1
|
Goldberg K, Lobov A, Antonello P, Shmueli MD, Yakir I, Weizman T, Ulman A, Sheban D, Laser E, Kramer MP, Shteinvil R, Chen G, Ibraheem A, Sysoeva V, Fishbain-Yoskovitz V, Mohapatra G, Abramov A, Shimshi S, Ogneva K, Nandy M, Amidror S, Bootz-Maoz H, Kuo SH, Dezorella N, Kacen A, Javitt A, Lau GW, Yissachar N, Hayouka Z, Merbl Y. Cell-autonomous innate immunity by proteasome-derived defence peptides. Nature 2025; 639:1032-1041. [PMID: 40044870 PMCID: PMC11946893 DOI: 10.1038/s41586-025-08615-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 01/08/2025] [Indexed: 03/25/2025]
Abstract
For decades, antigen presentation on major histocompatibility complex class I for T cell-mediated immunity has been considered the primary function of proteasome-derived peptides1,2. However, whether the products of proteasomal degradation play additional parts in mounting immune responses remains unknown. Antimicrobial peptides serve as a first line of defence against invading pathogens before the adaptive immune system responds. Although the protective function of antimicrobial peptides across numerous tissues is well established, the cellular mechanisms underlying their generation are not fully understood. Here we uncover a role for proteasomes in the constitutive and bacterial-induced generation of defence peptides that impede bacterial growth both in vitro and in vivo by disrupting bacterial membranes. In silico prediction of proteome-wide proteasomal cleavage identified hundreds of thousands of potential proteasome-derived defence peptides with cationic properties that may be generated en route to degradation to act as a first line of defence. Furthermore, bacterial infection induces changes in proteasome composition and function, including PSME3 recruitment and increased tryptic-like cleavage, enhancing antimicrobial activity. Beyond providing mechanistic insights into the role of proteasomes in cell-autonomous innate immunity, our study suggests that proteasome-cleaved peptides may have previously overlooked functions downstream of degradation. From a translational standpoint, identifying proteasome-derived defence peptides could provide an untapped source of natural antibiotics for biotechnological applications and therapeutic interventions in infectious diseases and immunocompromised conditions.
Collapse
Affiliation(s)
- Karin Goldberg
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Arseniy Lobov
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Paola Antonello
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Merav D Shmueli
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Idan Yakir
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agricultural, Food & Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Tal Weizman
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Adi Ulman
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Daoud Sheban
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Einav Laser
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Matthias P Kramer
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Ronen Shteinvil
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Guoyun Chen
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Angham Ibraheem
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Vera Sysoeva
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | | | - Gayatree Mohapatra
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Anat Abramov
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Sandy Shimshi
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Kseniia Ogneva
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Madhurima Nandy
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Sivan Amidror
- The Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
- Bar-Ilan Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, Israel
| | - Hadar Bootz-Maoz
- The Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
- Bar-Ilan Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, Israel
| | - Shanny H Kuo
- Department of Pathobiology, University of Illinois, Urbana, IL, USA
| | - Nili Dezorella
- Electron Microscopy Unit, Weizmann Institute of Science, Rehovot, Israel
| | - Assaf Kacen
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Aaron Javitt
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Gee W Lau
- Department of Pathobiology, University of Illinois, Urbana, IL, USA
| | - Nissan Yissachar
- The Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
- Bar-Ilan Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, Israel
| | - Zvi Hayouka
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agricultural, Food & Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Yifat Merbl
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
2
|
Abstract
Enteric viruses encounter various bacteria in the host, which can impact infection outcomes. The interactions between noroviruses and enteric bacteria are not well understood. Previous work determined that murine norovirus (MNV), a model norovirus, had decreased replication in antibiotic-treated mice compared with conventional mice. Although this suggests that the microbiota promotes MNV infection, the mechanisms are not completely understood. Additionally, prior work with other enteric viruses, such as poliovirus and coxsackievirus B3, demonstrated that virions bind bacteria, and exposure to bacteria stabilizes viral particles and limits premature RNA release. Therefore, we examined interactions between MNV and specific bacteria and the consequences of these interactions. We found that the majority of Gram-positive bacteria tested stabilized MNV, while Gram-negative bacteria did not stabilize MNV. Both Gram-positive and Gram-negative bacteria bound to MNV. However, bacterial binding alone was not sufficient for virion stabilization, since Gram-negative bacteria bound MNV but did not stabilize virions. Additionally, we found that bacteria conditioned medium also stabilized MNV and this stabilization may be due to a small heat-stable molecule. Overall, this work identifies specific bacteria and bacterial components that stabilize MNV and may impact virion stability in the environment. IMPORTANCE Enteric viruses are exposed to a wide variety of bacteria in the intestine, but the effects of bacteria on viral particles are incompletely understood. We found that murine norovirus (MNV) virion stability is enhanced in the presence of several Gram-positive bacterial strains. Virion-stabilizing activity was also present in bacterial culture medium, and activity was retained upon heat or protease treatment. These results suggest that certain bacteria and bacterial products may promote MNV stability in the environment, which could influence viral transmission.
Collapse
Affiliation(s)
- Melissa R. Budicini
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Julie K. Pfeiffer
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
3
|
Frazier K, Kambal A, Zale EA, Pierre JF, Hubert N, Miyoshi S, Miyoshi J, Ringus DL, Harris D, Yang K, Carroll K, Hermanson JB, Chlystek JS, Overmyer KA, Cham CM, Musch MW, Coon JJ, Chang EB, Leone VA. High-fat diet disrupts REG3γ and gut microbial rhythms promoting metabolic dysfunction. Cell Host Microbe 2022; 30:809-823.e6. [PMID: 35439436 PMCID: PMC9281554 DOI: 10.1016/j.chom.2022.03.030] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/22/2021] [Accepted: 03/23/2022] [Indexed: 11/24/2022]
Abstract
Gut microbial diurnal oscillations are important diet-dependent drivers of host circadian rhythms and metabolism ensuring optimal energy balance. However, the interplay between diet, microbes, and host factors sustaining intestinal oscillations is complex and poorly understood. Here, using a mouse model, we report the host C-type lectin antimicrobial peptide Reg3γ works with key ileal microbes to orchestrate these interactions in a bidirectional manner and does not correlate with the intestinal core circadian clock. High-fat diet is the primary driver of microbial oscillators that impair host metabolic homeostasis, resulting in arrhythmic host Reg3γ expression that secondarily drives abundance and oscillation of key gut microbes. This illustrates transkingdom coordination of biological rhythms primarily influenced by diet and reciprocal sensor-effector signals between host and microbial components, ultimately driving metabolism. Restoring the gut microbiota's capacity to sense dietary signals mediated by specific host factors such as Reg3γ could be harnessed to improve metabolic dysfunction.
Collapse
Affiliation(s)
- Katya Frazier
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Amal Kambal
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Elizabeth A Zale
- Infectious Diseases Division, Weill Cornell Medicine, New York, NY 10065, USA
| | - Joseph F Pierre
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Nathaniel Hubert
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Sawako Miyoshi
- Department of General Medicine, Kyorin University School of Medicine, Tokyo 1818611, Japan
| | - Jun Miyoshi
- Department of Gastroenterology and Hepatology, Kyorin University School of Medicine, Tokyo 1818611, Japan
| | - Daina L Ringus
- Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Dylan Harris
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Karen Yang
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Katherine Carroll
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Jake B Hermanson
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - John S Chlystek
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53506, USA
| | - Katherine A Overmyer
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53506, USA; National Center for Quantitative Biology of Complex Systems, Madison, WI 53706, USA; Morgridge Institute for Research, Madison, WI 53715, USA
| | - Candace M Cham
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Mark W Musch
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Joshua J Coon
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53506, USA; National Center for Quantitative Biology of Complex Systems, Madison, WI 53706, USA; Morgridge Institute for Research, Madison, WI 53715, USA; Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Eugene B Chang
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Vanessa A Leone
- Department of Animal & Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
4
|
Amorim Neto DP, Bosque BP, Pereira de Godoy JV, Rodrigues PV, Meneses DD, Tostes K, Costa Tonoli CC, Faustino de Carvalho H, González-Billault C, de Castro Fonseca M. Akkermansia muciniphila induces mitochondrial calcium overload and α -synuclein aggregation in an enteroendocrine cell line. iScience 2022; 25:103908. [PMID: 35243260 PMCID: PMC8881719 DOI: 10.1016/j.isci.2022.103908] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/06/2021] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota influence neurodevelopment, modulate behavior, and contribute to neurodegenerative disorders. Several studies have consistently reported a greater abundance of Akkermansia muciniphila in Parkinson disease (PD) fecal samples. Therefore, we investigated whether A.muciniphila-conditioned medium (CM) could initiate α-synuclein (αSyn) misfolding in enteroendocrine cells (EEC) — a component of the gut epithelium featuring neuron-like properties. We found that A. muciniphila CM composition is influenced by the ability of the strain to degrade mucin. Our in vitro experiments showed that the protein-enriched fraction of mucin-free CM induces RyR-mediated Ca2+ release and increased mitochondrial Ca2+ uptake leading to ROS generation and αSyn aggregation. Oral administration of A. muciniphila cultivated in the absence of mucin to mice led to αSyn aggregation in cholecystokinin (CCK)-positive EECs but no motor deficits were observed. Noteworthy, buffering mitochondrial Ca2+ reverted the damaging effects observed. These molecular insights offer evidence that bacterial proteins can induce αSyn aggregation in EECs. Gut bacterium Akkermansia muciniphila is increased in patients with Parkinson disease A. muciniphila-conditioned medium induces mitochondrial Ca2+ overload in EECs Mitochondrial Ca2+ overload leads to ROS generation and αSyn aggregation in vitro Buffering mitochondrial Ca2+ inhibits A. muciniphila-induced αSyn aggregation
Collapse
Affiliation(s)
- Dionísio Pedro Amorim Neto
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 10000 Giuseppe Maximo Scolfaro St., 13083-100 Campinas, São Paulo, Brazil
- Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Beatriz Pelegrini Bosque
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 10000 Giuseppe Maximo Scolfaro St., 13083-100 Campinas, São Paulo, Brazil
- Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - João Vitor Pereira de Godoy
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 10000 Giuseppe Maximo Scolfaro St., 13083-100 Campinas, São Paulo, Brazil
- Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Paulla Vieira Rodrigues
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 10000 Giuseppe Maximo Scolfaro St., 13083-100 Campinas, São Paulo, Brazil
- Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Dario Donoso Meneses
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 10000 Giuseppe Maximo Scolfaro St., 13083-100 Campinas, São Paulo, Brazil
| | - Katiane Tostes
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 10000 Giuseppe Maximo Scolfaro St., 13083-100 Campinas, São Paulo, Brazil
| | - Celisa Caldana Costa Tonoli
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 10000 Giuseppe Maximo Scolfaro St., 13083-100 Campinas, São Paulo, Brazil
| | | | - Christian González-Billault
- Department of Biology, Faculty of Sciences and Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
- The Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Matheus de Castro Fonseca
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 10000 Giuseppe Maximo Scolfaro St., 13083-100 Campinas, São Paulo, Brazil
- Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
- Corresponding author
| |
Collapse
|
5
|
Mucosal IFNγ production and potential role in protection in Escherichia coli O157:H7 vaccinated and challenged cattle. Sci Rep 2021; 11:9769. [PMID: 33963240 PMCID: PMC8105325 DOI: 10.1038/s41598-021-89113-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 04/15/2021] [Indexed: 11/16/2022] Open
Abstract
Shiga-toxin producing Escherichia coli O157:H7 (O157)-based vaccines can provide a potential intervention strategy to limit foodborne zoonotic transmission of O157. While the peripheral antibody response to O157 vaccination has been characterized, O157-specific cellular immunity at the rectoanal junction (RAJ), a preferred site for O157 colonization, remains poorly described. Vaccine induced mucosal O157-specific antibodies likely provide some protection, cellular immune responses at the RAJ may also play a role in protection. Distinct lymphoid follicles were increased in the RAJ of vaccinated/challenged animals. Additionally, increased numbers of interferon (IFN)γ-producing cells and γδ + T cells were detected in the follicular region of the RAJ of vaccinated/challenged animals. Likewise, adjuvanted-vaccine formulation is critical in immunogenicity of the O157 parenteral vaccine. Local T cell produced IFNγ may impact epithelial cells, subsequently limiting O157 adherence, which was demonstrated using in vitro attachment assays with bovine epithelial cells. Thus, distinct immune changes induced at the mucosa of vaccinated and challenged animals provide insight of mechanisms associated with limiting O157 fecal shedding. Enhancing mucosal immunity may be critical in the further development of efficacious vaccines for controlling O157 in ruminants and thus limiting O157 transmission to humans.
Collapse
|
6
|
Stromberg ZR, Van Goor A, Redweik GAJ, Wymore Brand MJ, Wannemuehler MJ, Mellata M. Pathogenic and non-pathogenic Escherichia coli colonization and host inflammatory response in a defined microbiota mouse model. Dis Model Mech 2018; 11:dmm035063. [PMID: 30275104 PMCID: PMC6262807 DOI: 10.1242/dmm.035063] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 09/24/2018] [Indexed: 12/17/2022] Open
Abstract
Most Escherichia coli strains in the human intestine are harmless. However, enterohemorrhagic Ecoli (EHEC) is a foodborne pathogen that causes intestinal disease in humans. Conventionally reared (CONV) mice are inconsistent models for human infections with EHEC because they are often resistant to Ecoli colonization, in part due to their gastrointestinal (GI) microbiota. Although antibiotic manipulation of the mouse microbiota has been a common means to overcome colonization resistance, these models have limitations. Currently, there are no licensed treatments for clinical EHEC infections and, thus, new tools to study EHEC colonization need to be developed. Here, we used a defined microbiota mouse model, consisting of the altered Schaedler flora (ASF), to characterize intestinal colonization and compare host responses following colonization with EHEC strain 278F2 or non-pathogenic Ecoli strain MG1655. Significantly higher (P<0.05) levels of both strains were found in feces and cecal and colonic contents of C3H/HeN ASF compared to C3H/HeN CONV mice. GI inflammation was significantly elevated (P<0.05) in the cecum of EHEC 278F2-colonized compared to E. coli MG1655-colonized C3H/HeN ASF mice. In addition, EHEC 278F2 differentially modulated inflammatory-associated genes in colonic tissue of C3H/HeN ASF mice compared to E. coli MG1655-colonized mice. This approach allowed for prolonged colonization of the murine GI tract by pathogenic and non-pathogenic Ecoli strains, and for evaluation of host inflammatory processes. Overall, this system can be used as a powerful tool for future studies to assess therapeutics, microbe-microbe interactions, and strategies for preventing EHEC infections.
Collapse
Affiliation(s)
- Zachary R Stromberg
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA 50011, USA
| | - Angelica Van Goor
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA 50011, USA
| | - Graham A J Redweik
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA 50011, USA
| | - Meghan J Wymore Brand
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50011, USA
| | - Michael J Wannemuehler
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50011, USA
| | - Melha Mellata
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
7
|
Han R, Xu L, Wang T, Liu B, Wang L. A Small Regulatory RNA Contributes to the Preferential Colonization of Escherichia coli O157:H7 in the Large Intestine in Response to a Low DNA Concentration. Front Microbiol 2017; 8:274. [PMID: 28289405 PMCID: PMC5326754 DOI: 10.3389/fmicb.2017.00274] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 02/09/2017] [Indexed: 11/13/2022] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) serotype O157:H7 (O157) is one of the most notorious human pathogens, causing severe disease in humans worldwide. O157 specifically colonizes the large intestine of mammals after passing through the small intestine, and this process is influenced by differential signals between the two regions. Small regulatory RNAs (sRNAs) are able to sense and respond to environmental changes and regulate diverse physiological processes in pathogenic bacteria. Although some sRNAs of O157 have been extensively investigated, whether these molecules can sense differences between the small and large intestine and influence the preferential colonization in the large intestine by O157 remains unknown. In this study, we identified a new sRNA, Esr055, in O157 which senses the low DNA concentration in the large intestine and contributes to the preferential colonization of the bacteria in this region. The number of O157 wild-type that adhered to the colon is 30.18 times higher than the number that adhered to the ileum of mice, while the number of the ΔEsr055 mutant that adhered to the colon decreased to 13.27 times higher than the number adhered to the ileum. Furthermore, we found that the expression of Esr055 is directly activated by the regulator, DeoR, and its expression is positively affected by DNA, which is significantly more abundant in the ileum than in the colon of mice. Additionally, combining the results of informatics predictions and transcriptomic analysis, we found that several virulence genes are up-regulated in the ΔEsr055 mutant and five candidate genes (z0568, z0974, z1356, z1926, and z5187) may be its direct targets.
Collapse
Affiliation(s)
- Runhua Han
- TEDA Institute of Biological Sciences and Biotechnology, Nankai UniversityTianjin, China; The Key Laboratory of Molecular Microbiology and Technology, Ministry of EducationTianjin, China
| | - Letian Xu
- TEDA Institute of Biological Sciences and Biotechnology, Nankai UniversityTianjin, China; The Key Laboratory of Molecular Microbiology and Technology, Ministry of EducationTianjin, China; Tianjin Key Laboratory of Microbial Functional GenomicsTianjin, China
| | - Ting Wang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai UniversityTianjin, China; The Key Laboratory of Molecular Microbiology and Technology, Ministry of EducationTianjin, China
| | - Bin Liu
- TEDA Institute of Biological Sciences and Biotechnology, Nankai UniversityTianjin, China; The Key Laboratory of Molecular Microbiology and Technology, Ministry of EducationTianjin, China; Tianjin Key Laboratory of Microbial Functional GenomicsTianjin, China
| | - Lei Wang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai UniversityTianjin, China; The Key Laboratory of Molecular Microbiology and Technology, Ministry of EducationTianjin, China; Tianjin Key Laboratory of Microbial Functional GenomicsTianjin, China; State Key Laboratory of Medicinal Chemical Biology, Nankai UniversityTianjin, China
| |
Collapse
|
8
|
Ting K, Aitken KJ, Penna F, Samiei AN, Sidler M, Jiang JX, Ibrahim F, Tolg C, Delgado-Olguin P, Rosenblum N, Bägli DJ. Uropathogenic E. coli (UPEC) Infection Induces Proliferation through Enhancer of Zeste Homologue 2 (EZH2). PLoS One 2016; 11:e0149118. [PMID: 26964089 PMCID: PMC4786126 DOI: 10.1371/journal.pone.0149118] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 01/27/2016] [Indexed: 01/13/2023] Open
Abstract
Host-pathogen interactions can induce epigenetic changes in the host directly, as well as indirectly through secreted factors. Previously, uropathogenic Escherichia coli (UPEC) was shown to increase DNA methyltransferase activity and expression, which was associated with methylation-dependent alterations in the urothelial expression of CDKN2A. Here, we showed that paracrine factors from infected cells alter expression of another epigenetic writer, EZH2, coordinate with proliferation. Urothelial cells were inoculated with UPEC, UPEC derivatives, or vehicle (mock infection) at low moi, washed, then maintained in media with Gentamycin. Urothelial conditioned media (CM) and extracellular vesicles (EV) were isolated after the inoculations and used to treat naïve urothelial cells. EZH2 increased with UPEC infection, inoculation-induced CM, and inoculation-induced EV vs. parallel stimulation derived from mock-inoculated urothelial cells. We found that infection also increased proliferation at one day post-infection, which was blocked by the EZH2 inhibitor UNC1999. Inhibition of demethylation at H3K27me3 had the opposite effect and augmented proliferation. CONCLUSION: Uropathogen-induced paracrine factors act epigenetically by altering expression of EZH2, which plays a key role in early host cell proliferative responses to infection.
Collapse
Affiliation(s)
- Kenneth Ting
- Faculty of Arts and Sciences, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Karen J. Aitken
- Developmental and Stem Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Urology Division, Department of Surgery, Hospital for Sick Children, Toronto, Ontario, Canada
- * E-mail:
| | - Frank Penna
- Urology Division, Department of Surgery, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Alaleh Najdi Samiei
- Developmental and Stem Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Martin Sidler
- Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Developmental and Stem Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Urology Division, Department of Surgery, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jia-Xin Jiang
- Developmental and Stem Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Fadi Ibrahim
- Developmental and Stem Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Cornelia Tolg
- Developmental and Stem Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Paul Delgado-Olguin
- Developmental and Stem Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Physiology and Experimental Medicine, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Norman Rosenblum
- Developmental and Stem Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Nephrology Division, Department of Surgery, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Darius J. Bägli
- Faculty of Arts and Sciences, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Developmental and Stem Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Urology Division, Department of Surgery, Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
9
|
Willy JA, Young SK, Stevens JL, Masuoka HC, Wek RC. CHOP links endoplasmic reticulum stress to NF-κB activation in the pathogenesis of nonalcoholic steatohepatitis. Mol Biol Cell 2015; 26:2190-204. [PMID: 25904325 PMCID: PMC4462938 DOI: 10.1091/mbc.e15-01-0036] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 04/16/2015] [Indexed: 01/10/2023] Open
Abstract
During metabolic stress, the UPR transcription factor CHOP activates NF-κB through a pathway involving IRAK2 expression, resulting in hepatocyte secretion of cytokines IL-8 and TNFα, which trigger inflammation and hepatocellular death. Free fatty acid induction of inflammation and cell death is an important feature of nonalcoholic steatohepatitis (NASH) and has been associated with disruption of the endoplasmic reticulum and activation of the unfolded protein response (UPR). After chronic UPR activation, the transcription factor CHOP (GADD153/DDIT3) triggers cell death; however, the mechanisms linking the UPR or CHOP to hepatoceullular injury and inflammation in the pathogenesis of NASH are not well understood. Using HepG2 and primary human hepatocytes, we found that CHOP induces cell death and inflammatory responses after saturated free fatty acid exposure by activating NF-κB through a pathway involving IRAK2 expression, resulting in secretion of cytokines IL-8 and TNFα directly from hepatocytes. TNFα facilitates hepatocyte death upon exposure to saturated free fatty acids, and secretion of both IL-8 and TNFα contribute to inflammation. Of interest, CHOP/NF-κB signaling is not conserved in primary rodent hepatocytes. Our studies suggest that CHOP plays a vital role in the pathophysiology of NASH by induction of secreted factors that trigger inflammation and hepatocellular death via a signaling pathway specific to human hepatocytes.
Collapse
Affiliation(s)
- Jeffrey A Willy
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Sara K Young
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - James L Stevens
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Howard C Masuoka
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Ronald C Wek
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
10
|
Yang B, Feng L, Wang F, Wang L. Enterohemorrhagic Escherichia coli senses low biotin status in the large intestine for colonization and infection. Nat Commun 2015; 6:6592. [PMID: 25791315 PMCID: PMC4382993 DOI: 10.1038/ncomms7592] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 02/10/2015] [Indexed: 12/17/2022] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) is an important foodborne pathogen that infects humans by colonizing the large intestine. Here we identify a virulence-regulating pathway in which the biotin protein ligase BirA signals to the global regulator Fur, which in turn activates LEE (locus of enterocyte effacement) genes to promote EHEC adherence in the low-biotin large intestine. LEE genes are repressed in the high-biotin small intestine, thus preventing adherence and ensuring selective colonization of the large intestine. The presence of this pathway in all nine EHEC serotypes tested indicates that it is an important evolutionary strategy for EHEC. The pathway is incomplete in closely related small-intestinal enteropathogenic E. coli due to the lack of the Fur response to BirA. Mice fed with a biotin-rich diet show significantly reduced EHEC adherence, indicating that biotin might be useful to prevent EHEC infection in humans. Enterohaemorrhagic Escherichia coli (EHEC) is an important foodborne pathogen that colonizes the large intestine. Here, the authors identify a signalling pathway that controls EHEC adherence to host cells in response to variations in biotin levels, ensuring selective colonization of the large intestine.
Collapse
Affiliation(s)
- Bin Yang
- 1] TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin 300457, P.R. China [2] Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin 300071, P.R. China
| | - Lu Feng
- 1] TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin 300457, P.R. China [2] Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin 300071, P.R. China [3] Tianjin Key Laboratory of Microbial Functional Genomics, Tianjin 300457, P.R. China [4] State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, P.R. China [5] SynBio Research Platform, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, P.R. China
| | - Fang Wang
- 1] TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin 300457, P.R. China [2] Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin 300071, P.R. China
| | - Lei Wang
- 1] TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin 300457, P.R. China [2] Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin 300071, P.R. China [3] Tianjin Key Laboratory of Microbial Functional Genomics, Tianjin 300457, P.R. China [4] State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, P.R. China
| |
Collapse
|
11
|
Pathogenesis of human enterovirulent bacteria: lessons from cultured, fully differentiated human colon cancer cell lines. Microbiol Mol Biol Rev 2014; 77:380-439. [PMID: 24006470 DOI: 10.1128/mmbr.00064-12] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hosts are protected from attack by potentially harmful enteric microorganisms, viruses, and parasites by the polarized fully differentiated epithelial cells that make up the epithelium, providing a physical and functional barrier. Enterovirulent bacteria interact with the epithelial polarized cells lining the intestinal barrier, and some invade the cells. A better understanding of the cross talk between enterovirulent bacteria and the polarized intestinal cells has resulted in the identification of essential enterovirulent bacterial structures and virulence gene products playing pivotal roles in pathogenesis. Cultured animal cell lines and cultured human nonintestinal, undifferentiated epithelial cells have been extensively used for understanding the mechanisms by which some human enterovirulent bacteria induce intestinal disorders. Human colon carcinoma cell lines which are able to express in culture the functional and structural characteristics of mature enterocytes and goblet cells have been established, mimicking structurally and functionally an intestinal epithelial barrier. Moreover, Caco-2-derived M-like cells have been established, mimicking the bacterial capture property of M cells of Peyer's patches. This review intends to analyze the cellular and molecular mechanisms of pathogenesis of human enterovirulent bacteria observed in infected cultured human colon carcinoma enterocyte-like HT-29 subpopulations, enterocyte-like Caco-2 and clone cells, the colonic T84 cell line, HT-29 mucus-secreting cell subpopulations, and Caco-2-derived M-like cells, including cell association, cell entry, intracellular lifestyle, structural lesions at the brush border, functional lesions in enterocytes and goblet cells, functional and structural lesions at the junctional domain, and host cellular defense responses.
Collapse
|
12
|
Protein kinase C mediates enterohemorrhagic Escherichia coli O157:H7-induced attaching and effacing lesions. Infect Immun 2014; 82:1648-56. [PMID: 24491575 DOI: 10.1128/iai.00534-13] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Enterohemorrhagic Escherichia coli serotype O157:H7 causes outbreaks of diarrhea, hemorrhagic colitis, and the hemolytic-uremic syndrome. E. coli O157:H7 intimately attaches to epithelial cells, effaces microvilli, and recruits F-actin into pedestals to form attaching and effacing lesions. Lipid rafts serve as signal transduction platforms that mediate microbe-host interactions. The aims of this study were to determine if protein kinase C (PKC) is recruited to lipid rafts in response to E. coli O157:H7 infection and what role it plays in attaching and effacing lesion formation. HEp-2 and intestine 407 tissue culture epithelial cells were challenged with E. coli O157:H7, and cell protein extracts were then separated by buoyant density ultracentrifugation to isolate lipid rafts. Immunoblotting for PKC was performed, and localization in lipid rafts was confirmed with an anti-caveolin-1 antibody. Isoform-specific PKC small interfering RNA (siRNA) was used to determine the role of PKC in E. coli O157:H7-induced attaching and effacing lesions. In contrast to uninfected cells, PKC was recruited to lipid rafts in response to E. coli O157:H7. Metabolically active bacteria and cells with intact lipid rafts were necessary for the recruitment of PKC. PKC recruitment was independent of the intimin gene, type III secretion system, and the production of Shiga toxins. Inhibition studies, using myristoylated PKCζ pseudosubstrate, revealed that atypical PKC isoforms were activated in response to the pathogen. Pretreating cells with isoform-specific PKC siRNA showed that PKCζ plays a role in E. coli O157:H7-induced attaching and effacing lesions. We concluded that lipid rafts mediate atypical PKC signal transduction responses to E. coli O157:H7. These findings contribute further to the understanding of the complex array of microbe-eukaryotic cell interactions that occur in response to infection.
Collapse
|
13
|
Enterohemorrhagic Escherichia coli O157:H7 Shiga toxins inhibit gamma interferon-mediated cellular activation. Infect Immun 2012; 80:2307-15. [PMID: 22526675 DOI: 10.1128/iai.00255-12] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) serotype O157:H7 is a food-borne pathogen that causes significant morbidity and mortality in developing and industrialized nations. EHEC infection of host epithelial cells is capable of inhibiting the gamma interferon (IFN-γ) proinflammatory pathway through the inhibition of Stat-1 phosphorylation, which is important for host defense against microbial pathogens. The aim of this study was to determine the bacterial factors involved in the inhibition of Stat-1 tyrosine phosphorylation. Human HEp-2 and Caco-2 epithelial cells were challenged directly with either EHEC or bacterial culture supernatants and stimulated with IFN-γ, and then the protein extracts were analyzed by immunoblotting. The data showed that IFN-γ-mediated Stat-1 tyrosine phosphorylation was inhibited by EHEC secreted proteins. Using two-dimensional difference gel electrophoresis, EHEC Shiga toxins were identified as candidate inhibitory factors. EHEC Shiga toxin mutants were then generated and complemented in trans, and mutant culture supernatant was supplemented with purified Stx to confirm their ability to subvert IFN-γ-mediated cell activation. We conclude that while other factors are likely involved in the suppression of IFN-γ-mediated Stat-1 tyrosine phosphorylation, E. coli-derived Shiga toxins represent a novel mechanism by which EHEC evades the host immune system.
Collapse
|
14
|
Li R, Hou J, Xu Q, Liu QJ, Shen YJ, Rodin G, Li M. High level interleukin-6 in the medium of human pancreatic cancer cell culture suppresses production of neurotransmitters by PC12 cell line. Metab Brain Dis 2012; 27:91-100. [PMID: 22109853 DOI: 10.1007/s11011-011-9270-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Accepted: 11/08/2011] [Indexed: 01/17/2023]
Abstract
It has been suggested that pancreatic cancer is associated with a greater prevalence of depression than many other cancers, but the mechanism accounting for this potential association has not yet been illustrated. In the present study, conditioned media (CM) from three pancreatic cancer cell lines and primary pancreatic cancer cells from two patients were added to culture system of differentiated pheochromocytoma cell line PC12. The release of dopamine (DA) and norepinephrine (NE) by PC12 was significantly inhibited after CM treatment (P < 0.05), similar to what happened after recombinant interleukin 6(IL-6) treatment. Furthermore, pretreatment with anti-IL-6 antibody significantly blocked the inhibitory effects of pancreatic cancer CM on DA and NE production (P < 0.05). We also demonstrated that tyrosine hydroxylase (TH), the rate-limiting enzyme for synthesis of catecholamine, was reduced after exposure to IL-6, which was accompanied by JAK-STAT3 pathway activation. Our results demonstrated that IL-6 in CM from pancreatic cancer down-regulated the production of DA and NE by PC12 cell. The possible underlying mechanisms might be decreasing TH production via activation of JAK-STAT3 signal transduction pathway. The present study might help to better understand the close relationship between pancreatic cancer and depression.
Collapse
Affiliation(s)
- Rong Li
- Department of Hematology, Chang Zheng Hospital, Second Military Medical University, Shanghai 200003, China
| | | | | | | | | | | | | |
Collapse
|
15
|
Gareau MG, Ho NK, Brenner D, Sousa AJ, LeBourhis L, Mak TW, Girardin SE, Philpott DJ, Sherman PM. Enterohaemorrhagic, but not enteropathogenic, Escherichia coli infection of epithelial cells disrupts signalling responses to tumour necrosis factor-alpha. Microbiology (Reading) 2011; 157:2963-2973. [DOI: 10.1099/mic.0.051094-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Enterohaemorrhagic Escherichia coli (EHEC), serotype O157 : H7 is a non-invasive, pathogenic bacterium that employs a type III secretion system (T3SS) to inject effector proteins into infected cells. In this study, we demonstrate that EHEC blocks tumour necrosis factor-alpha (TNFα)-induced NF-κB signalling in infected epithelial cells. HEK293T and INT407 epithelial cells were challenged with EHEC prior to stimulation with TNFα. Using complementary techniques, stimulation with TNFα caused activation of NF-κB, as determined by luciferase reporter assay (increase in gene expression), Western blotting (phosphorylation of IκBα), immunofluorescence (p65 nuclear translocation) and immunoassay (CXCL-8 secretion), and each was blocked by EHEC O157 : H7 infection. In contrast, subversion of host cell signalling was not observed following exposure to either enteropathogenic E. coli, strain E2348/69 (O127 : H6) or the laboratory E. coli strain HB101. Heat-killed EHEC had no effect on NF-κB activation by TNFα. Inhibition was mediated, at least in part, by Shiga toxins and by the O157 plasmid, but not by the T3SS or flagellin, as demonstrated by using isogenic mutant strains. These findings indicate the potential for developing novel therapeutic targets to interrupt the infectious process.
Collapse
Affiliation(s)
- Mélanie G. Gareau
- Research Institute, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - Nathan K. Ho
- Department of Laboratory Medicine and Pathobiology University of Toronto, Medical Sciences Building, 1 King’s College Circle, University of Toronto, Toronto, ON M5S 1A8, Canada
- Research Institute, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - Dirk Brenner
- Campbell Family Cancer Research Institute, Princess Margaret Hospital, 620 University Avenue, Toronto, ON M5G 2C1, Canada
| | - Andrew J. Sousa
- Research Institute, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - Lionel LeBourhis
- Department of Immunology, Medical Sciences Building, 1 King’s College Circle, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Tak W. Mak
- Campbell Family Cancer Research Institute, Princess Margaret Hospital, 620 University Avenue, Toronto, ON M5G 2C1, Canada
| | - Stephen E. Girardin
- Department of Laboratory Medicine and Pathobiology University of Toronto, Medical Sciences Building, 1 King’s College Circle, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Dana J. Philpott
- Department of Immunology, Medical Sciences Building, 1 King’s College Circle, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Philip M. Sherman
- Department of Laboratory Medicine and Pathobiology University of Toronto, Medical Sciences Building, 1 King’s College Circle, University of Toronto, Toronto, ON M5S 1A8, Canada
- Research Institute, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
16
|
MEDINA MARJORIEB, UKNALIS JOSEPH, TU SHUI. EFFECTS OF SUGAR ADDITION IN LURIA BERTANI (LB) MEDIA ON ESCHERICHIA COLI O157:H7. J Food Saf 2011. [DOI: 10.1111/j.1745-4565.2011.00311.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
17
|
Tahamtan Y, Kargar M, Namdar N, Rahimian A, Hayati M, Namavari M. Probiotic inhibits the cytopathic effect induced by Escherichia coli O157:H7 in Vero cell line model. Lett Appl Microbiol 2011; 52:527-31. [DOI: 10.1111/j.1472-765x.2011.03037.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
18
|
Shen-Tu G, Schauer DB, Jones NL, Sherman PM. Detergent-resistant microdomains mediate activation of host cell signaling in response to attaching-effacing bacteria. J Transl Med 2010; 90:266-81. [PMID: 19997063 DOI: 10.1038/labinvest.2009.131] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) O157:H7 causes outbreaks of bloody diarrhea and the hemolytic-uremic syndrome. EHEC intimately adheres to epithelial cells, effaces microvilli and induces attaching-effacing (AE) lesions. Detergent-resistant microdomains (lipid rafts) serve as membrane platforms for the recruitment of signaling complexes to mediate host responses to infection. The aim of this study was to define the role of lipid rafts in activating signal transduction pathways in response to AE bacterial pathogens. Epithelial cell monolayers were infected with EHEC (MOI 100:1, 3 h, 37 degrees C) and lipid rafts isolated by buoyant density ultracentrifugation. Phosphoinositide 3-kinase (PI3K) localization to lipid rafts was confirmed using PI3K and anti-caveolin-1 antibodies. Mice with cholesterol storage disease Niemann-Pick, type C were used as in vivo models to confirm the role of lipid rafts in mediating signaling response to AE organisms. In contrast to uninfected cells, PI3K was recruited to lipid rafts in response to EHEC infection. Metabolically active bacteria and cells with intact cholesterol-rich microdomains were necessary for the recruitment of second messengers to lipid rafts. Recruitment of PI3K to lipid rafts was independent of the intimin (eaeA) gene, type III secretion system, and production of Shiga-like toxins. Colonization of NPC(-/-) colonic mucosa by Citrobacter rodentium and AE lesion formation were both delayed, compared with wild-type mice infected with the murine-specific AE bacterial pathogen. C. rodentium-infected NPC(-/-) mice had reduced colonic epithelial hyperplasia (64+/-8.251 vs 112+/-2.958 microm; P<0.05) and decreased secretion of IFN-gamma (17.6+/-17.6 vs 71+/-26.3 pg/ml, P<0.001). Lipid rafts mediate host cell signal transduction responses to AE bacterial infections both in vitro and in vivo. These findings advance the current understanding of microbial-eukaryotic cell interactions in response to enteric pathogens that hijack signaling responses mediated through lipid rafts.
Collapse
Affiliation(s)
- Grace Shen-Tu
- Research Institute, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | | | | | | |
Collapse
|
19
|
Lapointe TK, O'Connor PM, Jones NL, Menard D, Buret AG. Interleukin-1 receptor phosphorylation activates Rho kinase to disrupt human gastric tight junctional claudin-4 during Helicobacter pylori infection. Cell Microbiol 2010; 12:692-703. [PMID: 20070312 DOI: 10.1111/j.1462-5822.2010.01429.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Helicobacter pylori infects more than half of the human population worldwide. In the absence of treatment, this persistent infection leads to asymptomatic gastritis, which in some cases can progress into gastric ulcers and adenocarcinomas. The host-microbial interactions that govern the clinical outcome of infection remain incompletely understood. H. pylori is known to disrupt gastric epithelial tight junctions, which may represent a significant component of disease pathogenesis. The present study demonstrates that H. pylori disrupt epithelial tight junctional claudin-4 in a Rho kinase (ROCK)-dependent manner in human gastric epithelial (HGE-20) cell monolayers, independently of the virulence factors CagA and VacA, and without altering claudin-4 transcription. In the same epithelial cell model, interleukin (IL)-1beta, mediated a similar ROCK-dependent pattern of tight junction disruption. Further experiments revealed that H. pylori infection induced IL-1 receptor type I (IL-1RI) phosphorylation, independently of epithelial secretion of its endogenous ligands IL-1alpha, IL-1beta or IL-18. Finally, inhibition of IL-1RI activation prevented H. pylori-induced ROCK activation and claudin-4 disruption. Taken together, these findings identify a novel pathophysiological mechanism by which H. pylori disrupts gastric epithelial barrier structure via IL-1RI-dependent activation of ROCK, which in turn mediates tight junctional claudin-4 disruption.
Collapse
Affiliation(s)
- Tamia K Lapointe
- Department of Biological Sciences and Inflammation Research Network, University of Calgary, Alberta, Canada
| | | | | | | | | |
Collapse
|
20
|
Pseudomonas aeruginosa Inhibition of Flagellin-activated NF-kappaB and interleukin-8 by human airway epithelial cells. Infect Immun 2009; 77:2857-65. [PMID: 19451246 DOI: 10.1128/iai.01355-08] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pseudomonas aeruginosa-induced activation of NF-kappaB and secretion of proinflammatory cytokines by airway epithelial cells require that the bacteria express flagellin. We tested whether P. aeruginosa and human airway epithelial cells secrete factors that modulated this response. Experiments were performed with both the Calu-3 cell line and primary cultures of tracheal epithelial cells. P. aeruginosa strain PAK DeltafliC (flagellin knockout) did not activate NF-kappaB or interleukin-8 (IL-8) but inhibited flagellin-activated NF-kappaB by 40 to 50% and IL-8 secretion by 20 to 25%. PAK DeltafliC also inhibited NF-kappaB induced by IL-1beta and Toll-like receptor 2 agonist Pam3CSK4. Similar inhibitions were observed with strains PAK, PAO1, and PA14. The inhibitory factor was present in conditioned medium isolated from PAK DeltafliC or Calu-3 plus PAK DeltafliC, but it was not present in conditioned medium isolated from Calu-3 cells alone or from PAK DeltafliC that had been heat treated. Inhibition by PAK DeltafliC-conditioned medium was exerted from either the apical or the basolateral side of the epithelium, was enhanced in simple Ringer's solution over that in tissue culture medium, and did not result from altered pH or depletion of glucose. The inhibitory effect of conditioned medium was abolished by boiling and appeared from filtration studies to result from effects of a factor with a molecular mass of <3 kDa. These and further studies with isogenic mutants led to the conclusion that the NF-kappaB and IL-8 response of airway epithelial cells to P. aeruginosa results from a balance of proinflammatory effects of flagellin and antiinflammatory effects of a small (<3-kDa), heat-sensitive factor(s) that is not lipopolysaccharide, C12 homoserine lactone, alginate, CIF, or exotoxin A, S, T, U, or Y.
Collapse
|
21
|
Jandu N, Ho NKL, Donato KA, Karmali MA, Mascarenhas M, Duffy SP, Tailor C, Sherman PM. Enterohemorrhagic Escherichia coli O157:H7 gene expression profiling in response to growth in the presence of host epithelia. PLoS One 2009; 4:e4889. [PMID: 19293938 PMCID: PMC2654852 DOI: 10.1371/journal.pone.0004889] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Accepted: 02/04/2009] [Indexed: 12/30/2022] Open
Abstract
Background The pathogenesis of enterohemorrhagic Escherichia coli (EHEC) O157∶H7 infection is attributed to virulence factors encoded on multiple pathogenicity islands. Previous studies have shown that EHEC O157∶H7 modulates host cell signal transduction cascades, independent of toxins and rearrangement of the cytoskeleton. However, the virulence factors and mechanisms responsible for EHEC-mediated subversion of signal transduction remain to be determined. Therefore, the purpose of this study was to first identify differentially regulated genes in response to EHEC O157∶H7 grown in the presence of epithelial cells, compared to growth in the absence of epithelial cells (that is, growth in minimal essential tissue culture medium alone, minimal essential tissue culture medium in the presence of 5% CO2, and Penassay broth alone) and, second, to identify EHEC virulence factors responsible for pathogen modulation of host cell signal transduction. Methodology/Principal Findings Overnight cultures of EHEC O157∶H7 were incubated for 6 hr at 37°C in the presence or absence of confluent epithelial (HEp-2) cells. Total RNA was then extracted and used for microarray analyses (Affymetrix E. coli Genome 2.0 gene chips). Relative to bacteria grown in each of the other conditions, EHEC O157∶H7 cultured in the presence of cultured epithelial cells displayed a distinct gene-expression profile. A 2.0-fold increase in the expression of 71 genes and a 2.0-fold decrease in expression of 60 other genes were identified in EHEC O157∶H7 grown in the presence of epithelial cells, compared to bacteria grown in media alone. Conclusion/Significance Microarray analyses and gene deletion identified a protease on O-island 50, gene Z1787, as a potential virulence factor responsible for mediating EHEC inhibition of the interferon (IFN)-γ-Jak1,2-STAT-1 signal transduction cascade. Up-regulated genes provide novel targets for use in developing strategies to interrupt the infectious process.
Collapse
Affiliation(s)
- Narveen Jandu
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
- Research Institute, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Nathan K. L. Ho
- Research Institute, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Kevin A. Donato
- Research Institute, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Mohamed A. Karmali
- Laboratory of Foodborne Zoonosis, Public Health Agency of Canada, Guelph, Ontario, Canada
| | - Mariola Mascarenhas
- Laboratory of Foodborne Zoonosis, Public Health Agency of Canada, Guelph, Ontario, Canada
| | - Simon P. Duffy
- Research Institute, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Chetankumar Tailor
- Research Institute, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Philip M. Sherman
- Research Institute, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
22
|
Jandu N, Zeng ZJ, Johnson-Henry KC, Sherman PM. Probiotics prevent enterohaemorrhagic Escherichia coli O157:H7-mediated inhibition of interferon-gamma-induced tyrosine phosphorylation of STAT-1. MICROBIOLOGY-SGM 2009; 155:531-540. [PMID: 19202101 DOI: 10.1099/mic.0.021931-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Enterohaemorrhagic Escherichia coli (EHEC) O157:H7 inhibits interferon (IFN)-gamma-stimulated tyrosine phosphorylation of signal transducer and activator of transcription (STAT)-1 in epithelial cells. We determined the effects of probiotics on EHEC-mediated disruption of IFN-gamma-stimulated STAT-1 activation in epithelial cell lines. Confluent Intestine 407, HEp-2 and Caco-2 epithelial cells were pre-treated (3 h) with either probiotics or surface-layer proteins derived from Lactobacillus helveticus R0052 prior to infection with EHEC O157:H7 strain CL56 (m.o.i. 100:1, 6 h, 37 degrees C in 5% CO2). Subsequently, cells were washed and stimulated with human recombinant IFN-gamma (50 ng ml(-1), 0.5 h, 37 degrees C) followed by whole-cell protein extraction and immunoblotting for tyrosine-phosphorylated STAT-1. Relative to uninfected cells, STAT-1-activation was reduced after EHEC O157:H7 infection. Pre-incubation with the probiotic L. helveticus R0052 followed by EHEC infection abrogated pathogen-mediated disruption of IFN-gamma-STAT-1 signalling. As determined using Transwell inserts, probiotic-mediated protection was independent of epithelial cell contact. In contrast, pre-incubation with boiled L. helveticus R0052, an equal concentration of viable Lactobacillus rhamnosus R0011, or surface-layer proteins (0.14 mg ml(-1)) did not restore STAT-1 signalling in EHEC-infected cells. The viable probiotic agent L. helveticus R0052 prevented EHEC O157:H7-mediated subversion of epithelial cell signal transduction responses.
Collapse
Affiliation(s)
- Narveen Jandu
- Research Institute, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Zoë Jingjing Zeng
- Research Institute, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | | | - Philip M Sherman
- Research Institute, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
23
|
Campylobacter jejuni cocultured with epithelial cells reduces surface capsular polysaccharide expression. Infect Immun 2009; 77:1959-67. [PMID: 19273563 DOI: 10.1128/iai.01239-08] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The host cell environment can alter bacterial pathogenicity. We employed a combination of cellular and molecular techniques to study the expression of Campylobacter jejuni polysaccharides cocultured with HCT-8 epithelial cells. After two passages, the amount of membrane-bound high-molecular-weight polysaccharide was considerably reduced. Microarray profiling confirmed significant downregulation of capsular polysaccharide (CPS) locus genes. Experiments using conditioned media showed that sugar depletion occurred only when the bacterial and epithelial cells were cocultured. CPS depletion occurred when C. jejuni organisms were exposed to conditioned media from a different C. jejuni strain but not when exposed to conditioned media from other bacterial species. Proteinase K or heat treatment of conditioned media under coculture conditions abrogated the effect on the sugars, as did formaldehyde fixation and cycloheximide treatment of host cells or chloramphenicol treatment of the bacteria. However, sugar depletion was not affected in flagellar export (fliQ) and quorum-sensing (luxS) gene mutants. Passaged C. jejuni showed reduced invasiveness and increased serum sensitivity in vitro. C. jejuni alters its surface polysaccharides when cocultured with epithelial cells, suggesting the existence of a cross talk mechanism that modulates CPS expression during infection.
Collapse
|
24
|
Lebeis SL, Sherman MA, Kalman D. Protective and destructive innate immune responses to enteropathogenic Escherichia coli and related A/E pathogens. Future Microbiol 2008; 3:315-28. [DOI: 10.2217/17460913.3.3.315] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Enteropathogenic Escherichia coli, enterohemorrhagic E. coli (O157:H7) and Citrobacter rodentium are classified as attaching and effacing (A/E) pathogens based on their ability to adhere to intestinal epithelium, destroy microvilli and induce pedestal formation at the site of infection. A/E bacterial infections also cause acute diarrheal episodes and intestinal inflammation. The use of model systems has led to an understanding of the innate immune response to A/E pathogens. The innate immune system plays a protective role, initiating a productive antibody response, directly killing bacteria and inducing repair mechanisms following tissue damage caused by infection. However, hyperactivation of the innate immune system can have negative consequences, including exacerbated tissue destruction following neutrophil infiltration. Here we review how innate immune cell types, including neutrophils, macrophages and dendritic cells, orchestrate both protective and destructive responses. Such information is crucial for the development of therapeutics that can mitigate destructive inflammatory responses while accentuating those that are protective.
Collapse
Affiliation(s)
- Sarah L Lebeis
- Microbiology & Molecular Genetics Graduate Program, Emory University School of Medicine, 615 Michael Street, Whitehead Research Building #155, Atlanta, GA 30322, USA and, Department of Pathology & Laboratory Medicine, Emory University School of Medicine, 615 Michael Street, Whitehead Research Building #144, Atlanta, GA 30322, USA
| | - Melanie A Sherman
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, 615 Michael Street, Whitehead Research Building #144, Atlanta, GA 30322, USA
| | - Daniel Kalman
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, 615 Michael Street, Whitehead Research Building #144, Atlanta, GA 30322, USA
| |
Collapse
|
25
|
Vareille M, Rannou F, Thélier N, Glasser AL, de Sablet T, Martin C, Gobert AP. Heme Oxygenase-1 Is a Critical Regulator of Nitric Oxide Production in EnterohemorrhagicEscherichia coli-Infected Human Enterocytes. THE JOURNAL OF IMMUNOLOGY 2008; 180:5720-6. [DOI: 10.4049/jimmunol.180.8.5720] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
26
|
Li R, Wang JJ, Wu X, Guo J, Wang MM, Wang X, Xie J, He XJ. Effect of BxPC-3-conditioned medium on the metabolism of DA and NE in PC12 cell line and its mechanism of action. Shijie Huaren Xiaohua Zazhi 2008; 16:484-487. [DOI: 10.11569/wcjd.v16.i5.484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the effect of BxPC-3-conditioned medium on the metabolism of DA and NE in PC12 cells and its mechanism of action.
METHODS: PC12 cells were treated with BxPC-3-conditioned medium without IL-6. The rate of apoptosis of PC12 cells was detected by flow cytometry. DA, 5-HT and NE in the medium were measured by HPLC. IL-6 (0.01 mg/L, 0.1 mg/L, 0.25 mg/L, 0.5 mg/L, 1 mg/L, 1.5 mg/L, 2 mg/L) was added into the medium deprived of serum, then the DA, 5-HT and NE in the medium were measured.
RESULTS: There was no significant difference in the rate of apoptosis of PC12 cells in each group. After anti-IL-6 was added into the conditioned medium, the quantity of DA and NE in the medium was increased. The concentration of IL-6 was increased. DA and NE were metabolized in a dose-dependent manner. Addition of 1mg/L IL-6 significantly decreased DA and NE.
CONCLUSION: BxPC-3-conditioned medium has certain effects on the metabolism of DA and NE of PC12 cells by adding IL-6.
Collapse
|
27
|
Gobert AP, Vareille M, Glasser AL, Hindré T, de Sablet T, Martin C. Shiga toxin produced by enterohemorrhagic Escherichia coli inhibits PI3K/NF-kappaB signaling pathway in globotriaosylceramide-3-negative human intestinal epithelial cells. THE JOURNAL OF IMMUNOLOGY 2007; 178:8168-74. [PMID: 17548655 DOI: 10.4049/jimmunol.178.12.8168] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Shiga toxin (Stx) produced by enterohemorrhagic Escherichia coli (EHEC) binds to endothelial cells expressing globotriaosylceramide-3 (Gb-3) and induces cell death by inhibiting translation. Nonetheless, the effects of Stx on human enterocytes, which lacks receptor Gb-3, remain less known. In this study, we questioned whether EHEC-derived Stx may modulate cellular signalization in the Gb-3-negative human epithelial cell line T84. Stx produced by EHEC was fixed and internalized by the cells. A weak activation of NF-kappaB was observed in T84 cells after EHEC infection. Cells infected with an isogenic mutant lacking stx1 and stx2, the genes encoding Stx, displayed an increased NF-kappaB DNA-binding activity. Consequently, the NF-kappaB-dependent CCL20 and IL-8 gene transcription and chemokine production were enhanced in T84 cells infected with the Stx mutant in comparison to the wild-type strain. Investigating the mechanism by which Stx modulates NF-kappaB activation, we showed that the PI3K/Akt signaling pathway was not induced by EHEC but was enhanced by the strain lacking Stx. Pharmacological inhibition of the PI3K/Akt signalization in EHEC DeltaStx-infected T84 cells yielded to a complete decrease of NF-kappaB activation and CCL20 and IL-8 mRNA expression. This demonstrates that the induction of the PI3K/Akt/NF-kappaB pathway is potentially induced by EHEC, but is inhibited by Stx in Gb-3-negative epithelial cells. Thus, Stx is an unrecognized modulator of the innate immune response of human enterocytes.
Collapse
Affiliation(s)
- Alain P Gobert
- Institut National de la Recherche Agronomique, UR454 Unité de Microbiologie, Centre de Theix, 63122 Saint-Genès-Champanelle, France.
| | | | | | | | | | | |
Collapse
|
28
|
Jandu N, Shen S, Wickham ME, Prajapati R, Finlay BB, Karmali MA, Sherman PM. Multiple seropathotypes of verotoxin-producing Escherichia coli (VTEC) disrupt interferon-γ-induced tyrosine phosphorylation of signal transducer and activator of transcription (Stat)-1. Microb Pathog 2007; 42:62-71. [PMID: 17174521 DOI: 10.1016/j.micpath.2006.10.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2006] [Revised: 10/30/2006] [Accepted: 10/30/2006] [Indexed: 12/22/2022]
Abstract
Verotoxin-producing Escherichia coli (VTEC) O157:H7 inhibits interferon-gamma-stimulated tyrosine phosphorylation of signal transducer and activator of transcription (Stat)-1 in epithelial cells, independent of Verotoxins and the locus of enterocyte effacement pathogenicity island. Although E. coli O157:H7 is the major cause of disease in humans, non-O157:H7 VTEC also cause human disease. However, the virulence properties of non-O157:H7 VTEC are less well characterized. The aims of this study were to define the ability of VTEC strains of differing seropathotypes (classified as A-E) to inhibit interferon-gamma stimulated Stat1-phosphorylation and to further characterize the bacterial-derived inhibitory factor. Confluent T84 and HEp-2 cells were infected with VTEC strains (MOI 100:1, 6h, 37 degrees C), and then stimulated with interferon-gamma (50 ng/mL) for 0.5h at 37 degrees C. Whole-cell protein extracts of infected cells were collected and prepared for immunoblotting to detect tyrosine phosphorylation of Stat1. The effects of E. coli O55 strains, the evolutionary precursors of VTEC, on Stat1-tyrosine phosphorylation were also determined. The effects of isogenic mutants of O-islands 47 and 122 were tested to determine the role of genes encoded on these putative pathogenicity islands in mediating VTEC inhibition of the interferon-gamma-Stat1 signaling cascade. To evaluate potential mechanism(s) of inhibition, VTEC O157:H7-infected cells were treated with pharmacological inhibitors, including, wortmannin and LY294002. Relative to uninfected cells, Stat1-tyrosine phosphorylation was significantly reduced after 6h infection of both T84 and HEp-2 cells by VTEC strains of all five seropathotypes. E. coli O55 strains, but not enteropathogenic E. coli (EPEC), also caused inhibition of Stat1-tyrosine phosphorylation, suggesting that this effect was acquired early in the evolution of VTEC. Stat1-activation did not recover in epithelial cells infected with isogenic mutants of O-islands 47 and 122, indicating that the inhibitory factor was not contained in these genomic regions. Stat1-phosphorylation remained intact when VTEC-infected cells were treated with wortmannin (0-100 nM), but not by treatment with the more specific PI3-kinase inhibitor, LY294002. Inhibition of interferon-gamma stimulated Stat1-tyrosine phosphorylation by VTEC of multiple seropathotypes indicates the presence of a common inhibitory factor that is independent of bacterial virulence in humans. The results of treatment with wortmannin suggest that the bacterial-derived inhibitory factor employs host cell signal transduction to mediate inhibition of Stat1-activation.
Collapse
Affiliation(s)
- Narveen Jandu
- Research Institute, Hospital for Sick Children, Ont., Canada
| | | | | | | | | | | | | |
Collapse
|