuen Ju
- Department of Animal Science and Biotechnology, Tunghai University, Taichung, Taiwan
| | - Fang-Hsin Chen
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Oleg V Chernikov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry FEB RAS, Vladivostok, Russia
| | - Wen-Chiuan Tsai
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Kuo-Feng Hua
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department of Biotechnology and Animal Science, National Ilan University, Ilan, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
24
|
Nigro G, Arena ET, Sachse M, Moya-Nilges M, Marteyn BS, Sansonetti PJ, Campbell-Valois FX. Mapping of Shigella flexneri's tissue distribution and type III secretion apparatus activity during infection of the large intestine of guinea pigs. Pathog Dis 2020; 77:5580288. [PMID: 31578543 PMCID: PMC6920510 DOI: 10.1093/femspd/ftz054] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022] Open
Abstract
Shigella spp. are bacterial pathogens that invade the human colonic mucosa using a type III secretion apparatus (T3SA), a proteinaceous device activated upon contact with host cells. Active T3SAs translocate proteins that carve the intracellular niche of Shigella spp. Nevertheless, the activation state of the T3SA has not been addressed in vivo. Here, we used a green fluorescent protein transcription-based secretion activity reporter (TSAR) to provide a spatio-temporal description of S. flexneri T3SAs activity in the colon of Guinea pigs. First, we observed that early mucus release is triggered in the vicinity of luminal bacteria with inactive T3SA. Subsequent mucosal invasion showed bacteria with active T3SA associated with the brush border, eventually penetrating into epithelial cells. From 2 to 8 h post-challenge, the infection foci expanded, and these intracellular bacteria displayed homogeneously high-secreting activity, while extracellular foci within the lamina propria featured bacteria with low secretion activity. We also found evidence that within lamina propria macrophages, bacteria reside in vacuoles instead of accessing the cytosol. Finally, bacteria were cleared from tissues between 8 and 24 h post-challenge, highlighting the hit-and-run colonization strategy of Shigella. This study demonstrates how genetically encoded reporters can contribute to deciphering pathogenesis in vivo.
Collapse
Affiliation(s)
- Giulia Nigro
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, INSERM U1202, 24-28 rue du Docteur-Roux, 75015 Paris, France
| | - Ellen T Arena
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, INSERM U1202, 24-28 rue du Docteur-Roux, 75015 Paris, France.,Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Laboratory for Optical and Computational Instrumentation, 271 Animal Sciences, 1675 Observatory Drive, Madison, WI 53706, USA
| | - Martin Sachse
- Ultrastructural Bioimaging unit, Institut Pasteur, 24-28 rue du Docteur-Roux, 75015 Paris, France
| | - Maryse Moya-Nilges
- Ultrastructural Bioimaging unit, Institut Pasteur, 24-28 rue du Docteur-Roux, 75015 Paris, France
| | - Benoit S Marteyn
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, INSERM U1202, 24-28 rue du Docteur-Roux, 75015 Paris, France.,Architecture et Réactivité de l'ARN, Université de Strasbourg, CNRS UPR9002, 2 Allée Konrad Roentgen, 67084 Strasbourg, France.,Unité Pathogenèse des Infections Vasculaires, Institut Pasteur, 24-28 rue du Docteur-Roux, 75015 Paris, France
| | - Philippe J Sansonetti
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, INSERM U1202, 24-28 rue du Docteur-Roux, 75015 Paris, France.,Chaire de Microbiologie et Maladies Infectieuses, Collège de France, 11 Place Marcelin Berthelot, 75231 Paris, France
| | - F-X Campbell-Valois
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, INSERM U1202, 24-28 rue du Docteur-Roux, 75015 Paris, France.,The Host-Microbe Interactions Laboratory, Department of Chemistry and Biomolecular Sciences, University of Ottawa, 150 Louis-Pasteur private, Ottawa, ON, K1N 6N5, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Rd, Ottawa, ON, K1N 6N5, Canada
| |
Collapse
|
25
|
Wang X, Chang CH, Jiang J, Liu X, Li J, Liu Q, Liao YP, Li L, Nel AE, Xia T. Mechanistic Differences in Cell Death Responses to Metal-Based Engineered Nanomaterials in Kupffer Cells and Hepatocytes. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2000528. [PMID: 32337854 PMCID: PMC7263057 DOI: 10.1002/smll.202000528] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/19/2020] [Accepted: 03/19/2020] [Indexed: 05/18/2023]
Abstract
The mononuclear phagocyte system in the liver is a frequent target for nanoparticles (NPs). A toxicological profiling of metal-based NPs is performed in Kupffer cell (KC) and hepatocyte cell lines. Sixteen NPs are provided by the Nanomaterial Health Implications Research Consortium of the National Institute of Environmental Health Sciences to study the toxicological effects in KUP5 (KC) and Hepa 1-6 cells. Five NPs (Ag, CuO, ZnO, SiO2 , and V2 O5 ) exhibit cytotoxicity in both cell types, while SiO2 and V2 O5 induce IL-1β production in KC. Ag, CuO, and ZnO induced caspase 3 generated apoptosis in both cell types is accompanied by ion shedding and generation of mitochondrial reactive oxygen species (ROS) in both cell types. However, the cell death response to SiO2 in KC differs by inducing pyroptosis as a result of potassium efflux, caspase 1 activation, NLRP3 inflammasome assembly, IL-1β release, and cleavage of gasdermin-D. This releases pore-performing peptide fragments responsible for pyroptotic cell swelling. Interestingly, although V2 O5 induces IL-1β release and delays caspase 1 activation by vanadium ion interference in membrane Na+ /K+ adenosine triphosphate (ATP)ase activity, the major cell death mechanism in KC (and Hepa 1-6) is caspase 3 mediated apoptosis. These findings improve the understanding of the mechanisms of metal-based engineered nanomaterial (ENM) toxicity in liver cells toward comprehensive safety evaluation.
Collapse
Affiliation(s)
- Xiang Wang
- Division of NanoMedicine, Department of Medicine; University of California, Los Angeles, CA 90095, United States, United States
- California NanoSystems Institute; University of California, Los Angeles, CA 90095, United States, United States
| | - Chong Hyun Chang
- California NanoSystems Institute; University of California, Los Angeles, CA 90095, United States, United States
| | - Jinhong Jiang
- California NanoSystems Institute; University of California, Los Angeles, CA 90095, United States, United States
| | - Xiangsheng Liu
- Division of NanoMedicine, Department of Medicine; University of California, Los Angeles, CA 90095, United States, United States
| | - Jiulong Li
- Division of NanoMedicine, Department of Medicine; University of California, Los Angeles, CA 90095, United States, United States
| | - Qi Liu
- Division of NanoMedicine, Department of Medicine; University of California, Los Angeles, CA 90095, United States, United States
| | - Yu-Pei Liao
- Division of NanoMedicine, Department of Medicine; University of California, Los Angeles, CA 90095, United States, United States
| | - Linjiang Li
- California NanoSystems Institute; University of California, Los Angeles, CA 90095, United States, United States
| | - André E. Nel
- Division of NanoMedicine, Department of Medicine; University of California, Los Angeles, CA 90095, United States, United States
- California NanoSystems Institute; University of California, Los Angeles, CA 90095, United States, United States
| | - Tian Xia
- Division of NanoMedicine, Department of Medicine; University of California, Los Angeles, CA 90095, United States, United States
- California NanoSystems Institute; University of California, Los Angeles, CA 90095, United States, United States
| |
Collapse
|
26
|
Al Mamun A, Wu Y, Jia C, Munir F, Sathy KJ, Sarker T, Monalisa I, Zhou K, Xiao J. Role of pyroptosis in liver diseases. Int Immunopharmacol 2020; 84:106489. [PMID: 32304992 DOI: 10.1016/j.intimp.2020.106489] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/03/2020] [Accepted: 04/05/2020] [Indexed: 12/17/2022]
Abstract
Pyroptosis is known as a novel form of pro-inflammatory cell death program, which is exceptional from other types of cell death programs. Particularly, pyroptosis is characterized by Gasdermin family-mediated pore formation and subsequently cellular lysis, also release of several pro-inflammatory intracellular cytokines. In terms of mechanism, there are two signaling pathways involved in pyroptosis, including caspase-1, and caspase-4/5/11 mediated pathways. However, pyroptosis plays important roles in immune defense mechanisms. Recent studies have demonstrated that pyroptosis plays significant roles in the development of liver diseases. In our review, we have focused on the role of pyroptosis based on the molecular and pathophysiological mechanisms in the development of liver diseases. We have also highlighted targeting of pyroptosis for the therapeutic implications in liver diseases in the near future.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Yanqing Wu
- Institute of Life Sciences, Wenzhou University, Wenzhou 325035, Zhejiang Province, China
| | - Chang Jia
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Fahad Munir
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Kasfia Jahan Sathy
- Department of Pharmacy, North South University, Bashundhara, Dhaka 1229, Bangladesh
| | - Tamanna Sarker
- Department of Pharmacy, Southeast University, Banani, Dhaka 1213, Bangladesh
| | - Ilma Monalisa
- Department of Pharmacy, Southeast University, Banani, Dhaka 1213, Bangladesh
| | - Kailiang Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Jian Xiao
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China.
| |
Collapse
|
27
|
Majer A, McGreevy A, Booth TF. Molecular Pathogenicity of Enteroviruses Causing Neurological Disease. Front Microbiol 2020; 11:540. [PMID: 32328043 PMCID: PMC7161091 DOI: 10.3389/fmicb.2020.00540] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 03/12/2020] [Indexed: 12/12/2022] Open
Abstract
Enteroviruses are single-stranded positive-sense RNA viruses that primarily cause self-limiting gastrointestinal or respiratory illness. In some cases, these viruses can invade the central nervous system, causing life-threatening neurological diseases including encephalitis, meningitis and acute flaccid paralysis (AFP). As we near the global eradication of poliovirus, formerly the major cause of AFP, the number of AFP cases have not diminished implying a non-poliovirus etiology. As the number of enteroviruses linked with neurological disease is expanding, of which many had previously little clinical significance, these viruses are becoming increasingly important to public health. Our current understanding of these non-polio enteroviruses is limited, especially with regards to their neurovirulence. Elucidating the molecular pathogenesis of these viruses is paramount for the development of effective therapeutic strategies. This review summarizes the clinical diseases associated with neurotropic enteroviruses and discusses recent advances in the understanding of viral invasion of the central nervous system, cell tropism and molecular pathogenesis as it correlates with host responses.
Collapse
Affiliation(s)
- Anna Majer
- Viral Diseases Division, National Microbiology Laboratory, Winnipeg, MB, Canada
| | - Alan McGreevy
- Viral Diseases Division, National Microbiology Laboratory, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.,Department of Biology, University of Winnipeg, Winnipeg, MB, Canada
| | - Timothy F Booth
- Viral Diseases Division, National Microbiology Laboratory, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
28
|
Ebersole JL, Al-Sabbagh M, Dawson DR. Heterogeneity of human serum antibody responses to P. gingivalis in periodontitis: Effects of age, race/ethnicity, and sex. Immunol Lett 2020; 218:11-21. [PMID: 31863783 PMCID: PMC6956649 DOI: 10.1016/j.imlet.2019.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 12/04/2019] [Accepted: 12/17/2019] [Indexed: 02/06/2023]
Abstract
Aging humans display an increased prevalence and severity of periodontitis, although the mechanisms underlying these findings remain poorly understood. This report examined antigenic diversity of P. gingivalis related to disease presence and patient demographics. Serum IgG antibody to P. gingivalis strains ATCC33277, FDC381, W50 (ATCC53978), W83, A7A1-28 (ATCC53977) and A7436 was measured in 426 participants [periodontally healthy (n = 61), gingivitis (N = 66) or various levels of periodontitis (N = 299)]. We hypothesized that antigenic diversity in P. gingivalis could contribute to a lack of "immunity" in the chronic infections of periodontal disease. Across the strains, the antibody levels in the oldest age group were lower than in the youngest groups, and severe periodontitis patients did not show higher antibody with aging. While 80 % of the periodontitis patients in any age group showed an elevated response to at least one of the P. gingivalis strains, the patterns of individual responses in the older group were also substantially different than the other age groups. Significantly greater numbers of older patients showed strain-specific antibody profiles to only 1 strain. The findings support that P. gingivalis may demonstrate antigenic diversity/drift within patients and could be one factor to help explain the inefficiency/ineffectiveness of the adaptive immune response in managing the infection.
Collapse
Affiliation(s)
- J L Ebersole
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada Las Vegas and Department of Periodontology, College of Dentistry, University of Kentucky, United States.
| | - M Al-Sabbagh
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada Las Vegas and Department of Periodontology, College of Dentistry, University of Kentucky, United States
| | - D R Dawson
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada Las Vegas and Department of Periodontology, College of Dentistry, University of Kentucky, United States
| |
Collapse
|
29
|
The Distinct Immune-Stimulatory Capacities of Porphyromonas gingivalis Strains 381 and ATCC 33277 Are Determined by the fimB Allele and Gingipain Activity. Infect Immun 2019; 87:IAI.00319-19. [PMID: 31570556 DOI: 10.1128/iai.00319-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 09/19/2019] [Indexed: 11/20/2022] Open
Abstract
The Porphyromonas gingivalis strain ATCC 33277 (33277) and 381 genomes are nearly identical. However, strain 33277 displays a significantly diminished capacity to stimulate host cell Toll-like receptor 2 (TLR2)-dependent signaling and interleukin-1β (IL-1β) production relative to 381, suggesting that there are strain-specific differences in one or more bacterial immune-modulatory factors. Genomic sequencing identified a single nucleotide polymorphism in the 33277 fimB allele (A→T), creating a premature stop codon in the 33277 fimB open reading frame relative to the 381 fimB allele. Gene exchange experiments established that the 33277 fimB allele reduces the immune-stimulatory capacity of this strain. Transcriptome comparisons revealed that multiple genes related to carboxy-terminal domain (CTD) family proteins, including the gingipains, were upregulated in 33277 relative to 381. A gingipain substrate degradation assay demonstrated that cell surface gingipain activity is higher in 33277, and an isogenic mutant strain deficient for the gingipains exhibited an increased ability to induce TLR2 signaling and IL-1β production. Furthermore, 33277 and 381 mutant strains lacking CTD cell surface proteins were more immune-stimulatory than the parental wild-type strains, consistent with an immune-suppressive role for the gingipains. Our data show that the combination of an intact fimB allele and limited cell surface gingipain activity in P. gingivalis 381 renders this strain more immune-stimulatory. Conversely, a defective fimB allele and high-level cell surface gingipain activity reduce the capacity of P. gingivalis 33277 to stimulate host cell innate immune responses. In summary, genomic and transcriptomic comparisons identified key virulence characteristics that confer divergent host cell innate immune responses to these highly related P. gingivalis strains.
Collapse
|
30
|
Zhou CB, Fang JY. The role of pyroptosis in gastrointestinal cancer and immune responses to intestinal microbial infection. Biochim Biophys Acta Rev Cancer 2019; 1872:1-10. [DOI: 10.1016/j.bbcan.2019.05.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 04/18/2019] [Accepted: 05/01/2019] [Indexed: 01/04/2023]
|
31
|
Owino CO, Chu JJH. Recent advances on the role of host factors during non-poliovirus enteroviral infections. J Biomed Sci 2019; 26:47. [PMID: 31215493 PMCID: PMC6582496 DOI: 10.1186/s12929-019-0540-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 06/11/2019] [Indexed: 02/06/2023] Open
Abstract
Non-polio enteroviruses are emerging viruses known to cause outbreaks of polio-like infections in different parts of the world with several cases already reported in Asia Pacific, Europe and in United States of America. These outbreaks normally result in overstretching of health facilities as well as death in children under the age of five. Most of these infections are usually self-limiting except for the neurological complications associated with human enterovirus A 71 (EV-A71). The infection dynamics of these viruses have not been fully understood, with most inferences made from previous studies conducted with poliovirus.Non-poliovirus enteroviral infections are responsible for major outbreaks of hand, foot and mouth disease (HFMD) often associated with neurological complications and severe respiratory diseases. The myriad of disease presentations observed so far in children calls for an urgent need to fully elucidate the replication processes of these viruses. There are concerted efforts from different research groups to fully map out the role of human host factors in the replication cycle of these viral infections. Understanding the interaction between viral proteins and human host factors will unravel important insights on the lifecycle of this groups of viruses.This review provides the latest update on the interplay between human host factors/processes and non-polio enteroviruses (NPEV). We focus on the interactions involved in viral attachment, entry, internalization, uncoating, replication, virion assembly and eventual egress of the NPEV from the infected cells. We emphasize on the virus- human host interplay and highlight existing knowledge gaps that needs further studies. Understanding the NPEV-human host factors interactions will be key in the design and development of vaccines as well as antivirals against enteroviral infections. Dissecting the role of human host factors during NPEV infection cycle will provide a clear picture of how NPEVs usurp the human cellular processes to establish an efficient infection. This will be a boost to the drug and vaccine development against enteroviruses which will be key in control and eventual elimination of the viral infections.
Collapse
Affiliation(s)
- Collins Oduor Owino
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
| | - Justin Jang Hann Chu
- Department of Microbiology and Immunology, National University of Singapore, Singapore, 117597, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore.
| |
Collapse
|
32
|
Capmany A, Gambarte Tudela J, Alonso Bivou M, Damiani MT. Akt/AS160 Signaling Pathway Inhibition Impairs Infection by Decreasing Rab14-Controlled Sphingolipids Delivery to Chlamydial Inclusions. Front Microbiol 2019; 10:666. [PMID: 31001235 PMCID: PMC6456686 DOI: 10.3389/fmicb.2019.00666] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 03/18/2019] [Indexed: 12/26/2022] Open
Abstract
Chlamydia trachomatis, an obligate intracellular bacterium, intercepts different trafficking pathways of the host cell to acquire essential lipids for its survival and replication, particularly from the Golgi apparatus via a Rab14-mediated transport. Molecular mechanisms underlying how these bacteria manipulate intracellular transport are a matter of intense study. Here, we show that C. trachomatis utilizes Akt/AS160 signaling pathway to promote sphingolipids delivery to the chlamydial inclusion through Rab14-controlled vesicular transport. C. trachomatis provokes Akt phosphorylation along its entire developmental life cycle and recruits phosphorylated Akt (pAkt) to the inclusion membrane. As a consequence, Akt Substrate of 160 kDa (AS160), also known as TBC1D4, a GTPase Activating Protein (GAP) for Rab14, is phosphorylated and therefore inactivated. Phosphorylated AS160 (pAS160) loses its ability to promote GTP hydrolysis, favoring Rab14 binding to GTP. Akt inhibition by an allosteric isoform-specific Akt inhibitor (iAkt) prevents AS160 phosphorylation and reduces Rab14 recruitment to chlamydial inclusions. iAkt further impairs sphingolipids acquisition by C. trachomatis-inclusion and provokes lipid retention at the Golgi apparatus. Consequently, treatment with iAkt decreases chlamydial inclusion size, bacterial multiplication, and infectivity in a dose-dependent manner. Similar results were found in AS160-depleted cells. By electron microscopy, we observed that iAkt generates abnormal bacterial forms as those reported after sphingolipids deprivation or Rab14 silencing. Taken together, our findings indicate that targeting the Akt/AS160/Rab14 axis could constitute a novel strategy to limit chlamydial infections, mainly for those caused by antibiotic-resistant bacteria.
Collapse
Affiliation(s)
- Anahí Capmany
- Laboratorio de Bioquímica e Inmunidad, Área de Química Biológica, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas, Mendoza, Argentina
| | - Julián Gambarte Tudela
- Laboratorio de Bioquímica e Inmunidad, Área de Química Biológica, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.,Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas, Mendoza, Argentina
| | - Mariano Alonso Bivou
- Laboratorio de Bioquímica e Inmunidad, Área de Química Biológica, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.,Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas, Mendoza, Argentina
| | - María T Damiani
- Laboratorio de Bioquímica e Inmunidad, Área de Química Biológica, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.,Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas, Mendoza, Argentina
| |
Collapse
|
33
|
Apoptosis of intestinal epithelial cells restricts Clostridium difficile infection in a model of pseudomembranous colitis. Nat Commun 2018; 9:4846. [PMID: 30451870 PMCID: PMC6242954 DOI: 10.1038/s41467-018-07386-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 10/26/2018] [Indexed: 12/11/2022] Open
Abstract
Clostridium difficile is the leading cause of pseudomembranous colitis in hospitalized patients. C. difficile enterotoxins TcdA and TcdB promote this inflammatory condition via a cytotoxic response on intestinal epithelial cells (IECs), but the underlying mechanisms are incompletely understood. Additionally, TcdA and TcdB engage the Pyrin inflammasome in macrophages, but whether Pyrin modulates CDI pathophysiology is unknown. Here we show that the Pyrin inflammasome is not functional in IECs and that Pyrin signaling is dispensable for CDI-associated IEC death and for in vivo pathogenesis. Instead, our studies establish that C. difficile enterotoxins induce activation of executioner caspases 3/7 via the intrinsic apoptosis pathway, and demonstrate that caspase-3/7-mediated IEC apoptosis is critical for in vivo host defense during early stages of CDI. In conclusion, our findings dismiss a critical role for inflammasomes in CDI pathogenesis, and identify IEC apoptosis as a host defense mechanism that restricts C. difficile infection in vivo.
Collapse
|
34
|
Coxiella burnetii Blocks Intracellular Interleukin-17 Signaling in Macrophages. Infect Immun 2018; 86:IAI.00532-18. [PMID: 30061378 DOI: 10.1128/iai.00532-18] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 07/13/2018] [Indexed: 12/15/2022] Open
Abstract
Coxiella burnetii is an obligate intracellular bacterium and the etiological agent of Q fever. Successful host cell infection requires the Coxiella type IVB secretion system (T4BSS), which translocates bacterial effector proteins across the vacuole membrane into the host cytoplasm, where they manipulate a variety of cell processes. To identify host cell targets of Coxiella T4BSS effector proteins, we determined the transcriptome of murine alveolar macrophages infected with a Coxiella T4BSS effector mutant. We identified a set of inflammatory genes that are significantly upregulated in T4BSS mutant-infected cells compared to mock-infected cells or cells infected with wild-type (WT) bacteria, suggesting that Coxiella T4BSS effector proteins downregulate the expression of these genes. In addition, the interleukin-17 (IL-17) signaling pathway was identified as one of the top pathways affected by the bacteria. While previous studies demonstrated that IL-17 plays a protective role against several pathogens, the role of IL-17 during Coxiella infection is unknown. We found that IL-17 kills intracellular Coxiella in a dose-dependent manner, with the T4BSS mutant exhibiting significantly more sensitivity to IL-17 than WT bacteria. In addition, quantitative PCR confirmed the increased expression of IL-17 downstream signaling genes in T4BSS mutant-infected cells compared to WT- or mock-infected cells, including the proinflammatory cytokine genes Il1a, Il1b, and Tnfa, the chemokine genes Cxcl2 and Ccl5, and the antimicrobial protein gene Lcn2 We further confirmed that the Coxiella T4BSS downregulates macrophage CXCL2/macrophage inflammatory protein 2 and CCL5/RANTES protein levels following IL-17 stimulation. Together, these data suggest that Coxiella downregulates IL-17 signaling in a T4BSS-dependent manner in order to escape the macrophage immune response.
Collapse
|
35
|
AIM2 Engages Active but Unprocessed Caspase-1 to Induce Noncanonical Activation of the NLRP3 Inflammasome. Cell Rep 2018; 20:794-805. [PMID: 28746866 DOI: 10.1016/j.celrep.2017.06.086] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 05/10/2017] [Accepted: 06/28/2017] [Indexed: 11/21/2022] Open
Abstract
Inflammasomes are multimeric protein complexes that initiate inflammatory cascades. Their activation is a hallmark of many infectious or inflammatory diseases. Their composition and activity are specified by proinflammatory stimuli. For example, the NLRP3 inflammasome is activated in response to cell damage and K+ efflux, whereas the AIM2 inflammasome is activated in response to cytosolic DNA. We used Legionella pneumophila, an intracellular bacterial pathogen that activates multiple inflammasomes, to elucidate the molecular mechanisms regulating inflammasome activation during infection. Upon infection, the AIM2 inflammasome engaged caspase-1 to induce pore formation in the cell membrane, which then caused K+-efflux-mediated activation of NLRP3. Thus, the AIM2 inflammasome amplifies signals of infection, triggering noncanonical activation of NLRP3. During infection, AIM2 and caspase-11 induced membrane damage, which was sufficient and essential for activating the NLRP3 inflammasome. Our data reveal that different inflammasomes regulate one another's activity to ensure an effective immune response to infection.
Collapse
|
36
|
Yuan YY, Xie KX, Wang SL, Yuan LW. Inflammatory caspase-related pyroptosis: mechanism, regulation and therapeutic potential for inflammatory bowel disease. Gastroenterol Rep (Oxf) 2018; 6:167-176. [PMID: 30151200 PMCID: PMC6101557 DOI: 10.1093/gastro/goy011] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/01/2018] [Accepted: 03/18/2018] [Indexed: 12/22/2022] Open
Abstract
As an essential part of programmed cell death, pyroptosis is an inflammatory response that is elicited upon infection by intracellular pathogens. Metabolic diseases, atherosclerosis and vital organ damage occur if pyroptosis is over-activated. Macrophages are the main cells that induce pyroptosis with the help of intracellular pattern-recognition receptors stimulated by danger signals and pathogenic microorganisms in the cytosol of host cells. Activated inflammatory caspases induce pyroptosis and produce pro-inflammatory cytokines, such as interleukin-1β and interleukin-18. Inflammatory programmed cell death is classified as canonical or non-canonical based on inflammatory caspases, which includes caspase-1 (in human and mouse) and caspase-11 (in mouse) or caspase-4 and -5 (in humans). Activated inflammatory caspases cleave the pore-forming effector protein, gasdermin-D, inducing osmotic pressure deregulation of internal fluids and subsequently rupturing the cell membranes. Inflammatory caspases could be attractive therapeutic targets for inflammatory bowel disease (IBD) in which pyroptosis may play an important role. This article reviews the current understanding of the mechanism of pyroptosis, focusing on the regulation of inflammatory caspases and therapeutic strategies for IBD.
Collapse
Affiliation(s)
- Yuan-Yuan Yuan
- Department of Geriatric Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Ke-Xin Xie
- Medical Laboratory Technology 1602, Central South University Xiangya School of Medicine, 172 Tongzipo Road, YueLu District, Changsha, Hunan 410011, China
| | - Sha-Long Wang
- Department of Geriatric Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Lian-Wen Yuan
- Department of Geriatric Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
37
|
Mirshafiee V, Sun B, Chang CH, Liao YP, Jiang W, Jiang J, Liu X, Wang X, Xia T, Nel AE. Toxicological Profiling of Metal Oxide Nanoparticles in Liver Context Reveals Pyroptosis in Kupffer Cells and Macrophages versus Apoptosis in Hepatocytes. ACS NANO 2018; 12:3836-3852. [PMID: 29543433 PMCID: PMC5946698 DOI: 10.1021/acsnano.8b01086] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
The liver and the mononuclear phagocyte system are a frequent target for engineered nanomaterials, either as a result of particle uptake and spread from primary exposure sites or systemic administration of therapeutic and imaging nanoparticles. In this study, we performed a comparative analysis of the toxicological impact of 29 metal oxide nanoparticles (NPs), some commonly used in consumer products, in transformed or primary Kupffer cells (KCs) and hepatocytes. We not only observed differences between KCs and hepatocytes, but also differences in the toxicological profiles of transition-metal oxides (TMOs, e. g., Co3O4) versus rare-earth oxide (REO) NPs ( e. g., Gd2O3). While pro-oxidative TMOs induced the activation of caspases 3 and 7, resulting in apoptotic cell death in both cell types, REOs induced lysosomal damage, NLRP3 inflammasome activation, caspase 1 activation, and pyroptosis in KCs. Pyroptosis was accompanied by cell swelling, membrane blebbing, IL-1β release, and increased membrane permeability, which could be reversed by knockdown of the pore forming protein, gasdermin D. Though similar features were not seen in hepatocytes, the investigation of the cytotoxic effects of REO NPs could also be seen to affect macrophage cell lines such as J774A.1 and RAW 264.7 cells as well as bone marrow-derived macrophages. These phagocytic cell types also demonstrated features of pyroptosis and increased IL-1β production. Collectively, these findings demonstrate important mechanistic considerations that can be used for safety evaluation of metal oxides, including commercial products that are developed from these materials.
Collapse
Affiliation(s)
- Vahid Mirshafiee
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
- Division of NanoMedicine, Department of Medicine, University of California Los Angeles, 10833 Le Conte Ave., Los Angeles, California 90095, United States
| | - Bingbing Sun
- Division of NanoMedicine, Department of Medicine, University of California Los Angeles, 10833 Le Conte Ave., Los Angeles, California 90095, United States
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, 2 Linggong Rd., Dalian 116024, China
| | - Chong Hyun Chang
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
| | - Yu Pei Liao
- Division of NanoMedicine, Department of Medicine, University of California Los Angeles, 10833 Le Conte Ave., Los Angeles, California 90095, United States
| | - Wen Jiang
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
| | - Jinhong Jiang
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
| | - Xiangsheng Liu
- Division of NanoMedicine, Department of Medicine, University of California Los Angeles, 10833 Le Conte Ave., Los Angeles, California 90095, United States
| | - Xiang Wang
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
| | - Tian Xia
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
- Division of NanoMedicine, Department of Medicine, University of California Los Angeles, 10833 Le Conte Ave., Los Angeles, California 90095, United States
- Address correspondence to: ;
| | - André E. Nel
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
- Division of NanoMedicine, Department of Medicine, University of California Los Angeles, 10833 Le Conte Ave., Los Angeles, California 90095, United States
- Address correspondence to: ;
| |
Collapse
|
38
|
Multitalented EspB of enteropathogenic Escherichia coli (EPEC) enters cells autonomously and induces programmed cell death in human monocytic THP-1 cells. Int J Med Microbiol 2018; 308:387-404. [PMID: 29550166 DOI: 10.1016/j.ijmm.2018.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/06/2018] [Accepted: 03/11/2018] [Indexed: 12/26/2022] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) subvert host cell signaling pathways by injecting effector proteins via a Type 3 Secretion System (T3SS). The T3SS-dependent EspB protein is a multi-functional effector protein, which contributes to adherence and translocator pore formation and after injection exhibits several intracellular activities. In addition, EspB is also secreted into the environment. Effects of secreted EspB have not been reported thus far. As a surrogate for secreted EspB we employed recombinant EspB (rEspB) derived from the prototype EPEC strain E2348/69 and investigated the interactions of the purified protein with different human epithelial and immune cells including monocytic THP-1 cells, macrophages, dendritic cells, U-937, epithelial T84, Caco-2, and HeLa cells. To assess whether these proteins might exert a cytotoxic effect we monitored the release of lactate dehydrogenase (LDH) as well as propidium iodide (PI) uptake. For comparison, we also investigated several homologs of EspB such as IpaD of Shigella, and SipC, SipD, SseB, and SseD of Salmonella as purified recombinant proteins. Interestingly, cytotoxicity was only observed in THP-1 cells and macrophages, whereas epithelial cells remained unaffected. Cell fractionation and immune fluorescence experiments showed that rEspB enters cells autonomously, which suggests that EspB might qualify as a novel cell-penetrating effector protein (CPE). Using specific organelle tracers and inhibitors of signaling pathways we found that rEspB destroys the mitochondrial membrane potential - an indication of programmed cell death induction in THP-1 cells. Here we show that EspB not only constitutes an essential part of the T3SS-nanomachine and contributes to the arsenal of injected effector proteins but, furthermore, that secreted (recombinant) EspB autonomously enters host cells and selectively induces cell death in immune cells.
Collapse
|
39
|
Pahari S, Kaur G, Negi S, Aqdas M, Das DK, Bashir H, Singh S, Nagare M, Khan J, Agrewala JN. Reinforcing the Functionality of Mononuclear Phagocyte System to Control Tuberculosis. Front Immunol 2018; 9:193. [PMID: 29479353 PMCID: PMC5811511 DOI: 10.3389/fimmu.2018.00193] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 01/23/2018] [Indexed: 12/12/2022] Open
Abstract
The mononuclear phagocyte system (MPS) constitutes dendritic cells, monocytes, and macrophages. This system contributes to various functions that are essential for maintaining homeostasis, activation of innate immunity, and bridging it with the adaptive immunity. Consequently, MPS is highly important in bolstering immunity against the pathogens. However, MPS is the frontline cells in destroying Mycobacterium tuberculosis (Mtb), yet the bacterium prefers to reside in the hostile environment of macrophages. Therefore, it may be very interesting to study the struggle between Mtb and MPS to understand the outcome of the disease. In an event when MPS predominates Mtb, the host remains protected. By contrast, the situation becomes devastating when the pathogen tames and tunes the host MPS, which ultimately culminates into tuberculosis (TB). Hence, it becomes extremely crucial to reinvigorate MPS functionality to overwhelm Mtb and eliminate it. In this article, we discuss the strategies to bolster the function of MPS by exploiting the molecules associated with the innate immunity and highlight the mechanisms involved to overcome the Mtb-induced suppression of host immunity. In future, such approaches may provide an insight to develop immunotherapeutics to treat TB.
Collapse
Affiliation(s)
- Susanta Pahari
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Gurpreet Kaur
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Shikha Negi
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Mohammad Aqdas
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Deepjyoti K Das
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Hilal Bashir
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Sanpreet Singh
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Mukta Nagare
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Junaid Khan
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Javed N Agrewala
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| |
Collapse
|
40
|
NOD-like receptor(s) and host immune responses with Pseudomonas aeruginosa infection. Inflamm Res 2018; 67:479-493. [PMID: 29353310 DOI: 10.1007/s00011-018-1132-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/12/2018] [Accepted: 01/16/2018] [Indexed: 01/01/2023] Open
Abstract
INTRODUCTION Molecular mechanisms underlying the interactions between Pseudomonas aeruginosa, the common opportunistic pathogen in cystic fibrosis individuals, and host induce a number of marked inflammatory responses and associate with complex therapeutic problems due to bacterial resistance to antibiotics in chronic stage of infection. METHODS Pseudomonas aeruginosa is recognized by number of pattern recognition receptors (PRRs); NOD-like receptors (NLRs) are a class of PRRs, which can recognize a variety of endogenous and exogenous ligands, thereby playing a critical role in innate immunity. RESULTS NLR activation initiates forming of a multi-protein complex called inflammasome that induces activation of caspase-1 and resulted in cleavage of pro-inflammatory cytokines interleukin (IL)-1β and IL-18. When the IL-1β is secreted excessively, this causes tissue damage and extensive inflammatory responses that are potentially hazardous for the host. CONCLUSIONS Recent evidence has laid out inflammasome-forming NLR far beyond inflammation. This review summarizes current knowledge regarding the various roles played by different NLRs and associated down-signals, either in recognition of P. aeruginosa or may be associated with such bacterial pathogen infection, which may relate to for the complexity of lung diseases caused by P. aeruginosa.
Collapse
|
41
|
Moreira-Souza ACA, Almeida-da-Silva CLC, Rangel TP, Rocha GDC, Bellio M, Zamboni DS, Vommaro RC, Coutinho-Silva R. The P2X7 Receptor Mediates Toxoplasma gondii Control in Macrophages through Canonical NLRP3 Inflammasome Activation and Reactive Oxygen Species Production. Front Immunol 2017; 8:1257. [PMID: 29075257 PMCID: PMC5643413 DOI: 10.3389/fimmu.2017.01257] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/21/2017] [Indexed: 12/12/2022] Open
Abstract
Toxoplasma gondii (T. gondii) is the protozoan parasite that causes toxoplasmosis, a potentially fatal disease to immunocompromised patients, and which affects approximately 30% of the world’s population. Previously, we showed that purinergic signaling via the P2X7 receptor contributes to T. gondii elimination in macrophages, through reactive oxygen species (ROS) production and lysosome fusion with the parasitophorous vacuole. Moreover, we demonstrated that P2X7 receptor activation promotes the production of anti-parasitic pro-inflammatory cytokines during early T. gondii infection in vivo. However, the cascade of signaling events that leads to parasite elimination via P2X7 receptor activation remained to be elucidated. Here, we investigated the cellular pathways involved in T. gondii elimination triggered by P2X7 receptor signaling, during early infection in macrophages. We focused on the potential role of the inflammasome, a protein complex that can be co-activated by the P2X7 receptor, and which is involved in the host immune defense against T. gondii infection. Using peritoneal and bone marrow-derived macrophages from knockout mice deficient for inflammasome components (NLRP3−/−, Caspase-1/11−/−, Caspase-11−/−), we show that the control of T. gondii infection via P2X7 receptor activation by extracellular ATP (eATP) depends on the canonical inflammasome effector caspase-1, but not on caspase-11 (a non-canonical inflammasome effector). Parasite elimination via P2X7 receptor and inflammasome activation was also dependent on ROS generation and pannexin-1 channel. Treatment with eATP increased IL-1β secretion from infected macrophages, and this effect was dependent on the canonical NLRP3 inflammasome. Finally, treatment with recombinant IL-1β promoted parasite elimination via mitochondrial ROS generation (as assessed using Mito-TEMPO). Together, our results support a model where P2X7 receptor activation by eATP inhibits T. gondii growth in macrophages by triggering NADPH-oxidase-dependent ROS production, and also by activating a canonical NLRP3 inflammasome, which increases IL-1β production (via caspase-1 activity), leading to mitochondrial ROS generation.
Collapse
Affiliation(s)
- Aline Cristina Abreu Moreira-Souza
- Immunobiology Program, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Parasitology and Cell Biology Program, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Thuany Prado Rangel
- Immunobiology Program, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Parasitology and Cell Biology Program, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gabrielle da Costa Rocha
- Immunobiology Program, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maria Bellio
- Department of Immunology, Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Dario Simões Zamboni
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Rossiane Claudia Vommaro
- Parasitology and Cell Biology Program, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Robson Coutinho-Silva
- Immunobiology Program, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
42
|
Cecil JD, O'Brien-Simpson NM, Lenzo JC, Holden JA, Singleton W, Perez-Gonzalez A, Mansell A, Reynolds EC. Outer Membrane Vesicles Prime and Activate Macrophage Inflammasomes and Cytokine Secretion In Vitro and In Vivo. Front Immunol 2017; 8:1017. [PMID: 28890719 PMCID: PMC5574916 DOI: 10.3389/fimmu.2017.01017] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 08/08/2017] [Indexed: 12/16/2022] Open
Abstract
Outer membrane vesicles (OMVs) are proteoliposomes blebbed from the surface of Gram-negative bacteria. Chronic periodontitis is associated with an increase in subgingival plaque of Gram-negative bacteria, Porphyromonas gingivalis, Treponema denticola, and Tannerella forsythia. In this study, we investigated the immune-modulatory effects of P. gingivalis, T. denticola, and T. forsythia OMVs on monocytes and differentiated macrophages. All of the bacterial OMVs were phagocytosed by monocytes, M(naïve) and M(IFNγ) macrophages in a dose-dependent manner. They also induced NF-κB activation and increased TNFα, IL-8, and IL-1β cytokine secretion. P. gingivalis OMVs were also found to induce anti-inflammatory IL-10 secretion. Although unprimed monocytes and macrophages were resistant to OMV-induced cell death, lipopolysaccharide or OMV priming resulted in a significantly reduced cell viability. P. gingivalis, T. denticola, and T. forsythia OMVs all activated inflammasome complexes, as monitored by IL-1β secretion and ASC speck formation. ASC was critical for OMV-induced inflammasome formation, while AIM2-/- and Caspase-1-/- cells had significantly reduced inflammasome formation and NLRP3-/- cells exhibited a slight reduction. OMVs were also found to provide both priming and activation of the inflammasome complex. High-resolution microscopy and flow cytometry showed that P. gingivalis OMVs primed and activated macrophage inflammasomes in vivo with 80% of macrophages exhibiting inflammasome complex formation. In conclusion, periodontal pathogen OMVs were found to have significant immunomodulatory effects upon monocytes and macrophages and should therefore influence pro-inflammatory host responses associated with disease.
Collapse
Affiliation(s)
- Jessica D Cecil
- Oral Health CRC, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - Neil M O'Brien-Simpson
- Oral Health CRC, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - Jason C Lenzo
- Oral Health CRC, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - James A Holden
- Oral Health CRC, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - William Singleton
- Oral Health CRC, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - Alexis Perez-Gonzalez
- Oral Health CRC, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - Ashley Mansell
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Eric C Reynolds
- Oral Health CRC, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
43
|
Yogarajah T, Ong KC, Perera D, Wong KT. AIM2 Inflammasome-Mediated Pyroptosis in Enterovirus A71-Infected Neuronal Cells Restricts Viral Replication. Sci Rep 2017; 7:5845. [PMID: 28724943 PMCID: PMC5517550 DOI: 10.1038/s41598-017-05589-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 05/31/2017] [Indexed: 01/17/2023] Open
Abstract
Encephalomyelitis is a well-known complication of hand, foot, and mouth disease (HFMD) due to Enterovirus 71 (EV71) infection. Viral RNA/antigens could be detected in the central nervous system (CNS) neurons in fatal encephalomyelitis but the mechanisms of neuronal cell death is not clearly understood. We investigated the role of absent in melanoma 2 (AIM2) inflammasome in neuronal cell death, and its relationship to viral replication. Our transcriptomic analysis, RT-qPCR, Western blot, immunofluorescence and flow cytometry studies consistently showed AIM2 gene up-regulation and protein expression in EV-A71-infected SK-N-SH cells. Downstream AIM2-induced genes, CARD16, caspase-1 and IL-1β were also up-regulated and caspase-1 was activated to form cleaved caspase-1 p20 subunits. As evidenced by 7-AAD positivity, pyroptosis was confirmed in infected cells. Overall, these findings have a strong correlation with decreases in viral titers, copy numbers and proteins, and reduced proportions of infected cells. AIM2 and viral antigens were detected by immunohistochemistry in infected neurons in inflamed areas of the CNS in EV-A71 encephalomyelitis. In infected AIM2-knockdown cells, AIM2 and related downstream gene expressions, and pyroptosis were suppressed, resulting in significantly increased virus infection. These results support the notion that AIM2 inflammasome-mediated pyroptosis is an important mechanism of neuronal cell death and it could play an important role in limiting EV-A71 replication.
Collapse
Affiliation(s)
- Thinesshwary Yogarajah
- Department of Pathology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Kien Chai Ong
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - David Perera
- Institute of Health and Community Medicine, University Malaysia Sarawak, Sarawak, Malaysia
| | - Kum Thong Wong
- Department of Pathology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| |
Collapse
|
44
|
Garib FY, Rizopulu AP, Kuchmiy AA, Garib VF. Inactivation of Inflammasomes by Pathogens Regulates Inflammation. BIOCHEMISTRY (MOSCOW) 2017; 81:1326-1339. [PMID: 27914458 DOI: 10.1134/s0006297916110109] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Inflammatory response is initiated and sustained by the action of quintessential pro-inflammatory cytokines of immune system namely IL-1β and IL-18. The maturation process of those cytokines is ensured by caspase-1 enzymatic activity, that is in turn is tightly controlled by multiprotein complexes called inflammasomes. Inflammasomes are activated in cells of innate immune system in response to recognition of conservative parts of microbes (pathogen-associated molecular patterns) or by sensing molecular signs of tissue damage (damage-associated molecular patterns). Inflammasome activation apart of cytokines secretion leads to pro-inflammatory cell death, so-called pyroptosis. That culminates in release of cytoplasmatic content of cells including cytokines and alarmins that boost immune response against pathogens, as well as pyroptosis destroys replicative niches of intracellular pathogens. During co-evolution with the host, bacterial and viral pathogens developed a range of molecular inhibitors targeting each step of inflammasome activation. In current review, we will discuss the latest knowledge of inflammasomes' signaling pathways and tricks that pathogens use to avoid immune recognition and clearance. Our better understanding of inflammasome inhibition by pathogens can lead to better therapeutic approaches for the treatment of infectious diseases.
Collapse
Affiliation(s)
- F Yu Garib
- Lomonosov Moscow State University, Biological Faculty, Moscow, 119991, Russia
| | | | | | | |
Collapse
|
45
|
Karaji N, Sattentau QJ. Efferocytosis of Pathogen-Infected Cells. Front Immunol 2017; 8:1863. [PMID: 29312342 PMCID: PMC5743670 DOI: 10.3389/fimmu.2017.01863] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/07/2017] [Indexed: 12/20/2022] Open
Abstract
The prompt and efficient clearance of unwanted and abnormal cells by phagocytes is termed efferocytosis and is crucial for organism development, maintenance of tissue homeostasis, and regulation of the immune system. Dying cells are recognized by phagocytes through pathways initiated via "find me" signals, recognition via "eat me" signals and down-modulation of regulatory "don't eat me" signals. Pathogen infection may trigger cell death that drives phagocytic clearance in an immunologically silent, or pro-inflammatory manner, depending on the mode of cell death. In many cases, efferocytosis is a mechanism for eliminating pathogens and pathogen-infected cells; however, some pathogens have subverted this process and use efferocytic mechanisms to avoid innate immune detection and assist phagocyte infection. In parallel, phagocytes can integrate signals received from infected dying cells to elicit the most appropriate effector response against the infecting pathogen. This review focuses on pathogen-induced cell death signals that drive infected cell recognition and uptake by phagocytes, and the outcomes for the infected target cell, the phagocyte, the pathogen and the host.
Collapse
Affiliation(s)
- Niloofar Karaji
- The Sir William Dunn School of Pathology, The University of Oxford, Oxford, United Kingdom
| | - Quentin J Sattentau
- The Sir William Dunn School of Pathology, The University of Oxford, Oxford, United Kingdom
| |
Collapse
|
46
|
Escoll P, Rolando M, Buchrieser C. MAMs are attractive targets for bacterial repurposing of the host cell: MAM-functions might be key for undermining an infected cell. Bioessays 2016; 39. [PMID: 28026026 DOI: 10.1002/bies.201600171] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pathogenic bacteria frequently target the endoplasmic reticulum (ER) and mitochondria in order to exploit host functions. ER-mitochondria inter-organelle communication is topologically sub-compartmentalized at mitochondria-associated ER membranes (MAMs). MAMs are specific membranous microdomains with unique regulatory functions such as lipid synthesis and trafficking, calcium homeostasis, mitochondrial morphology, inflammasome activation, autophagosome formation, and apoptosis. These important cellular processes are all modulated by pathogens to subvert host functions and promote infection, thus it is tempting to assume that pathogenic bacteria target MAMs to subvert these different pathways in their hosts. First lines of evidence that support this hypothesis come from Legionella pneumophila. This intracellular bacterium secretes an effector that exhibits sphingosine-1 phosphate lyase activity (LpSpl) that seems to target MAMs to modulate the autophagy response to infection. Here we thus propose the concept that MAMs could be targeted by pathogenic bacteria to undermine key host cellular processes.
Collapse
Affiliation(s)
- Pedro Escoll
- Institut Pasteur, Biologie des Bactéries Intracellulaires, Paris, France.,CNRS UMR 3525, Paris, France
| | - Monica Rolando
- Institut Pasteur, Biologie des Bactéries Intracellulaires, Paris, France.,CNRS UMR 3525, Paris, France
| | - Carmen Buchrieser
- Institut Pasteur, Biologie des Bactéries Intracellulaires, Paris, France.,CNRS UMR 3525, Paris, France
| |
Collapse
|
47
|
Yen H, Karino M, Tobe T. Modulation of the Inflammasome Signaling Pathway by Enteropathogenic and Enterohemorrhagic Escherichia coli. Front Cell Infect Microbiol 2016; 6:89. [PMID: 27617233 PMCID: PMC4999430 DOI: 10.3389/fcimb.2016.00089] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 08/11/2016] [Indexed: 01/07/2023] Open
Abstract
Innate immunity is an essential component in the protection of a host against pathogens. Enteropathogenic and enterohemorrhagic Escherichia coli (EPEC and EHEC, respectively) are known to modulate the innate immune responses of infected cells. The interference is dependent on their type III secretion system (T3SS) and T3SS-dependent effector proteins. Furthermore, these cytosolically injected effectors have been demonstrated to engage multiple immune signaling pathways, including the IFN/STAT, MAPK, NF-κB, and inflammasome pathways. In this review, recent work describing the interaction between EPEC/EHEC and the inflammasome pathway will be discussed.
Collapse
Affiliation(s)
- Hilo Yen
- Department of Biomedical Informatics, Graduate School of Medicine, Osaka University Osaka, Japan
| | - Masaki Karino
- Department of Biomedical Informatics, Graduate School of Medicine, Osaka University Osaka, Japan
| | - Toru Tobe
- Department of Biomedical Informatics, Graduate School of Medicine, Osaka University Osaka, Japan
| |
Collapse
|
48
|
Chitinase 3-Like 1 (Chil1) Regulates Survival and Macrophage-Mediated Interleukin-1β and Tumor Necrosis Factor Alpha during Pseudomonas aeruginosa Pneumonia. Infect Immun 2016; 84:2094-2104. [PMID: 27141083 DOI: 10.1128/iai.00055-16] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 04/27/2016] [Indexed: 01/06/2023] Open
Abstract
Pseudomonas aeruginosa causes hospital-acquired pneumonia and is associated with high mortality. An effective response to such an infection includes efficient clearance of pathogenic organisms while limiting collateral damage from the host inflammatory response, known as host resistance and host tolerance, respectively. P. aeruginosa expresses a type III secretion system (T3SS) needle complex that induces NLRC4 (NOD-like receptor C4) activation, interleukin-1β (IL-1β) production, and host tissue damage. Chitinase 3-like-1 (Chil1) is expressed during infection and binds to its receptor, IL-13 receptor α2 (IL-13Rα2), to regulate the pathogen-host response during Streptococcus pneumoniae infection, but the role Chil1 plays in balancing the host resistance and host tolerance during P. aeruginosa pneumonia is not known. We conducted experiments using C57BL/6 mice with or without a genetic deficiency of Chil1 and demonstrated that Chil1-deficient mice succumb to P. aeruginosa infection more rapidly than the wild type (WT). The decreased survival time in infected Chil1-deficient mice is associated with more neutrophils recruited to the airways, more lung parenchymal damage, and increased pulmonary consolidation while maintaining equivalent bacterial killing compared to WT mice. Infected Chil1-deficient mice and bone marrow-derived macrophages (BMDMs) from Chil1-deficient mice have increased production of tumor necrosis factor alpha (TNF-α) and IL-1β compared to infected WT mice and macrophages. Infection of Chil1-deficient BMDMs with non-NLRC4-triggering P. aeruginosa, which is deficient in the T3SS needle complex, did not alter the excessive IL-1β production compared to BMDMs from WT mice. The addition of recombinant Chil1 decreases the excessive IL-1β production but only partially rescues stimulated BMDMs from IL-13Rα2-deficient mice. Our data provide mechanistic insights into how Chil1 regulates P. aeruginosa-induced host responses.
Collapse
|
49
|
Mitchell G, Chen C, Portnoy DA. Strategies Used by Bacteria to Grow in Macrophages. Microbiol Spectr 2016; 4:10.1128/microbiolspec.MCHD-0012-2015. [PMID: 27337444 PMCID: PMC4922531 DOI: 10.1128/microbiolspec.mchd-0012-2015] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Indexed: 12/24/2022] Open
Abstract
Intracellular bacteria are often clinically relevant pathogens that infect virtually every cell type found in host organisms. However, myeloid cells, especially macrophages, constitute the primary cells targeted by most species of intracellular bacteria. Paradoxically, macrophages possess an extensive antimicrobial arsenal and are efficient at killing microbes. In addition to their ability to detect and signal the presence of pathogens, macrophages sequester and digest microorganisms using the phagolysosomal and autophagy pathways or, ultimately, eliminate themselves through the induction of programmed cell death. Consequently, intracellular bacteria influence numerous host processes and deploy sophisticated strategies to replicate within these host cells. Although most intracellular bacteria have a unique intracellular life cycle, these pathogens are broadly categorized into intravacuolar and cytosolic bacteria. Following phagocytosis, intravacuolar bacteria reside in the host endomembrane system and, to some extent, are protected from the host cytosolic innate immune defenses. However, the intravacuolar lifestyle requires the generation and maintenance of unique specialized bacteria-containing vacuoles and involves a complex network of host-pathogen interactions. Conversely, cytosolic bacteria escape the phagolysosomal pathway and thrive in the nutrient-rich cytosol despite the presence of host cell-autonomous defenses. The understanding of host-pathogen interactions involved in the pathogenesis of intracellular bacteria will continue to provide mechanistic insights into basic cellular processes and may lead to the discovery of novel therapeutics targeting infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Gabriel Mitchell
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Chen Chen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Daniel A. Portnoy
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- School of Public Health, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
50
|
Type I Interferon Counters or Promotes Coxiella burnetii Replication Dependent on Tissue. Infect Immun 2016; 84:1815-1825. [PMID: 27068091 DOI: 10.1128/iai.01540-15] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 04/01/2016] [Indexed: 01/09/2023] Open
Abstract
Coxiella burnetii is an intracellular pathogen and the cause of Q fever. Gamma interferon (IFN-γ) is critical for host protection from infection, but a role for type I IFN in C. burnetii infection has not been determined. Type I IFN supports host protection from a related pathogen, Legionella pneumophila, and we hypothesized that it would be similarly protective in C. burnetii infection. In contrast to our prediction, IFN-α receptor-deficient (IFNAR(-/-)) mice were protected from C. burnetii-induced infection. Therefore, the role of type I IFN in C. burnetii infection was distinct from that in L. pneumophila Mice treated with a double-stranded-RNA mimetic were protected from C. burnetii-induced weight loss through an IFNAR-independent pathway. We next treated mice with recombinant IFN-α (rIFN-α). When rIFN-α was injected by the intraperitoneal route during infection, disease-induced weight loss was exacerbated. Mice that received rIFN-α by this route had dampened interleukin 1β (IL-1β) expression in bronchoalveolar lavage fluids. However, when rIFN-α was delivered to the lung, bacterial replication was decreased in all tissues. Thus, the presence of type I IFN in the lung protected from infection, but when delivered to the periphery, type I IFN enhanced disease, potentially by dampening inflammatory cytokines. To better characterize the capacity for type I IFN induction by C. burnetii, we assessed expression of IFN-β transcripts by human macrophages following stimulation with lipopolysaccharide (LPS) from C. burnetii Understanding innate responses in C. burnetii infection will support the discovery of novel therapies that may be alternative or complementary to the current antibiotic treatment.
Collapse
|